WO2015048653A1 - Methods of identifying, assessing, preventing, and treating cardiac disorders using gsk-3b and phosphorylation targets thereof - Google Patents

Methods of identifying, assessing, preventing, and treating cardiac disorders using gsk-3b and phosphorylation targets thereof Download PDF

Info

Publication number
WO2015048653A1
WO2015048653A1 PCT/US2014/058062 US2014058062W WO2015048653A1 WO 2015048653 A1 WO2015048653 A1 WO 2015048653A1 US 2014058062 W US2014058062 W US 2014058062W WO 2015048653 A1 WO2015048653 A1 WO 2015048653A1
Authority
WO
WIPO (PCT)
Prior art keywords
level
gsk
expression
activity
sample
Prior art date
Application number
PCT/US2014/058062
Other languages
French (fr)
Inventor
Jonathan Kirk
David Kass
Jennifer Van Eyk
Original Assignee
The Johns Hopkins University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Johns Hopkins University filed Critical The Johns Hopkins University
Publication of WO2015048653A1 publication Critical patent/WO2015048653A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/32Cardiovascular disorders
    • G01N2800/325Heart failure or cardiac arrest, e.g. cardiomyopathy, congestive heart failure

Definitions

  • CRT cardiac resynchronization therapy
  • the disclosure provides a method of determining whether a subject is afflicted with a cardiac disorder, the method comprising: a) determining the level of expression or level of activity of one or more biomarkers selected from glycogen synthase kinase 3 ⁇ (GSK-3P) and the phosphorylation targets of GSK-3P in a biological sample obtained from the subject; b) determining the level of expression or level of activity of the one or more biomarkers in a control sample; and c) comparing the level of expression or level of activity of said one or more biomarkers detected in steps a) and b).
  • a significant decrease in the level of expression or level of activity of the one or more biomarkers in the subject sample relative to the control level of expression or level of activity of the one or more biomarkers indicates that the subject is afflicted with the cardiac disorder.
  • the disclosure features a method of determining whether a subject afflicted with a cardiac disorder or at risk for developing a cardiac disorder would benefit from cardiac resynchronization therapy (CRT) or enhanced activity of GSK-3P, which method comprises: a) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P in a biological sample obtained from the subject; b) determining the level of expression or level of activity of the one or more biomarkers in a control sample; and c) comparing the level of expression or level of activity of said one or more biomarkers detected in steps a) and b), optionally wherein between the first point in time and the subsequent point in time, the subject has undergone treatment for the cardiac disorder; wherein a significant decrease in the level of expression or level of activity of the one or more biomarkers in the subject sample relative to the control level of expression or level of activity of the one or more biomarkers indicates that the subject afflicted with the cardiac disorder or
  • the disclosure features a method for monitoring the progression of a cardiac disorder in a subject.
  • the method comprises: a) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P in a biological sample obtained from the subject at a first point; b) repeating step a) at a subsequent point in time; and c) comparing the level of expression or level of activity of said one or more biomarkers detected in steps a) and b) to monitor the progression of the cardiac disorder.
  • the disclosure features a method for stratifying subjects afflicted with a cardiac disorder according to predicted clinical outcome of treatment with CRT or one or more GSK-3P modulators, optionally wherein the predicted clinical outcome is (a) restored mechanical cardiac synchrony, (b) decreased morbidity, or (c) increased survival time resulting from treatment with one or more modulators of GSK-3p.
  • the method comprises: a) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P, in a biological sample obtained from the subject; b) determining the level of expression or level of activity of the one or more biomarkers in a control sample; and c) comparing the level of expression or level of activity of said one or more biomarkers detected in steps a) and b); wherein a significant modulation, optionally a decrease, in the level of expression or level of activity of the one or more biomarkers in the subject sample relative to the control level of expression or level of activity of the one or more biomarkers predicts the clinical outcome of the patient to treatment with CRT or one or more GSK-3P modulators.
  • any of the methods described herein can comprise determining the level of expression of the one or more biomarkers in each sample.
  • the level of expression may be determined by contacting each sample, or one or more nucleic acids derived from each sample, with one or more nucleic acid arrays or one or more nucleic acid probes.
  • the level of expression may be determined by performing qPCR on each sample or by performing qPCR on one or more nucleic acids derived from each sample.
  • any of the methods described herein can comprise determining the level of activity of the one or more biomarkers in each sample.
  • the level of activity may be determined by analyzing one or more biomarkers in each sample by mass spectroscopy.
  • the level of activity may be determined by contacting each sample with an antibody that specifically binds to the one or more biomarkers.
  • any of the methods described herein can comprise treating the subject with a therapeutic agent that specifically modulates the level of expression or level of activity of the one or more biomarkers.
  • the method may comprise treating the subject with one or more modulators of GSK-3p.
  • the modulator may increase the expression level of GSK-3P or increases the phosphorylation state of GSK-3p.
  • the methods described herein may comprise treating the subject in need thereof with a therapeutically effective amount of a therapeutic agent that specifically modulates the level of expression or level of activity of the one or more biomarkers.
  • the method may comprise treating the subject with one or more modulators of GSK- 3 ⁇ .
  • the modulator may increase the expression level of GSK-3P or increase the
  • the disclosure features a method of determining the efficacy of a test compound for treating a cardiac disorder in a subject.
  • the method comprises: a) exposing a first sample obtained from a subject to a test compound b) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P in the first sample , optionally wherein the sample is analyzed in vivo, ex vivo, or in vitro; c) determining the level of expression or level of activity of the one or more biomarkers in a second sample obtained from the subject, wherein the second sample is not exposed to the test compound; and d) comparing the level of expression or level of activity of the one or more biomarkers in the first and second samples, wherein the test compound has efficacy for treating a cardiac disorder if the level of expression or level of activity is greater in the first sample than in the second sample.
  • the disclosure features a method of determining the efficacy of a therapy for treating a cardiac disorder in a subject.
  • the method comprises: a) obtaining a first sample from a subject prior to providing at least a portion of the therapy to the subject; b) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P in the first sample, optionally wherein the sample or therapy is analyzed in vivo, ex vivo, or in vitro; c) obtaining a second sample from the subject following provision of the portion of the therapy; d) determining the level of expression or level of activity of the one or more biomarkers in the second sample; and e) comparing the level of expression or level of activity of the one or more biomarkers in the first and second samples, wherein the therapy has efficacy for treating a cardiac disorder if the level of expression or level of activity is greater in the second sample than in the first sample.
  • the disclosure features a method for identifying a compound which treats a cardiac disorder, the method comprising: a) contacting one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P with a test compound, optionally wherein the contacting occurs in vivo, ex vivo, or in vitro; and b) determining the effect of the test compound on the level of expression or level of activity of the one or more biomarkers to thereby identify a compound which treats the cardiac disorder.
  • the one or more biomarkers may be expressed on or in a cell.
  • the cells may be isolated from an animal model of the cardiac disorder.
  • the cells are from a subject afflicted with the cardiac disorder.
  • the disclosure features a method for treating a cardiac disorder, the method comprising contacting a cell with an agent that modulates, optionally increases, the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P to thereby treat the cardiac disorder.
  • the step of contacting may occur in vivo, ex vivo, or in vitro.
  • the method may further comprise contacting the cell with an additional agent that treats the cardiac disorder.
  • control may be determined from a sample from a subject not afflicted with the cardiac disorder.
  • the sample may consist of or comprises body fluid, cells, cell lines, histological slides, paraffin embedded tissue, fresh frozen tissue, fresh tissue, biopsies, blood, plasma, serum, buccal scrape, saliva, cerebrospinal fluid, urine, stool, mucus, or bone marrow, obtained from the subject.
  • the sample may comprise body fluid, and the body fluid may be selected from amniotic fluid, aqueous humor, bile, blood and blood plasma, cerebrospinal fluid, cerumen and earwax, cowper's fluid or pre-ejaculatory fluid, chyle, chyme, stool, female ejaculate, interstitial fluid, intracellular fluid, lymph, menses, breast milk, mucus, pleural fluid, peritoneal fluid, pus, saliva, sebum, semen, serum, sweat, synovial fluid, tears, urine, vaginal lubrication, vitreous humor, and vomit.
  • the sample is blood.
  • the expression level of the one or more biomarkers may be assessed by detecting the presence in the samples of a polynucleotide molecule encoding the biomarker or a portion of said polynucleotide molecule.
  • the polynucleotide molecule may be a mR A, cDNA, or functional variants or fragments thereof and, optionally, wherein the step of detecting further comprises amplifying the polynucleotide molecule.
  • the expression level of the one or more biomarkers may be assessed by annealing a nucleic acid probe with the sample of the polynucleotide encoding the one or more biomarkers or a portion of said polynucleotide molecule under stringent hybridization conditions.
  • the expression level of the biomarker is assessed by detecting the presence in the samples, whether phosphorylated, unphosphorylated, or both, of a protein of the biomarker, a polypeptide, or protein fragment thereof comprising said protein.
  • the presence of the protein, polypeptide or protein fragment thereof may be detected using a reagent which specifically binds with said protein, polypeptide or protein fragment thereof, optionally wherein the reagent is selected from an antibody, an antibody derivative, and an antibody fragment.
  • the activity level of the biomarker may be assessed by determining the magnitude of modulation of the activity or expression level of downstream targets of the one or more biomarkers.
  • determining the level of expression or level of activity of the one or more biomarkers comprises determining the phosphorylation state of GSK-3P or one or more phosphorylation targets of GSK-3p.
  • the phosphorylation state may be determined, for example, either by mass spectroscopy or by using an antibody that specifically binds to the phosphorylated form of the protein.
  • the phosphorylation targets of GSK-3P may be myofilament phosphorylation targets.
  • the myofilament phosphorylation targets may be selected from the group listed in Tables 2-5.
  • the phosphorylation targets of GSK-3P are selected from Tnl, MyBPC, TnT, MLC2, cMyBP-C, cTnl, a-Tropomyosin, titin, obscurin, Actin binding LIM protein 1 (AbLIMl), Tensin-1, Thyroid hormone receptor-associated protein 3 (THRAP-3), Nestin, Sorbin and SH3 domain-containing protein 2 (SORB-2), LIM domain only protein 7 (LMO-7), LIM domain-binding protein 3 (LDB3/Cypher), Striated muscle preferentially expressed kinase (SPEG), Filamin-C, Myotilin 1, and Leiomodin-2.
  • the phosphorylation targets GSK-3P may be selected from titin, obscurin, AbLIMl, Tensin-1, THRAP-3, Nestin, SORB-2, LMO-7, LDB3, SPEG, Filamin-C, Myotilin 1, and Leiomodin 2.
  • the cardiac disorder is heart failure or ventricular
  • the subject is a mammal, optionally a human.
  • the expression level, level of activity, or phosphorylation status of at least two (e.g., at least three, four, five, six, seven, or eight) biomarkers is determined.
  • any of the methods described herein can further include generating a risk score related to the expression level, level of activity, or phosphorylation status (e.g., level of phosphorylation) of any of the biomarkers described herein.
  • the risk score can be used, e.g., to classify a subject as one in need of treatment, one at risk for developing a cardiac disorder, or one who has benefited from therapy.
  • Figure 1 shows a timeline of pacing protocols in each of the canine models: Control, dyssynchronous heart failure (HFd ys ), cardiac resynchronization therapy (CRT), synchronous heart failure (HF sync ), V3A3 (3 weeks RV pacing to induce dyssynchrony, 3 weeks atrial pacing to restore synchrony), and AVA (synchronous heart failure with two weeks of induced transient dyssynchrony via RV pacing).
  • the HFd ys and CRT models receive left bundle branch block via radio-frequency ablation. After a one-week recovery period from the pacemaker implant, each pacing protocol lasts 6 weeks before the animal is sacrificed.
  • FIGS. 2A-2G show that CRT globally restores myofilament function.
  • F max force normalized to maximum calcium- activated force
  • HF dys causes desensitization to calcium (increase in EC 50 ), but is restored to Control levels with CRT.
  • Figures 3A-3D show that skinned myocytes from the left anterior septum exhibited the same functional changes as those from the LV lateral wall.
  • A, B Force-calcium data and curves, showing (A) actual force and (B) normalized force.
  • HF dys was desensitized to calcium, but CRT restored the value to near the Con value. *, P ⁇ 0.05 vs. Con. #, P ⁇ 0.05 vs HF dys .
  • Figures 4A-4D show that always synchronous heart failure (HF sync ) exhibited reduced myofilament function.
  • C Maximum force was reduced similarly in both HF dys and HF sync .
  • D HF sync exhibited a hyper-sensitization to calcium. *, P ⁇ 0.05 vs Con. #, P ⁇ 0.05
  • Figures 5A-5D show that myofilament function can be improved via a brief period of induced dyssynchrony.
  • A, B Both maximum force and calcium sensitivity are improved in the HF sync model when the middle two weeks of the pacing protocol is made dyssynchronous (AVA model).
  • C, D When dyssynchrony is induced via right ventricular pacing (instead of radio frequency ablation of the left bundle branch), and resynchronization is restored by moving the pacing site to the atrium (V3A3), the effects of CRT are recapitulated.
  • C Dyssynchrony caused a decrease in maximum force (F max ), which was increased by V3A3.
  • Figures 6A-6E show that there were no changes in protein degradation or isoforms between Control, HF dys , and CRT.
  • FIG. 7A-7E shows that the apparent decrease in F max is due to myocyte
  • Values are mean ⁇ s.e.m. *, P ⁇ 0.05 vs. Control, #, P ⁇ 0.05 vs. HF dys by one-way ANOVA.
  • Figures 8A-8F show that calcium sensitization is mediated by phosphorylation, but not at the known targets.
  • A Representative force-calcium curves for Con, HF dys , and CRT before protein phosphatase- 1 (PP1) and post treatment.
  • C Top, phos-Tag gel blotted for troponin I (Tnl).
  • Figure 10 shows that the phos-Tag gel (Figure 8C) showed that overall Tnl phosphorylation was decreased in HFd ys and CRT, but could not show whether the PKA or PKC sites specifically were decreasing.
  • PKA phosphorylation was unchanged between HF dys and CRT, as was total phosphorylation, it suggests that there was also no change in any of the other highly phosphorylated sites.
  • FIG 11 shows that, in order to confirm there were no phosphorylation changes among the most abundant myofilament proteins between HFd ys and CRT, 2D-DiGE gels were performed.
  • the two images represent the HF dys (top) and CRT (bottom) channels that were differentially labeled with Cy5 and Cy3, respectively, and run simultaneously on the same gel.
  • the two channels have been independently presented here for clarity.
  • the boxes indicate a selection of myofilament proteins. Note: troponin I is not represented on these 2D gels because it has a very high pi.
  • Figures 12A-12C show mass spectrometry (MS) data for phosphorylation at Titin (N2BA isoform) S9901.
  • MS mass spectrometry
  • N2BA isoform
  • A MS spectra for the peptide containing S9901.
  • B Fragmentation Table.
  • C Known domains and protein binding regions shown with location of the identified phosphorylation site.
  • Figures 13A-13C show mass spectrometry (MS) data for phosphorylation at Obscurin S4809.
  • MS mass spectrometry
  • A MS spectra for the peptide containing S4809.
  • B Fragmentation Table.
  • C Known domains and protein binding regions shown with location of the identified
  • Figures 14A-14C show mass spectrometry (MS) data for phosphorylation at ABLIM1 S421.
  • MS spectra for the peptide containing S421.
  • B Fragmentation Table.
  • C Known domains and protein binding regions shown with location of the identified phosphorylation site.
  • Figures 15A-15C show mass spectrometry (MS) data for phosphorylation at Tensin-1 S1274.
  • B Fragmentation Table.
  • C Known domains and protein binding regions shown with location of the identified phosphorylation site.
  • Figures 16A-16C show mass spectrometry (MS) data for phosphorylation at TRAP3
  • Figures 17A-17C show mass spectrometry (MS) data for phosphorylation at Nestin S 1062.
  • MS mass spectrometry
  • A MS spectra for the peptide containing S 1062.
  • B Fragmentation Table.
  • C Known domains and protein binding regions shown with location of the identified phosphorylation site.
  • Figures 18A-18C show mass spectrometry (MS) data for phosphorylation at SORB2 S40.
  • MS mass spectrometry
  • A MS spectra for the peptide containing S40.
  • B Fragmentation Table.
  • C Known domains and protein binding regions shown with location of the identified phosphorylation site.
  • FIGS 19A-19C show mass spectrometry (MS) data for phosphorylation at SORB2 S231.
  • MS mass spectrometry
  • A MS spectra for the peptide containing S231.
  • B Fragmentation Table.
  • C Known domains and protein binding regions shown with location of the identified phosphorylation site.
  • Figures 20A-20C show mass spectrometry (MS) data for phosphorylation at SORB2 S307.
  • MS mass spectrometry
  • A MS spectra for the peptide containing S307.
  • B Fragmentation Table.
  • C Known domains and protein binding regions shown with location of the identified phosphorylation site.
  • Figures 21A-21C show mass spectrometry (MS) data for phosphorylation at LM07
  • SI 346 (A) MS spectra for the peptide containing SI 346. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site.
  • Figures 22A-22C show mass spectrometry (MS) data for phosphorylation at LDB3 (Cypher/Zasp) S507.
  • MS mass spectrometry
  • A MS spectra for the peptide containing S507.
  • B Fragmentation
  • FIGS 23A-23C show mass spectrometry (MS) data for phosphorylation at SPEG S2039.
  • MS mass spectrometry
  • A MS spectra for the peptide containing SS2039.
  • B Fragmentation Table.
  • C Known domains and protein binding regions shown with location of the identified phosphorylation site.
  • FIGS 24A-24C show mass spectrometry (MS) data for phosphorylation at Filamin- C S2228.
  • MS mass spectrometry
  • A MS spectra for the peptide containing S2228.
  • B Fragmentation Table.
  • C Known domains and protein binding regions shown with location of the identified
  • FIGS 25A-25C show mass spectrometry (MS) data for phosphorylation at Myotilin S231.
  • MS mass spectrometry
  • A MS spectra for the peptide containing S231.
  • B Fragmentation Table.
  • C Known domains and protein binding regions shown with location of the identified phosphorylation site.
  • Figures 26A-26C show mass spectrometry (MS) data for phosphorylation at
  • Leiomodin-2 S513. MS spectra for the peptide containing S513.
  • B Fragmentation Table.
  • C Known domains and protein binding regions shown with location of the identified phosphorylation site.
  • Figures 27A-27C show increased phosphorylation at GSK-3P sites in CRT mediated the calcium sensitizing effect.
  • A Schematic of the cardiac sarcomere showing Z-disk (vertical lines), titin (labeled; diagonal lines), thick filament (branched structures), M-line (middle/center ), and thin filament (horizontal lines). The proteins (and sites) that were identified as increased with CRT are shown, with their best-known localization. *, matches GSK-3P consensus sequence.
  • B Representative force-calcium relationships in Con (top), HF dys (middle) and CRT (bottom) before (solid line) and after GSK-3P treatment (dashed line).
  • Figures 28A-28B show the effect of PKA treatment on HF dys and CRT myocytes.
  • Figures 29A-29E show the effect of Akt treatment on HF dys and CRT myocytes.
  • a and B Akt treatment had no effect on either F max or EC 50 in either group.
  • C Akt did not induce any detectable phosphorylation of myofilament targets, by Pro-Q Diamond phospho- stain.
  • D Akt treatment did induce phosphorylation of known targets in both groups, as shown by phosphorylation of serine 9 on GSK-3P (cytosolic fraction).
  • Figures 30A-30E show the time course of function and phosphorylation during dyssynchrony and heart failure.
  • A Endocardial biopsies taken at the 3-week time point (n, Con: 8, HF dys 3 weeks: 11, HF sync 3 weeks: 5, HF dys 6 weeks: 17, HF sync 6 weeks: 18 myocytes) showed that F max decreased steadily over the entire 6-week protocol.
  • B Calcium sensitivity was altered in HF sync by 3 weeks, with no changes between 3 and 6 weeks.
  • Figures 31A-31E show the details of an embodiment of a mouse cardiac pacemaker.
  • Figures 31A-31B show a mouse pacemaker chip (Figure 31 A) and a pacing lead (Figure 3 IB).
  • Figure 31C shows an ECG confirming capture of cardiac pacing data.
  • Figure 3 ID shows an echo showing dyssynchrony with RV pacing.
  • Figure 3 IE shows that RV pacing decreases systolic function.
  • FIGs 32A-32C show results of cardiac paced mice. Cardiac dyssynchrony was induced in mice using a custom RV pacing lead and pacemaker system.
  • Figure 32A shows that RV pacing, causing dyssynchrony, caused calcium desensitization compared to those mice with an implanted pacing lead, but no pacing. There was no effect on maximum force, however, with RV pacing. This replicates what was observed in the canine model of dyssynchrony, which resulted in reduced calcium sensitivity, as a consequence of reduced phosphorylation of GSK-3P's myofilament targets.
  • Figure 32B shows that with RV pacing, there was an increase in the ratio of phospho-serine 9 GSK-3P to total GSK-3P, indicating kinase de-activation in dyssynchrony. That both GSK-3P de-activation and calcium desensitization occurred in the mouse model with dyssynchrony but no heart failure, supports that these phenotypes are specific to cardiac dyssynchrony.
  • Figure 32C shows that genetic knock-out of GSK-3P in a mouse results in myofilament Ca 2+ desensitization. Skinned myocytes from inducible cardiomyocyte-specific GSK-3P knockout mice (Woulfe, Circ Res, 2010) were compared to myocytes from mice expressing WT GSK-3p.
  • Figure 33 shows an embodiment of a pacing protocol.
  • Figure 34 shows the expected results from the pacing protocol shown in Figure 33 and related experiments described herein.
  • Figure 35A-35D show GSK-3P expression results.
  • Figure 36A shows that GSK-3P is present in myo filament-enriched samples.
  • Figure 36B shows GSK-3P myofilament localization.
  • Figure 36C shows that this localization is with GSK-3P de-activation.
  • Figure 36D shows cardiac myofilament electron microscopy (EM).
  • Figure 36A-36B show biological relationships between GSK-3P, PKCa, p90 rsk , and Akt.
  • Figure 36A shows increased PKCa and p90 rsk activity with HF dys .
  • Figure 36B shows that Akt has no effect on myofilament function.
  • Figure 37 shows a schematic diagram of GSK-3P targets with known binding domains.
  • the invention relates, at least in part, to the discovery that the kinase glycogen synthase kinase 3 ⁇ (GSK-3P) is specifically de-activated in ventricular dyssynchrony. This de-activation occurs via its phosphorylation at serine 9. This de-activation is reversed in response to CRT, and does not occur in synchronous heart failure, indicating that it is a specific biomarker for cardiac dyssynchrony. While a biomarker for GSK-3P activation would require a cardiac biopsy, GSK-3P has several myofilament targets, which have been discovered herein to be detectable in the blood when the myocardium is damaged.
  • phosphorylation sites therefore represent biomarkers correlating with myocardial GSK-3P activity and cardiac dyssynchrony.
  • these phosphorylation sites are detected with very high sensitivity using multiple reaction monitoring (MRM).
  • MRM multiple reaction monitoring
  • GSK-3P activity and the myofilament phosphorylation targets represent a useful tool for drug screening. Allowing for in vitro testing of therapeutics to replace CRT, or translate CRT to a more diverse patient population. Knowing the phosphorylation status of one or more of these proteins (e.g. , in biopsies or in circulating blood) provides molecular insights into whether an individual will be a responder to therapy.
  • embodiments of the present invention can make use of well-known reagents and techniques for determining the status of a biomarker described herein (e.g. , publicly available antibody, mass spectrometry, nucleic acid probe, and similar reagents).
  • routine adaptions of the methods described herein such as assaying the presence of phosphorylation or quantitative increase or decrease in levels over time, over different courses of therapy, in therapeutic agent screening protocols, and the like, are contemplated.
  • assays for screening test agents to determine whether they activate the GSK-3P pathway and mimic CRT cellular contractile effects are contemplated.
  • an element means one element or more than one element.
  • the amount of a biomarker in a subject is "significantly" higher or lower than the normal amount of the biomarker, if the amount of the biomarker is greater or less,
  • the amount of the biomarker in the subject can be considered "significantly" higher or lower than the normal amount if the amount is at least about two, and preferably at least about three, four, or five times, higher or lower, respectively, than the normal amount of the biomarker.
  • signaling can also be applied to any other measured parameter described herein, such as for expression, inhibition, phosphorylation, and the like.
  • altered level of expression of a biomarker refers to an expression level or copy number of the biomarker in a test sample, that is greater or less than the standard error of the assay employed to assess expression or copy number, and is preferably at least twice, and more preferably three, four, five or ten or more times the expression level or copy number of the biomarker in a control sample (e.g., a sample from a healthy subject) and preferably, the average expression level or copy number of the biomarker in several control samples.
  • a control sample e.g., a sample from a healthy subject
  • the altered level of expression is greater or less than the standard error of the assay employed to assess expression or copy number, and is preferably at least twice, and more preferably three, four, five or ten or more times the expression level or copy number of the biomarker in a control sample (e.g., sample from a healthy subjects not having the associated disease) and preferably, the average expression level or copy number of the biomarker in several control samples.
  • a control sample e.g., sample from a healthy subjects not having the associated disease
  • This also applies to changes in the level of phosphorylation or phosphorylation status of a biomarker protein.
  • altered activity of a biomarker refers to an activity of the biomarker which is increased or decreased in a disease state as compared to the activity of the biomarker in a normal, control sample.
  • Altered activity of the biomarker may be the result of, for example, altered expression of the biomarker, altered phosphorylation the biomarker, or, e.g., an altered interaction with other proteins involved in the same or different pathway as the biomarker or altered interaction with transcriptional activators or inhibitors.
  • antibody broadly encompass naturally-occurring forms of antibodies (e.g. IgG, IgA, IgM, IgE) and recombinant antibodies such as single-chain antibodies, chimeric and humanized antibodies and multi- specific antibodies, as well as fragments and derivatives of all of the foregoing, which fragments and derivatives have at least an antigenic binding site.
  • Antibody derivatives may comprise a protein or chemical moiety conjugated to an antibody.
  • Antibodies may be polyclonal or monoclonal; xenogeneic, allogeneic, or syngeneic; or modified forms thereof (e.g. humanized, chimeric, etc.). Antibodies may also be fully human. Preferably, antibodies of the invention bind specifically or substantially specifically to a biomarker polypeptide or fragment thereof.
  • monoclonal antibody composition refers to a population of antibody polypeptides that contain only one species of an antigen binding site capable of
  • biomarker refers to a measurable entity of the present invention that has been determined to be predictive of a cardiac response. Biomarkers can include, without limitation, nucleic acids, proteins, and metabolites, particularly those shown in Tables 2-6.
  • body fluid refers to fluids that are excreted or secreted from the body as well as fluids that are normally not (e.g., bronchoalveolar lavage fiuid, amniotic fluid, aqueous humor, bile, blood and blood plasma, cerebrospinal fluid, cerumen and earwax, cowper's f uid or pre-ejaculatory fluid, chyle, chyme, stool, female ejaculate, interstitial fluid, intracellular f uid, lymph, menses, breast milk, mucus, pleural f uid, pus, saliva, sebum, semen, serum, sweat, synovial fluid, tears, urine, vaginal lubrication, vitreous humor, vomit).
  • bronchoalveolar lavage fiuid e.g., amniotic fluid, aqueous humor, bile, blood and blood plasma, cerebrospinal fluid, cerumen and earwax
  • complementary refers to the broad concept of sequence complementarity between regions of two nucleic acid strands or between two regions of the same nucleic acid strand. It is known that an adenine residue of a first nucleic acid region is capable of forming specific hydrogen bonds ("base pairing") with a residue of a second nucleic acid region which is antiparallel to the first region if the residue is thymine or uracil. Similarly, it is known that a cytosine residue of a first nucleic acid strand is capable of base pairing with a residue of a second nucleic acid strand which is antiparallel to the first strand if the residue is guanine.
  • a first region of a nucleic acid is complementary to a second region of the same or a different nucleic acid if, when the two regions are arranged in an antiparallel fashion, at least one nucleotide residue of the first region is capable of base pairing with a residue of the second region.
  • the first region comprises a first portion and the second region comprises a second portion, whereby, when the first and second portions are arranged in an antiparallel fashion, at least about 50%, and preferably at least about 75%, at least about 90%, or at least about 95%) of the nucleotide residues of the first portion are capable of base pairing with nucleotide residues in the second portion. More preferably, all nucleotide residues of the first portion are capable of base pairing with nucleotide residues in the second portion.
  • control refers to any reference standard suitable to provide a comparison to the expression products in the test sample.
  • the control comprises obtaining a "control sample” from which expression product levels are detected and compared to the expression product levels from the test sample.
  • a control sample may comprise any suitable sample, including but not limited to a sample from a control patient (can be stored sample or previous sample measurement) with a known outcome; normal tissue, blood, or cells isolated from a subject, such as a normal patient or the patient with a known condition, cultured primary cells/tissues isolated from a subject such as a normal subject or the patient with a known condition, a tissue or cell sample isolated from a normal subject, or primary cells/blood/tissues obtained from a depository.
  • control may comprise a reference standard expression product level from any suitable source, including but not limited to housekeeping genes, an expression product level range from normal tissue (or other previously analyzed control sample), a previously determined expression product level range within a test sample from a group of patients, or a set of patients with a certain outcome (for example, survival for one, two, three, four years, etc.) or receiving a certain treatment (for example, standard of care therapy).
  • a certain outcome for example, survival for one, two, three, four years, etc.
  • a certain treatment for example, standard of care therapy
  • control samples and reference standard expression product levels can be used in combination as controls in the methods of the present invention.
  • control may comprise normal cells/blood/tissue.
  • control may comprise an expression level for a set of patients, such as a set of patients with a known condition, or for a set of patients receiving a certain treatment, or for a set of patients with one outcome versus another outcome.
  • specific expression product level of each patient can be assigned to a percentile level of expression, or expressed as either higher or lower than the mean or average of the reference standard expression level.
  • control may comprise normal cells, blood, or tissue from patients treated with therapy.
  • control may also comprise a measured value for example, average level of expression of a particular gene in a population compared to the level of expression of a housekeeping gene in the same population.
  • control comprises a ratio transformation of expression product levels, including but not limited to determining a ratio of expression product levels of two genes in the test sample and comparing it to any suitable ratio of the same two genes in a reference standard; determining expression product levels of the two or more genes in the test sample and determining a difference in expression product levels in any suitable control; and determining expression product levels of the two or more genes in the test sample, normalizing their expression to expression of housekeeping genes in the test sample, and comparing to any suitable control.
  • control comprises a control sample which is of the same lineage and/or type as the test sample.
  • control may comprise expression product levels grouped as percentiles within or based on a set of patient samples, such as all patients with a certain condition.
  • a control expression product level is established wherein higher or lower levels of expression product relative to, for instance, a particular percentile, are used as the basis for predicting outcome.
  • a control expression product level is established using expression product levels from control patients with a known outcome, and the expression product levels from the test sample are compared to the control expression product level as the basis for predicting outcome.
  • the methods of the invention are not limited to use of a specific cut-point in comparing the level of expression product in the test sample to the control.
  • expression signature refers to a group of two or more coordinately expressed biomarkers.
  • the genes, proteins, metabolites, and the like making up this signature may be expressed in a specific cell lineage or during a particular biological response.
  • Expression data and gene expression levels can be stored on computer readable media, e.g., the computer readable medium used in conjunction with a microarray or chip reading device, or in conjunction with a qPCR detection system. Such expression data can be manipulated to generate expression signatures.
  • interaction when referring to an interaction between two molecules, refers to the physical contact (e.g., binding) of the molecules with one another.
  • An interaction may cause phosphorylation or Dephosphorylation of a molecule.
  • an interaction results in an activity (which produces a biological effect) of one or both of said molecules.
  • isolated protein refers to a protein that is substantially free of other proteins, cellular material, separation medium, and culture medium when isolated from cells or produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized.
  • isolated or purified protein or biologically active portion thereof is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the antibody, polypeptide, peptide or fusion protein is derived, or substantially free from chemical precursors or other chemicals when chemically synthesized.
  • substantially free of cellular material includes preparations of a biomarker polypeptide or fragment thereof, in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly produced.
  • the language "substantially free of cellular material” includes preparations of a biomarker protein or fragment thereof, having less than about 30% (by dry weight) of non-biomarker protein (also referred to herein as a "contaminating protein”), more preferably less than about 20% of non-biomarker protein, still more preferably less than about 10% of non-biomarker protein, and most preferably less than about 5% non-biomarker protein.
  • non-biomarker protein also referred to herein as a "contaminating protein”
  • polypeptide, peptide or fusion protein or fragment thereof e.g., a biologically active fragment thereof
  • it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the protein preparation.
  • the "normal" level of expression of a biomarker is the level of expression of the biomarker in cells of a subject, e.g., a healthy human patient.
  • An “over-expression” or “significantly higher level of expression” of a biomarker refers to an expression level in a test sample that is greater than the standard error of the assay employed to assess expression, and is preferably at least twice, and more preferably 2.1 , 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20 times or more higher than the expression activity or level of the biomarker in a control sample (e.g.
  • a "significantly lower level of expression" of a biomarker refers to an expression level in a test sample that is at least twice, and more preferably 2.1 , 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20 times or more lower than the expression level of the biomarker in a control sample (e.g. , sample from a healthy subject not having the biomarker associated disease) and preferably, the average expression level of the biomarker in several control samples.
  • a control sample e.g. , sample from a healthy subject not having the biomarker associated disease
  • an “over-expression” or “significantly higher level of expression” of a biomarker refers to an expression level in a test sample that is greater than the standard error of the assay employed to assess expression, and is preferably at least twice, and more preferably 2.1 , 2.2,
  • a "significantly lower level of expression" of a biomarker refers to an expression level in a test sample that is at least twice, and more preferably 2.1 , 2.2, 2.3,
  • prevent refers to reducing the probability of developing a disease, disorder, or condition in a subject, who does not have, but is at risk of or susceptible to developing a disease, disorder, or condition.
  • probe refers to any molecule which is capable of selectively binding to a specifically intended target molecule, for example, a nucleotide transcript or protein encoded by or corresponding to a biomarker nucleic acid. Probes can be either synthesized by one skilled in the art, or derived from appropriate biological preparations. For purposes of detection of the target molecule, probes may be specifically designed to be labeled, as described herein. Examples of molecules that can be utilized as probes include, but are not limited to, R A, DNA, proteins, antibodies, and organic molecules.
  • sample used for detecting or determining the presence or level of at least one biomarker is typically whole blood, plasma, serum, saliva, urine, stool (e.g., feces), tears, and any other bodily fluid (e.g., as described above under the definition of "body fluids"), or a tissue sample (e.g., biopsy) such as a surgical resection tissue.
  • body fluids e.g., as described above under the definition of "body fluids”
  • tissue sample e.g., biopsy
  • the method of the present invention further comprises obtaining the sample from the individual prior to detecting or determining the presence or level of at least one marker in the sample.
  • subject refers to any animal, mammal or human.
  • subject is interchangeable with “patient.”
  • therapeutic effect refers to a local or systemic effect in animals, particularly mammals, and more particularly humans, caused by a pharmacologically active substance.
  • the term thus means any substance intended for use in the diagnosis, cure, mitigation, treatment or prevention of disease or in the enhancement of desirable physical or mental development and conditions in an animal or human.
  • therapeutically- effective amount means that amount of such a substance that produces some desired local or systemic effect at a reasonable benefit/risk ratio applicable to any treatment.
  • a therapeutically effective amount of a compound will depend on its therapeutic index, solubility, and the like.
  • certain compounds discovered by the methods of the present invention may be administered in a sufficient amount to produce a reasonable benefit/risk ratio applicable to such treatment.
  • terapéuticaally-effective amount and “effective amount” as used herein means that amount of a compound, material, or composition comprising a compound of the present invention which is effective for producing some desired therapeutic effect in at least a sub -population of cells in an animal at a reasonable benefit/risk ratio applicable to any medical treatment.
  • a “transcribed polynucleotide” or “nucleotide transcript” is a polynucleotide (e.g. an mRNA, hnRNA, a cDNA, or an analog of such RNA or cDNA) which is complementary to or homologous with all or a portion of a mature mRNA made by transcription of a biomarker nucleic acid and normal post-transcriptional processing (e.g. splicing), if any, of the RNA transcript, and reverse transcription of the RNA transcript.
  • a polynucleotide e.g. an mRNA, hnRNA, a cDNA, or an analog of such RNA or cDNA
  • Arginine AGA, ACG, CGA, CGC, CGG, CGT
  • Glycine Gly, G
  • GGC GGG, GGT
  • Serine (Ser, S) AGC, AGT, TCA, TCC, TCG, TCT
  • Trp Tryptophan
  • Tyrosine TAC, TAT
  • nucleotide triplet An important and well known feature of the genetic code is its redundancy, whereby, for most of the amino acids used to make proteins, more than one coding nucleotide triplet may be employed (illustrated above). Therefore, a number of different nucleotide sequences may code for a given amino acid sequence. Such nucleotide sequences are considered functionally equivalent since they result in the production of the same amino acid sequence in all organisms (although certain organisms may translate some sequences more efficiently than they do others). Moreover, occasionally, a methylated variant of a purine or pyrimidine may be found in a given nucleotide sequence. Such methylations do not affect the coding relationship between the trinucleotide codon and the corresponding amino acid.
  • nucleotide sequence of a DNA or R A encoding a biomarker nucleic acid can be used to derive the polypeptide amino acid sequence, using the genetic code to translate the DNA or RNA into an amino acid sequence.
  • corresponding nucleotide sequences that can encode the polypeptide can be deduced from the genetic code (which, because of its redundancy, will produce multiple nucleic acid sequences for any given amino acid sequence).
  • description and/or disclosure herein of a nucleotide sequence which encodes a polypeptide should be considered to also include description and/or disclosure of the amino acid sequence encoded by the nucleotide sequence.
  • description and/or disclosure of a polypeptide amino acid sequence herein should be considered to also include description and/or disclosure of all possible nucleotide sequences that can encode the amino acid sequence.
  • nucleic acid and amino acid sequence information for the loci and biomarkers of the present invention are well known in the art and readily available on publicly available databases, such as the National Center for
  • NCBI Biotechnology Information
  • biomarker amount and/or activity measurement(s) in a sample from a subject is compared to a predetermined control (standard) sample.
  • the control sample can be from the same subject or from a different subject.
  • the control sample can be a combination of samples from several different subjects.
  • the biomarker amount and/or activity measurement(s) from a subject is compared to a pre-determined level. This pre-determined level is typically obtained from normal samples.
  • a "pre-determined" biomarker amount and/or activity measurement(s) may be a biomarker amount and/or activity measurement(s) used to, by way of example only, evaluate a subject that may be selected for treatment, evaluate a response to a treatment.
  • a pre-determined biomarker amount and/or activity measurement(s) may be determined in populations of patients with or without a condition.
  • the pre-determined biomarker amount and/or activity measurement(s) can be a single number, equally applicable to every patient, or the pre- determined biomarker amount and/or activity measurement(s) can vary according to specific subpopulations of patients. Age, weight, height, and other factors of a subject may affect the pre-determined biomarker amount and/or activity measurement(s) of the individual.
  • the pre-determined biomarker amount and/or activity can be determined for each subject individually.
  • the amounts determined and/or compared in a method described herein are based on absolute measurements.
  • the amounts determined and/or compared in a method described herein are based on relative measurements, such as ratios (e.g., expression and/or activity of biomarkers to that of wild type biomarkers and expression and/or activity of a biomarker of interest normalized to that of a housekeeping gene).
  • the pre-determined biomarker amount and/or activity measurement(s) can be any suitable standard.
  • the pre-determined biomarker amount and/or activity measurement(s) can be obtained from the same or a different human for whom a patient selection is being assessed.
  • the pre-determined biomarker amount and/or activity measurement(s) can be obtained from a previous assessment of the same patient. In such a manner, the progress of the selection of the patient can be monitored over time.
  • the control can be obtained from an assessment of another human or multiple humans, e.g., selected groups of humans, if the subject is a human.
  • the extent of the selection of the human for whom selection is being assessed can be compared to suitable other humans, e.g., other humans who are in a similar situation to the human of interest, such as those suffering from similar or the same condition(s) and/or of the same ethnic group.
  • the change of biomarker amount and/or activity measurement(s) from the pre-determined level is about 0.5 fold, about 1.0 fold, about 1.5 fold, about 2.0 fold, about 2.5 fold, about 3.0 fold, about 3.5 fold, about 4.0 fold, about 4.5 fold, or about 5.0 fold or greater.
  • the fold change is less than about 1, less than about 5, less than about 10, less than about 20, less than about 30, less than about 40, or less than about 50.
  • the fold change in biomarker amount and/or activity measurement(s) compared to a predetermined level is more than about 1 , more than about 5, more than about 10, more than about 20, more than about 30, more than about 40, or more than about 50.
  • Body fluids refer to fluids that are excreted or secreted from the body as well as fluids that are normally not (e.g.
  • bronchoalevolar lavage fluid amniotic fluid, aqueous humor, bile, blood and blood plasma, cerebrospinal fluid, cerumen and earwax, cowper's fluid or pre- ejaculatory fluid, chyle, chyme, stool, female ejaculate, interstitial fluid, intracellular fluid, lymph, menses, breast milk, mucus, pleural fluid, pus, saliva, sebum, semen, serum, sweat, synovial fluid, tears, urine, vaginal lubrication, vitreous humor, vomit).
  • the subject and/or control sample is selected from the group consisting of cells, cell lines, histological slides, paraffin embedded tissues, biopsies, whole blood, nipple aspirate, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, and bone marrow.
  • the sample is blood, serum, plasma, or urine. In another embodiment, the sample is serum.
  • the samples can be collected from individuals repeatedly over a longitudinal period of time (e.g., once or more on the order of days, weeks, months, annually, biannually, etc.). Obtaining numerous samples from an individual over a period of time can be used to verify results from earlier detections and/or to identify an alteration in biological pattern as a result of, for example, disease progression, drug treatment, etc. For example, subject samples can be taken and monitored every month, every two months, or combinations of one, two, or three month intervals according to the invention.
  • biomarker amount and/or activity measurements of the subject obtained over time can be conveniently compared with each other, as well as with those of normal controls during the monitoring period, thereby providing the subject's own values, as an internal, or personal, control for long-term monitoring.
  • Sample preparation and separation can involve any of the procedures, depending on the type of sample collected and/or analysis of biomarker measurement(s).
  • Such procedures include, by way of example only, concentration, dilution, adjustment of pH, removal of high abundance polypeptides (e.g., albumin, gamma globulin, and transferrin, etc.), addition of preservatives and calibrants, addition of protease inhibitors, addition of denaturants, desalting of samples, concentration of sample proteins, extraction and purification of lipids.
  • the sample preparation can also isolate molecules that are bound in non-covalent complexes to other protein (e.g., carrier proteins).
  • carrier proteins e.g., albumin
  • This process may isolate those molecules bound to a specific carrier protein (e.g., albumin), or use a more general process, such as the release of bound molecules from all carrier proteins via protein denaturation, for example using an acid, followed by removal of the carrier proteins.
  • Removal of undesired proteins from a sample can be achieved using high affinity reagents, high molecular weight filters, ultracentrifugation and/or electrodialysis.
  • High affinity reagents include antibodies or other reagents (e.g., aptamers) that selectively bind to high abundance proteins.
  • Sample preparation could also include ion exchange chromatography, metal ion affinity
  • Molecular weight filters include membranes that separate molecules on the basis of size and molecular weight. Such filters may further employ reverse osmosis, nanofiltration, ultrafiltration and microfiltration.
  • Ultracentrifugation is a method for removing undesired polypeptides from a sample. Ultracentrifugation is the centrifugation of a sample at about 15,000-60,000 rpm while monitoring with an optical system the sedimentation (or lack thereof) of particles.
  • Electrodialysis is a procedure which uses an electromembrane or semipermable membrane in a process in which ions are transported through semi-permeable membranes from one solution to another under the influence of a potential gradient. Since the membranes used in electrodialysis may have the ability to selectively transport ions having positive or negative charge, reject ions of the opposite charge, or to allow species to migrate through a
  • Separation and purification in the present invention may include any procedure known in the art, such as capillary electrophoresis (e.g. , in capillary or on-chip) or chromatography (e.g., in capillary, column or on a chip).
  • Electrophoresis is a method which can be used to separate ionic molecules under the influence of an electric field. Electrophoresis can be conducted in a gel, capillary, or in a microchannel on a chip. Examples of gels used for electrophoresis include starch, acrylamide, polyethylene oxides, agarose, or combinations thereof.
  • a gel can be modified by its cross-linking, addition of detergents, or denaturants, immobilization of enzymes or antibodies (affinity electrophoresis) or substrates (zymography) and incorporation of a pH gradient.
  • capillaries used for electrophoresis include capillaries that interface with an electrospray.
  • CE Capillary electrophoresis
  • CZE capillary zone electrophoresis
  • CIEF capillary isoelectric focusing
  • cITP capillary isotachophoresis
  • CE electrochromatography
  • An embodiment to couple CE techniques to electrospray ionization involves the use of volatile solutions, for example, aqueous mixtures containing a volatile acid and/or base and an organic such as an alcohol or acetonitrile.
  • Capillary isotachophoresis is a technique in which the analytes move through the capillary at a constant speed but are nevertheless separated by their respective mobilities.
  • Capillary zone electrophoresis also known as free-solution CE (FSCE)
  • FSCE free-solution CE
  • CIEF Capillary isoelectric focusing
  • Chromatography can be based on the differential adsorption and elution of certain analytes or partitioning of analytes between mobile and stationary phases.
  • Different examples of chromatography include, but not limited to, liquid chromatography (LC), gas chromatography (GC), high performance liquid chromatography (HPLC), etc.
  • nucleic acid molecules that correspond to biomarker nucleic acids that encode a biomarker polypeptide or a portion of such a polypeptide.
  • nucleic acid molecule is intended to include DNA molecules (e.g., cDNA or genomic DNA) and RNA molecules (e.g., mRNA) and analogs of the DNA or RNA generated using nucleotide analogs.
  • the nucleic acid molecule can be single-stranded or double-stranded, but preferably is double-stranded DNA.
  • An "isolated" nucleic acid molecule is one which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid molecule.
  • an "isolated" nucleic acid molecule is free of sequences (preferably protein-encoding sequences) which naturally flank the nucleic acid (i.e., sequences located at the 5' and 3' ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived.
  • the isolated nucleic acid molecule can contain less than about 5 kB, 4 kB, 3 kB, 2 kB, 1 kB, 0.5 kB or 0.1 kB of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived.
  • an "isolated" nucleic acid molecule such as a cDNA molecule, can be substantially free of other cellular material or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • a biomarker nucleic acid molecule of the present invention can be isolated using standard molecular biology techniques and the sequence information in the database records described herein. Using all or a portion of such nucleic acid sequences, nucleic acid molecules of the invention can be isolated using standard hybridization and cloning techniques (e.g., as described in Sambrook et ah, ed., Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989).
  • a nucleic acid molecule of the invention can be amplified using cDNA, mRNA, or genomic DNA as a template and appropriate oligonucleotide primers according to standard PCR amplification techniques.
  • the nucleic acid molecules so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis.
  • oligonucleotides corresponding to all or a portion of a nucleic acid molecule of the invention can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
  • nucleic acid molecule of the invention can comprise only a portion of a nucleic acid sequence, wherein the full length nucleic acid sequence comprises a marker of the invention or which encodes a polypeptide corresponding to a marker of the invention.
  • nucleic acid molecules can be used, for example, as a probe or primer.
  • probe/primer typically is used as one or more substantially purified oligonucleotides.
  • the oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 7, preferably about 15, more preferably about 25, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, or 400 or more consecutive nucleotides of a biomarker nucleic acid sequence.
  • Probes based on the sequence of a biomarker nucleic acid molecule can be used to detect transcripts or genomic sequences corresponding to one or more markers of the invention.
  • the probe comprises a label group attached thereto, e.g., a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor.
  • a biomarker nucleic acid molecules that differ, due to degeneracy of the genetic code, from the nucleotide sequence of nucleic acid molecules encoding a protein which corresponds to the biomarker, and thus encode the same protein, are also contemplated.
  • DNA sequence polymorphisms that lead to changes in the amino acid sequence can exist within a population (e.g., the human population). Such genetic polymorphisms can exist among individuals within a population due to natural allelic variation. An allele is one of a group of genes which occur alternatively at a given genetic locus.
  • DNA polymorphisms that affect RNA expression levels can also exist that may affect the overall expression level of that gene (e.g., by affecting regulation or degradation).
  • allele refers to alternative forms of a gene or portions thereof. Alleles occupy the same locus or position on homologous chromosomes. When a subject has two identical alleles of a gene, the subject is said to be homozygous for the gene or allele. When a subject has two different alleles of a gene, the subject is said to be heterozygous for the gene or allele.
  • biomarker alleles can differ from each other in a single nucleotide, or several nucleotides, and can include substitutions, deletions, and insertions of nucleotides.
  • An allele of a gene can also be a form of a gene containing one or more mutations.
  • allelic variant of a polymorphic region of gene refers to an alternative form of a gene having one of several possible nucleotide sequences found in that region of the gene in the population.
  • allelic variant is meant to encompass functional allelic variants, non-functional allelic variants, SNPs, mutations and polymorphisms.
  • single nucleotide polymorphism refers to a polymorphic site occupied by a single nucleotide, which is the site of variation between allelic sequences.
  • the site is usually preceded by and followed by highly conserved sequences of the allele (e.g. , sequences that vary in less than 1/100 or 1/1000 members of a population).
  • a SNP usually arises due to substitution of one nucleotide for another at the polymorphic site.
  • SNPs can also arise from a deletion of a nucleotide or an insertion of a nucleotide relative to a reference allele.
  • the polymorphic site is occupied by a base other than the reference base.
  • the altered allele can contain a "C” (cytidine), “G” (guanine), or "A” (adenine) at the polymorphic site.
  • SNP's may occur in protein-coding nucleic acid sequences, in which case they may give rise to a defective or otherwise variant protein, or genetic disease. Such a SNP may alter the coding sequence of the gene and therefore specify another amino acid (a "missense” SNP) or a SNP may introduce a stop codon (a "nonsense” SNP).
  • SNP When a SNP does not alter the amino acid sequence of a protein, the SNP is called "silent.” SNP's may also occur in noncoding regions of the nucleotide sequence. This may result in defective protein expression, e.g., as a result of alternative spicing, or it may have no effect on the function of the protein.
  • the terms "gene” and “recombinant gene” refer to nucleic acid molecules comprising an open reading frame encoding a polypeptide corresponding to a marker of the invention. Such natural allelic variations can typically result in 1-5% variance in the nucleotide sequence of a given gene. Alternative alleles can be identified by
  • a biomarker nucleic acid molecule is at least 7, 15, 20, 25, 30, 40, 60, 80, 100, 150, 200, 250, 300, 350, 400, 450, 550, 650, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2200, 2400, 2600, 2800, 3000, 3500, 4000, 4500, or more nucleotides in length and hybridizes under stringent conditions to a nucleic acid molecule corresponding to a marker of the invention or to a nucleic acid molecule encoding a protein corresponding to a marker of the invention.
  • hybridizes under stringent conditions is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 60% (65%>, 70%>, 75%, 80%, preferably 85%) identical to each other typically remain hybridized to each other.
  • stringent conditions are known to those skilled in the art and can be found in sections 6.3.1-6.3.6 of Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989).
  • a preferred, non-limiting example of stringent hybridization conditions are hybridization in 6X sodium chloride/sodium citrate (SSC) at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 50- 65°C.
  • allelic variants of a nucleic acid molecule of the invention can exist in the population, the skilled artisan will further appreciate that sequence changes can be introduced by mutation thereby leading to changes in the amino acid sequence of the encoded protein, without altering the biological activity of the protein encoded thereby.
  • sequence changes can be introduced by mutation thereby leading to changes in the amino acid sequence of the encoded protein, without altering the biological activity of the protein encoded thereby.
  • a "non-essential" amino acid residue is a residue that can be altered from the wild-type sequence without altering the biological activity, whereas an "essential" amino acid residue is required for biological activity.
  • amino acid residues that are not conserved or only semi-conserved among homologs of various species may be non-essential for activity and thus would be likely targets for alteration.
  • amino acid residues that are conserved among the homologs of various species e.g. , murine and human
  • amino acid residues that are conserved among the homologs of various species may be essential for activity and thus would not be likely targets for alteration.
  • nucleic acid molecules encoding a polypeptide of the invention that contain changes in amino acid residues that are not essential for activity.
  • polypeptides differ in amino acid sequence from the naturally- occurring proteins which correspond to the markers of the invention, yet retain biological activity.
  • a biomarker protein has an amino acid sequence that is at least about 40% identical, 50%, 60%, 70%, 75%, 80%, 83%, 85%, 87.5%, 90%, 91%, 92%, 93%, 94%), 95%), 96%), 97%), 98%>, 99% or identical to the amino acid sequence of a biomarker protein described herein.
  • An isolated nucleic acid molecule encoding a variant protein can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of nucleic acids of the invention, such that one or more amino acid residue substitutions, additions, or deletions are introduced into the encoded protein. Mutations can be introduced by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis. Preferably, conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues.
  • a "conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine
  • non-polar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • beta-branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • mutations can be introduced randomly along all or part of the coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for biological activity to identify mutants that retain activity.
  • the encoded protein can be expressed recombinantly and the activity of the protein can be determined.
  • the present invention further contemplates the use of anti- biomarker antisense nucleic acid molecules, i.e., molecules which are complementary to a sense nucleic acid of the invention, e.g., complementary to the coding strand of a double- stranded cDNA molecule corresponding to a marker of the invention or complementary to an mR A sequence corresponding to a marker of the invention.
  • an antisense nucleic acid molecule of the invention can hydrogen bond to (i.e. anneal with) a sense nucleic acid of the invention.
  • the antisense nucleic acid can be complementary to an entire coding strand, or to only a portion thereof, e.g.
  • an antisense nucleic acid molecule can also be antisense to all or part of a non-coding region of the coding strand of a nucleotide sequence encoding a polypeptide of the invention.
  • the non-coding regions (“5' and 3' untranslated regions") are the 5' and 3' sequences which flank the coding region and are not translated into amino acids.
  • An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 or more nucleotides in length.
  • An antisense nucleic acid can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art.
  • an antisense nucleic acid e.g., an antisense oligonucleotide
  • an antisense nucleic acid e.g., an antisense oligonucleotide
  • modified nucleotides which can be used to generate the antisense nucleic acid include 5-fluorouracil, 5- bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5- (carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5- carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6- isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2- methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7- methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D- mannosylqueosine, 5'-methoxy
  • the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been sub-cloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).
  • the antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mR A and/or genomic DNA encoding a polypeptide corresponding to a selected marker of the invention to thereby inhibit expression of the marker, e.g., by inhibiting transcription and/or translation.
  • the hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule which binds to DNA duplexes, through specific interactions in the major groove of the double helix.
  • antisense nucleic acid molecules of the invention examples include direct injection at a tissue site or infusion of the antisense nucleic acid into a blood- or bone marrow- associated body fluid.
  • antisense nucleic acid molecules can be modified to target selected cells and then administered systemically.
  • antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface, e.g., by linking the antisense nucleic acid molecules to peptides or antibodies which bind to cell surface receptors or antigens.
  • the antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient intracellular concentrations of the antisense molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II or pol III promoter are preferred.
  • the nucleic acid molecules of the present invention can be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g., the stability, hybridization, or solubility of the molecule.
  • the deoxyribose phosphate backbone of the nucleic acid molecules can be modified to generate peptide nucleic acid molecules (see Hyrup et ah, 1996, Bioorganic & Medicinal Chemistry 4(1): 5-23).
  • peptide nucleic acids refer to nucleic acid mimics, e.g., DNA mimics, in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained.
  • the neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength.
  • the synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described in Hyrup et al. (1996), supra; Perry-O'Keefe et al. (1996) Proc. Natl. Acad. Sci. USA 93: 14670-675.
  • PNAs can be used in therapeutic and diagnostic applications.
  • PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, e.g., inducing transcription or translation arrest or inhibiting replication.
  • PNAs can also be used, e.g. , in the analysis of single base pair mutations in a gene by, e.g. , PNA directed PCR clamping; as artificial restriction enzymes when used in combination with other enzymes, e.g., SI nucleases (Hyrup (1996), supra; or as probes or primers for DNA sequence and hybridization (Hyrup, 1996, supra; Perry-O'Keefe et al, 1996, Proc. Natl. Acad. Sci. USA 93: 14670-675).
  • PNAs can be modified, e.g. , to enhance their stability or cellular uptake, by attaching lipophilic or other helper groups to PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art.
  • PNA-DNA chimeras can be generated which can combine the advantageous properties of PNA and DNA.
  • Such chimeras allow DNA recognition enzymes, e.g., RNASE H and DNA polymerases, to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity.
  • PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleobases, and orientation (Hyrup, 1996, supra).
  • the synthesis of PNA- DNA chimeras can be performed as described in Hyrup (1996), supra, and Finn et al. (1996) Nucleic Acids Res. 24(17):3357-63.
  • a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry and modified nucleoside analogs.
  • substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the protein is derived, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • substantially free of cellular material includes preparations of protein in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly produced.
  • protein that is substantially free of cellular material includes preparations of protein having less than about 30%, 20%, 10%>, or 5% (by dry weight) of heterologous protein (also referred to herein as a "contaminating protein").
  • the protein or biologically active portion thereof is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%>, 10%>, or 5% of the volume of the protein preparation.
  • culture medium represents less than about 20%>, 10%>, or 5% of the volume of the protein preparation.
  • the protein is produced by chemical synthesis, it is preferably substantially free of chemical precursors or other chemicals, i.e., it is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein. Accordingly such preparations of the protein have less than about 30%>, 20%>, 10%>, 5% (by dry weight) of chemical precursors or compounds other than the polypeptide of interest.
  • Biomarker polypeptides include polypeptides comprising amino acid sequences sufficiently identical to or derived from a biomarker protein amino acid sequence described herein, but which includes fewer amino acids than the full length protein, and exhibit at least one activity of the corresponding full-length protein.
  • biologically active portions comprise a domain or motif with at least one activity of the corresponding protein.
  • a biologically active portion of a protein of the invention can be a polypeptide which is, for example, 10, 25, 50, 100 or more amino acids in length.
  • other biologically active portions, in which other regions of the protein are deleted can be prepared by recombinant techniques and evaluated for one or more of the functional activities of the native form of a polypeptide of the invention.
  • Preferred polypeptides have an amino acid sequence of a biomarker protein encoded by a nucleic acid molecule described herein.
  • Other useful proteins are substantially identical (e.g., at least about 40%, preferably 50%, 60%, 70%, 75%, 80%, 83%, 85%, 88%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) to one of these sequences and retain the functional activity of the protein of the corresponding naturally-occurring protein yet differ in amino acid sequence due to natural allelic variation or mutagenesis.
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence).
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
  • the determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • a preferred, non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul (1990) Proc. Natl. Acad. Sci. USA 87:2264-2268, modified as in Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5877. Such an algorithm is incorporated into the
  • Gapped BLAST can be utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25:3389-3402.
  • PSI-Blast can be used to perform an iterated search which detects distant relationships between molecules.
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • Another preferred, non- limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, (1988) Comput Appl Biosci, 4: 11-7.
  • Such an algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package.
  • PAM120 weight residue table a gap length penalty of 12, and a gap penalty of 4 can be used.
  • Yet another useful algorithm for identifying regions of local sequence similarity and alignment is the FASTA algorithm as described in Pearson and Lipman (1988) Proc. Natl. Acad. Sci. USA 85:2444-2448.
  • a PAM120 weight residue table can, for example, be used with a k- tuple value of 2.
  • the percent identity between two sequences can be determined using techniques similar to those described above, with or without allowing gaps. In calculating percent identity, only exact matches are counted.
  • the invention also provides chimeric or fusion proteins corresponding to a biomarker protein.
  • a "chimeric protein” or “fusion protein” comprises all or part (preferably a biologically active part) of a polypeptide corresponding to a marker of the invention operably linked to a heterologous polypeptide (i.e., a polypeptide other than the polypeptide corresponding to the marker).
  • a heterologous polypeptide i.e., a polypeptide other than the polypeptide corresponding to the marker.
  • the term "operably linked” is intended to indicate that the polypeptide of the invention and the heterologous polypeptide are fused in- frame to each other.
  • the heterologous polypeptide can be fused to the amino-terminus or the carboxyl-terminus of the polypeptide of the invention.
  • One useful fusion protein is a GST fusion protein in which a polypeptide
  • fusion proteins can facilitate the purification of a recombinant polypeptide of the invention.
  • the fusion protein contains a heterologous signal sequence, immunoglobulin fusion protein, toxin, or other useful protein sequence.
  • Chimeric and fusion proteins of the invention can be produced by standard recombinant DNA techniques.
  • the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and re-amplified to generate a chimeric gene sequence (see, e.g., Ausubel et ah, supra).
  • fusion moiety e.g., a GST polypeptide
  • a nucleic acid encoding a polypeptide of the invention can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the polypeptide of the invention.
  • a signal sequence can be used to facilitate secretion and isolation of the secreted protein or other proteins of interest.
  • Signal sequences are typically characterized by a core of hydrophobic amino acids which are generally cleaved from the mature protein during secretion in one or more cleavage events.
  • Such signal peptides contain processing sites that allow cleavage of the signal sequence from the mature proteins as they pass through the secretory pathway.
  • the invention pertains to the described polypeptides having a signal sequence, as well as to polypeptides from which the signal sequence has been proteolytically cleaved (i.e., the cleavage products).
  • a nucleic acid sequence encoding a signal sequence can be operably linked in an expression vector to a protein of interest, such as a protein which is ordinarily not secreted or is otherwise difficult to isolate.
  • the signal sequence directs secretion of the protein, such as from a eukaryotic host into which the expression vector is transformed, and the signal sequence is subsequently or concurrently cleaved.
  • the protein can then be readily purified from the extracellular medium by art recognized methods.
  • the signal sequence can be linked to the protein of interest using a sequence which facilitates purification, such as with a GST domain.
  • the present invention also pertains to variants of the biomarker polypeptides described herein.
  • Such variants have an altered amino acid sequence which can function as either agonists (mimetics) or as antagonists.
  • Variants can be generated by mutagenesis, e.g., discrete point mutation or truncation.
  • An agonist can retain substantially the same, or a subset, of the biological activities of the naturally occurring form of the protein.
  • An antagonist of a protein can inhibit one or more of the activities of the naturally occurring form of the protein by, for example, competitively binding to a downstream or upstream member of a cellular signaling cascade which includes the protein of interest.
  • specific biological effects can be elicited by treatment with a variant of limited function. Treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein can have fewer side effects in a subject relative to treatment with the naturally occurring form of the protein.
  • Variants of a biomarker protein which function as either agonists (mimetics) or as antagonists can be identified by screening combinatorial libraries of mutants, e.g., truncation mutants, of the protein of the invention for agonist or antagonist activity.
  • a variegated library of variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library.
  • a variegated library of variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential protein sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins ⁇ e.g., for phage display).
  • a degenerate set of potential protein sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins ⁇ e.g., for phage display.
  • a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of the coding sequence of interest with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA to form double stranded DNA which can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with S 1 nuclease, and ligating the resulting fragment library into an expression vector.
  • an expression library can be derived which encodes amino terminal and internal fragments of various sizes of the protein of interest.
  • REM Recursive ensemble mutagenesis
  • biomarker nucleic acid and/or biomarker polypeptide molecules described herein can be facilitated by using standard recombinant techniques.
  • such techniques use vectors, preferably expression vectors, containing a nucleic acid encoding a biomarker polypeptide or a portion of such a polypeptide.
  • vector refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • plasmid refers to a circular double stranded DNA loop into which additional DNA segments can be ligated.
  • viral vector Another type of vector, wherein additional DNA segments can be ligated into the viral genome.
  • vectors are capable of autonomous replication in a host cell into which they are introduced ⁇ e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • Other vectors ⁇ e.g., non-episomal mammalian vectors
  • certain vectors namely expression vectors, are capable of directing the expression of genes to which they are operably linked.
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids (vectors).
  • the present invention is intended to include such other forms of expression vectors, such as viral vectors ⁇ e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
  • the recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell.
  • the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which is operably linked to the nucleic acid sequence to be expressed.
  • operably linked is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell).
  • regulatory sequence is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, Methods in Enzymology: Gene Expression
  • Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in many types of host cell and those which direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, and the like.
  • the expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein.
  • the recombinant expression vectors for use in the invention can be designed for expression of a polypeptide corresponding to a marker of the invention in prokaryotic (e.g. , E. coli) or eukaryotic cells (e.g., insect cells ⁇ using baculovirus expression vectors ⁇ , yeast cells or mammalian cells). Suitable host cells are discussed further in Goeddel, supra.
  • prokaryotic e.g. , E. coli
  • eukaryotic cells e.g., insect cells ⁇ using baculovirus expression vectors ⁇ , yeast cells or mammalian cells. Suitable host cells are discussed further in Goeddel, supra.
  • the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
  • Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein.
  • Such fusion vectors typically serve three purposes: 1) to increase expression of recombinant protein; 2) to increase the solubility of the recombinant protein; and 3) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification.
  • a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein.
  • enzymes, and their cognate recognition sequences include Factor Xa, thrombin and enterokinase.
  • Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith and Johnson, 1988, Gene 67:31-40), pMAL (New England Biolabs, Beverly, MA) and pRIT5 (Pharmacia, Piscataway, NJ) which fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein.
  • GST glutathione S-transferase
  • maltose E binding protein or protein A, respectively, to the target recombinant protein.
  • Suitable inducible non-fusion E. coli expression vectors include pTrc (Amann et al, 1988, Gene 69:301-315) and pET l id (Studier et al, p. 60-89, In Gene Expression Technology: Methods in Enzymology vol.185, Academic Press, San Diego, CA, 1991).
  • Target biomarker nucleic acid expression from the pTrc vector relies on host RNA polymerase transcription from a hybrid trp-lac fusion promoter.
  • Target biomarker nucleic acid expression from the pET l id vector relies on transcription from a T7 gnlO-lac fusion promoter mediated by a co-expressed viral RNA polymerase (T7 gnl).
  • This viral polymerase is supplied by host strains BL21 (DE3) or HMS174(DE3) from a resident prophage harboring a T7 gnl gene under the transcriptional control of the lacUV 5 promoter.
  • the expression vector is a yeast expression vector.
  • yeast expression vectors for expression in yeast S. cerevisiae include pYepSecl (Baldari et al, 1987, EMBO J. 6:229-234), pMFa (Kurjan and Herskowitz, 1982, Cell 30:933-943), pJRY88 (Schultz et al, 1987, Gene 54:113-123), pYES2 (Invitrogen Corporation, San Diego, CA), and pPicZ (Invitrogen Corp, San Diego, CA).
  • the expression vector is a baculovirus expression vector.
  • Baculovirus vectors available for expression of proteins in cultured insect cells include the pAc series (Smith et al, 1983, Mol. Cell Biol. 3:2156-2165) and the pVL series (Lucklow and Summers, 1989, Virology 170:31-39).
  • a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector.
  • mammalian expression vectors include pCDM8 (Seed, 1987, Nature 329:840) and pMT2PC (Kaufman et al, 1987, EMBO J. 6: 187-195).
  • the expression vector's control functions are often provided by viral regulatory elements.
  • commonly used promoters are derived from polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40.
  • suitable expression systems for both prokaryotic and eukaryotic cells see chapters 16 and 17 of Sambrook et al. , supra.
  • the invention further provides a recombinant expression vector comprising a DNA molecule cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operably linked to a regulatory sequence in a manner which allows for expression (by transcription of the DNA molecule) of an R A molecule which is antisense to the mR A encoding a polypeptide of the invention.
  • Regulatory sequences operably linked to a nucleic acid cloned in the antisense orientation can be chosen which direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen which direct constitutive, tissue-specific or cell type specific expression of antisense RNA.
  • the antisense expression vector can be in the form of a recombinant plasmid, phagemid, or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced.
  • Another aspect of the invention pertains to host cells into which a recombinant expression vector of the invention has been introduced.
  • host cell and
  • progeny refers not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • a host cell can be any prokaryotic ⁇ e.g., E. coli) or eukaryotic cell (e.g., insect cells, yeast or mammalian cells).
  • prokaryotic ⁇ e.g., E. coli
  • eukaryotic cell e.g., insect cells, yeast or mammalian cells.
  • Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques.
  • transformation and “transfection” are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid into a host cell, including calcium phosphate or calcium chloride co- precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. ⁇ supra), and other laboratory manuals.
  • a gene that encodes a selectable marker ⁇ e.g., for resistance to antibiotics is generally introduced into the host cells along with the gene of interest.
  • selectable markers include those which confer resistance to drugs, such as G418, hygromycin and methotrexate.
  • Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die).
  • Biomarker nucleic acids and/or biomarker polypeptides can be analyzed according to the methods described herein and techniques known to the skilled artisan to identify such genetic or expression alterations useful for the present invention including, but not limited to, 1) an alteration in the level of a biomarker transcript or polypeptide, 2) a deletion or addition of one or more nucleotides from a biomarker gene, 4) a substitution of one or more nucleotides of a biomarker gene, 5) aberrant modification of a biomarker gene, such as an expression regulatory region, 6) an alteration in the phosphorylation status or level of phosphorylation of a biomarker protein described herein, and the like,
  • Biomarker expression may be assessed by any of a wide variety of well known methods for detecting expression of a transcribed molecule or protein.
  • Non-limiting examples of such methods include immunological methods for detection of secreted, cell-surface, cytoplasmic, or nuclear proteins, protein purification methods, protein function or activity assays, nucleic acid hybridization methods, nucleic acid reverse transcription methods, and nucleic acid amplification methods.
  • activity of a particular gene is characterized by a measure of gene transcript (e.g. m NA), by a measure of the quantity of translated protein, or by a measure of gene product activity.
  • Marker expression can be monitored in a variety of ways, including by detecting mRNA levels, protein levels, or protein activity, any of which can be measured using standard techniques. Detection can involve quantification of the level of gene expression (e.g., genomic DNA, cDNA, mRNA, protein, or enzyme activity), or,
  • detecting or determining expression levels of a biomarker and functionally similar homologs thereof, including a fragment or genetic alteration thereof (e.g., in regulatory or promoter regions thereof) comprises detecting or determining RNA levels for the marker of interest.
  • one or more cells from the subject to be tested are obtained and RNA is isolated from the cells.
  • a sample of breast tissue cells is obtained from the subject.
  • RNA is obtained from a single cell.
  • a cell can be isolated from a tissue sample by laser capture microdissection (LCM).
  • a cell can be isolated from a tissue section, including a stained tissue section, thereby assuring that the desired cell is isolated (see, e.g., Bonner et al. (1997) Science 278: 1481 ; Emmert- Buck et al. (1996) Science 274:998; Fend et al. (1999) Am. J. Path. 154: 61 and Murakami et al. (2000) Kidney Int. 58: 1346).
  • Murakami et al, supra describe isolation of a cell from a previously immunostained tissue section.
  • RNA can be extracted.
  • Methods for establishing cultures of non-transformed cells, i.e., primary cell cultures, are known in the art.
  • RNA in the tissue and cells may quickly become degraded.
  • the tissue or cells obtained from a subject is snap frozen as soon as possible.
  • RNA can be extracted from the tissue sample by a variety of methods, e.g., the guanidium thiocyanate lysis followed by CsCl centrifugation (Chirgwin et al, 1979,
  • RNA from single cells can be obtained as described in methods for preparing cDNA libraries from single cells, such as those described in Dulac, C. (1998) Curr. Top. Dev. Biol. 36, 245 and Jena et al. (1996) J. Immunol. Methods 190: 199. Care to avoid RNA degradation must be taken, e.g., by inclusion of RNAsin.
  • RNA sample can then be enriched in particular species.
  • poly(A)+ RNA is isolated from the RNA sample. In general, such purification takes advantage of the poly-A tails on mRNA.
  • poly-T poly-T
  • oligonucleotides may be immobilized within on a solid support to serve as affinity ligands for mRNA.
  • Kits for this purpose are commercially available, e.g. , the MessageMaker kit (Life Technologies, Grand Island, NY).
  • the RNA population is enriched in marker sequences. Enrichment can be undertaken, e.g., by primer-specific cDNA synthesis, or multiple rounds of linear amplification based on cDNA synthesis and template-directed in vitro transcription (see, e.g. , Wang et al. (1989) PNAS 86, 9717; Dulac et al., supra, and Jena et al., supra).
  • the population of RNA, enriched or not in particular species or sequences can further be amplified. As defined herein, an "amplification process" is designed to strengthen, increase, or augment a molecule within the RNA.
  • RNA is mRNA
  • an amplification process such as RT-PCR can be utilized to amplify the mRNA, such that a signal is detectable or detection is enhanced.
  • RT-PCR can be utilized to amplify the mRNA, such that a signal is detectable or detection is enhanced.
  • Such an amplification process is beneficial particularly when the biological, tissue, or tumor sample is of a small size or volume.
  • RNAscribe mRNA into cDNA followed by polymerase chain reaction RT-PCR
  • RT-AGLCR reverse transcribe mRNA into cDNA followed by symmetric gap ligase chain reaction
  • amplification methods which can be utilized herein include but are not limited to the so-called "NASBA” or “3SR” technique described in PNAS USA 87: 1874- 1878 (1990) and also described in Nature 350 (No. 6313): 91-92 (1991); Q-beta amplification as described in published European Patent Application (EPA) No. 4544610; strand displacement amplification (as described in G. T. Walker et al., Clin. Chem. 42: 9-13 (1996) and European Patent Application No.
  • Northern analysis involves running a preparation of RNA on a denaturing agarose gel, and transferring it to a suitable support, such as activated cellulose, nitrocellulose or glass or nylon
  • Radiolabeled cDNA or RNA is then hybridized to the preparation, washed and analyzed by autoradiography.
  • In situ hybridization visualization may also be employed, wherein a radioactively labeled antisense RNA probe is hybridized with a thin section of a biopsy sample, washed, cleaved with RNase and exposed to a sensitive emulsion for autoradiography.
  • the samples may be stained with hematoxylin to demonstrate the histological composition of the sample, and dark field imaging with a suitable light filter shows the developed emulsion.
  • Nonradioactive labels such as digoxigenin may also be used.
  • mRNA expression can be detected on a DNA array, chip or a microarray.
  • Labeled nucleic acids of a test sample obtained from a subject may be hybridized to a solid surface comprising biomarker DNA. Positive hybridization signal is obtained with the sample containing biomarker transcripts.
  • mRNA is extracted from the biological sample to be tested, reverse transcribed, and fluorescently-labeled cDNA probes are generated.
  • the microarrays capable of hybridizing to marker cDNA are then probed with the labeled cDNA probes, the slides scanned and fluorescence intensity measured. This intensity correlates with the hybridization intensity and expression levels.
  • probes that can be used in the methods described herein include cDNA, riboprobes, synthetic oligonucleotides and genomic probes.
  • the type of probe used will generally be dictated by the particular situation, such as riboprobes for in situ hybridization, and cDNA for Northern blotting, for example.
  • the probe is directed to nucleotide regions unique to the RNA.
  • the probes may be as short as is required to differentially recognize marker mRNA transcripts, and may be as short as, for example, 15 bases; however, probes of at least 17, 18, 19 or 20 or more bases can be used.
  • the primers and probes hybridize specifically under stringent conditions to a DNA fragment having the nucleotide sequence corresponding to the marker.
  • stringent conditions means hybridization will occur only if there is at least 95% identity in nucleotide sequences. In another embodiment, hybridization under "stringent conditions" occurs when there is at least 97% identity between the sequences.
  • the form of labeling of the probes may be any that is appropriate, such as the use of radioisotopes, for example, 32 P and 35 S. Labeling with radioisotopes may be achieved, whether the probe is synthesized chemically or biologically, by the use of suitably labeled bases.
  • the biological sample contains polypeptide molecules from the test subject.
  • the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject.
  • the methods further involve obtaining a control biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting marker polypeptide, mRNA, genomic DNA, or fragments thereof, such that the presence of the marker polypeptide, mRNA, genomic DNA, or fragments thereof, is detected in the biological sample, and comparing the presence of the marker polypeptide, mRNA, genomic DNA, or fragments thereof, in the control sample with the presence of the marker polypeptide, mRNA, genomic DNA, or fragments thereof in the test sample.
  • the activity or level of a biomarker protein can be detected and/or quantified by detecting or quantifying the expressed polypeptide.
  • the polypeptide can be detected and quantified by any of a number of means well known to those of skill in the art. Any method known in the art for detecting polypeptides can be used. Such methods include, but are not limited to, immunodiffusion, Immunoelectrophoresis, radioimmunoassay (RIA), enzyme- linked immunosorbent assays (ELISAs), immunofluorescent assays, Western blotting, binder- ligand assays, immunohistochemical techniques, agglutination, complement assays, high performance liquid chromatography (HPLC), thin layer chromatography (TLC),
  • hyperdiffusion chromatography and the like (e.g., Basic and Clinical Immunology, Sites and Terr, eds., Appleton and Lange, Norwalk, Conn, pp 217-262, 1991 which is incorporated by reference).
  • binder-ligand immunoassay methods including reacting antibodies with an epitope or epitopes and competitively displacing a labeled polypeptide or derivative thereof.
  • ELISA and RIA procedures may be conducted such that a desired biomarker protein standard is labeled (with a radioisotope such as 125 I or 35 S, or an assayable enzyme, such as horseradish peroxidase or alkaline phosphatase), and, together with the unlabelled sample, brought into contact with the corresponding antibody, whereon a second antibody is used to bind the first, and radioactivity or the immobilized enzyme assayed (competitive assay).
  • a radioisotope such as 125 I or 35 S, or an assayable enzyme, such as horseradish peroxidase or alkaline phosphatase
  • biomarker protein in the sample is allowed to react with the corresponding immobilized antibody, radioisotope- or enzyme-labeled anti- biomarker proteinantibody is allowed to react with the system, and radioactivity or the enzyme assayed (ELISA-sandwich assay).
  • radioactivity or the enzyme assayed ELISA-sandwich assay.
  • Other conventional methods may also be employed as suitable.
  • a “one-step” assay involves contacting antigen with immobilized antibody and, without washing, contacting the mixture with labeled antibody.
  • a “two-step” assay involves washing before contacting, the mixture with labeled antibody.
  • Other conventional methods may also be employed as suitable.
  • a method for measuring biomarker protein levels comprises the steps of: contacting a biological specimen with an antibody or variant (e.g., fragment) thereof which selectively binds the biomarker protein, and detecting whether said antibody or variant thereof is bound to said sample and thereby measuring the levels of the biomarker protein.
  • an antibody or variant e.g., fragment
  • Enzymatic and radiolabeling of biomarker protein and/or the antibodies may be effected by conventional means.
  • Such means will generally include covalent linking of the enzyme to the antigen or the antibody in question, such as by glutaraldehyde, specifically so as not to adversely affect the activity of the enzyme, by which is meant that the enzyme must still be capable of interacting with its substrate, although it is not necessary for all of the enzyme to be active, provided that enough remains active to permit the assay to be effected.
  • Enzymes employable for labeling are not particularly limited, but may be selected from the members of the oxidase group, for example. These catalyze production of hydrogen peroxide by reaction with their substrates, and glucose oxidase is often used for its good stability, ease of availability and cheapness, as well as the ready availability of its substrate
  • glucose activity of the oxidase may be assayed by measuring the concentration of hydrogen peroxide formed after reaction of the enzyme-labeled antibody with the substrate under controlled conditions well-known in the art.
  • Other techniques may be used to detect biomarker protein according to a practitioner's preference based upon the present disclosure.
  • One such technique is Western blotting (Towbin et at., Proc. Nat. Acad. Sci. 76:4350 (1979)), wherein a suitably treated sample is run on an SDS-PAGE gel before being transferred to a solid support, such as a nitrocellulose filter.
  • Anti-biomarker protein antibodies are then brought into contact with the support and assayed by a secondary immunological reagent, such as labeled protein A or antiimmunoglobulin (suitable labels including 125 I, horseradish peroxidase and alkaline phosphatase). Chromatographic detection may also be used.
  • a secondary immunological reagent such as labeled protein A or antiimmunoglobulin (suitable labels including 125 I, horseradish peroxidase and alkaline phosphatase). Chromatographic detection may also be used.
  • Immunohistochemistry may be used to detect expression of biomarker protein, e.g., in a biopsy sample.
  • a suitable antibody is brought into contact with, for example, a thin layer of cells, washed, and then contacted with a second, labeled antibody.
  • Labeling may be by fluorescent markers, enzymes, such as peroxidase, avidin, or radiolabelling. The assay is scored visually, using microscopy.
  • Anti- biomarker protein antibodies may also be used for imaging purposes, for example, to detect the presence of biomarker protein in cells and tissues of a subject.
  • Suitable labels include radioisotopes, iodine ( 125 I, 121 I), carbon ( 14 C), sulphur ( 35 S), tritium ( 3 H), indium ( 112 In), and technetium ( 99 mTc), fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • antibodies are not detectable, as such, from outside the body, and so must be labeled, or otherwise modified, to permit detection.
  • Markers for this purpose may be any that do not substantially interfere with the antibody binding, but which allow external detection.
  • Suitable markers may include those that may be detected by X- radiography, NMR or MRI.
  • suitable markers include any radioisotope that emits detectable radiation but that is not overtly harmful to the subject, such as barium or cesium, for example.
  • Suitable markers for NMR and MRI generally include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the antibody by suitable labeling of nutrients for the relevant hybridoma, for example.
  • the size of the subject, and the imaging system used, will determine the quantity of imaging moiety needed to produce diagnostic images.
  • the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of technetium-99.
  • the labeled antibody or antibody fragment will then preferentially accumulate at the location of cells which contain biomarker protein.
  • the labeled antibody or antibody fragment can then be detected using known techniques.
  • Antibodies that may be used to detect biomarker protein include any antibody, whether natural or synthetic, full length or a fragment thereof, monoclonal or polyclonal, that binds sufficiently strongly and specifically to the biomarker protein to be detected.
  • An antibody may have a IQ of at most about 10 "6 M, 10 "7 M, 10 “8 M, 10 "9 M, 10 "10 M, 10 "U M, 10 "12 M.
  • the phrase "specifically binds” refers to binding of, for example, an antibody to an epitope or antigen or antigenic determinant in such a manner that binding can be displaced or competed with a second preparation of identical or similar epitope, antigen or antigenic determinant.
  • An antibody may bind preferentially to the biomarker protein relative to other proteins, such as related proteins.
  • Antibodies are commercially available or may be prepared according to methods known in the art.
  • Antibodies and derivatives thereof that may be used encompass polyclonal or monoclonal antibodies, chimeric, human, humanized, primatized (CDR-grafted), veneered or single-chain antibodies as well as functional fragments, i.e., biomarker protein binding fragments, of antibodies.
  • antibody fragments capable of binding to a biomarker protein or portions thereof including, but not limited to, Fv, Fab, Fab' and F(ab') 2 fragments can be used.
  • Such fragments can be produced by enzymatic cleavage or by recombinant techniques. For example, papain or pepsin cleavage can generate Fab or F(ab') 2 fragments, respectively.
  • Fab or F(ab') 2 fragments can also be used to generate Fab or F(ab') 2 fragments.
  • Antibodies can also be produced in a variety of truncated forms using antibody genes in which one or more stop codons have been introduced upstream of the natural stop site.
  • a chimeric gene encoding a F(ab') 2 heavy chain portion can be designed to include DNA sequences encoding the CH, domain and hinge region of the heavy chain.
  • Boss et al European Patent No. 0,120,694 Bl; Neuberger, M. S. et al, WO 86/01533; Neuberger, M. S. et al, European Patent No. 0,194,276 Bl; Winter, U.S. Pat. No. 5,225,539; Winter, European Patent No. 0,239,400 Bl; Queen et al., European Patent No. 0451216 Bl; and Padlan, E. A. et al, EP 0519596 Al . See also, Newman, R. et al,
  • Antibodies produced from a library may also be used.
  • agents that specifically bind to a biomarker protein other than antibodies are used, such as peptides.
  • Peptides that specifically bind to a biomarker protein can be identified by any means known in the art. For example, specific peptide binders of a biomarker protein can be screened for using peptide phage display libraries.
  • the methods described herein can include determining the amount of serine 9 phosphorylated human GSK-3 or a corresponding phosphorylatable amino acid in a homolog of the human protein in the sample relative to said amount in the absence of the compound or at an earlier timepoint.
  • Phosphorylation is a biochemical reaction in which a phosphate group is added to Ser, Thr or Tyr residues of a protein and is catalyzed by protein kinase enzymes. Phosphorylation normally modifies the functions of target proteins, often causing activation. As part of the cell's homeostatic mechanisms, phosphorylation is only a transient process which is reversed by other enzyme called phosphatases. Therefore, protein phosphorylation levels change over time and can be evaluated in a number of well-known manners, including, for example, by immunological approaches.
  • the amount of serine 9 phosphorylated GSK-3 can be determined by an immunoassay using a reagent which specifically binds with serine 9 phosphorylated GSK-3P .
  • an immunoassay comprises a number of well-known forms, including, without limitation, a radioimmunoassay, a Western blot assay, an
  • immunofluoresence assay an enzyme immunoassay, an immunoprecipitation assay, a chemiluminescence assay, an immunohistochemical assay, a dot blot assay, or a slot blot assay.
  • PDA in situ proximity ligation assay
  • FPIA fluorescence polarization immunoassay
  • FFIA fluorescence immunoassay
  • EIA enzyme immunoassay
  • NIA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • the enzyme immunoassay is a sandwich enzyme immunoassay using a capture antibody or fragment thereof which specifically binds with GSK-3 regardless of phosphorylation status and a detection antibody or fragment thereof which specifically binds with serine 9 phosphorylated form of the protein.
  • Immunological reagents for identifying a protein, as well as phosphorylated forms of GSK-3 are well known in the art (e.g., Phospho-GSK-3 (ser9) antibody, Cell Signaling Technology®, catalogue number 9336 and the phospho-GSK-3 (ser9) antibody from abeam®, catalogue number ab75814).
  • Methods for generating antibodies that bind to phospho-forms of proteins are well known in the art.
  • Such antibody reagents e.g.
  • monoclonal antibody can be used to isolate and/or determine the amount of the respective proteins such as in a cellular lysate.
  • Such reagents can also be used to monitor protein levels in a cell or tissue, e.g., white blood cells or lymphocytes, as part of a clinical testing procedure, e.g., in order to monitor an optimal dosage of an inhibitory agent.
  • Detection can be facilitated by coupling (e.g., physically linking) the antibody to a detectable substance.
  • detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, ⁇ - galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine,
  • radioactive material examples include I, 13 1 1, 35 S or 3 H.
  • screening assays described herein can be adapted to identify candidate/test compounds which modulate (e.g., stimulate or inhibit) the expression, activity, or
  • phosphorylation status of a protein e.g., GSK-3P
  • a protein target of the kinase examples include phosphorylated target proteins in the signaling pathway of GSK-3 .
  • the disclosure provides assays for screening candidate/test compounds which interact with (e.g., bind to) GSK-3 protein.
  • Binding compound shall refer to a binding composition, such as a small molecule, an antibody, a peptide, a peptide or non-peptide ligand, a protein, an oligonucleotide, an oligonucleotide analog, such as a peptide nucleic acid, a lectin, or any other molecular entity that is capable of specifically binding to a target protein or molecule or stable complex formation with an analyte of interest, such as a complex of proteins.
  • Binding moiety means any molecule to which molecular tags can be directly or indirectly attached that is capable of specifically binding to an analyte. Binding moieties include, but are not limited to, antibodies, antibody binding compositions, peptides, proteins, nucleic acids and organic molecules having a molecular weight of up to about 1000 daltons and containing atoms selected from the group consisting of hydrogen, fluoride, carbon, oxygen, nitrogen, sulfur and phosphorus. Typically, the assays are cell-based assays. The cell, for example, can be of mammalian origin expressing GSK-3 .
  • the assays are cell-free assays which include the steps of combining a GSK-3P protein or a biologically active portion thereof, and a candidate/test compound, e.g., under conditions which allow for interaction of ⁇ e.g., binding of) the candidate/test compound to the GSK-3 protein or portion thereof to form a complex, and detecting the formation of a complex, in which the ability of the candidate compound to interact with ⁇ e.g., bind to) the GSK-3 polypeptide or fragment thereof is indicated by the presence of the candidate compound in the complex.
  • Formation of complexes between the GSK-3 protein and the candidate compound can be quantitated, for example, using standard immunoassays. Such analyses would identify test compounds as GSK-3 ligands.
  • GSK-3 or its target molecules to facilitate separation of complexes from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay.
  • Interaction ⁇ e.g., binding of) of GSK-3 to a target molecule, in the presence and absence of a candidate compound can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtitre plates, test tubes, and micro-centrifuge tubes.
  • a fusion polypeptide can be provided which adds a domain that allows the polypeptide to be bound to a matrix.
  • glutathione-S-transferase/GSK-3 fusion polypeptides can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St.
  • the complexes can be dissociated from the matrix, separated by SDS-PAGE, and the level of GSK-3 -binding polypeptide found in the bead fraction quantitated from the gel using standard electrophoretic techniques.
  • GSK-3 or its target molecule can be immobilized utilizing conjugation of biotin and streptavidin.
  • Biotinylated GSK-3 molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, 111.), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
  • antibodies reactive with GSK-3P but which do not interfere with binding of the polypeptide to its target molecule can be derivatized to the wells of the plate, and GSK-3 trapped in the wells by antibody conjugation.
  • preparations of a GSK-3 - binding polypeptide and a candidate compound are incubated in the GSK-3 -presenting wells of the plate, and the amount of complex trapped in the well can be quantitated.
  • Methods for detecting such complexes include immunodetection of complexes using antibodies reactive with the GSK- 3 ⁇ target molecule, or which are reactive with GSK-3 polypeptide and compete with the target molecule; as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the target molecule.
  • time course shall refer to the amount of time between an initial event and a subsequent event.
  • time course may relate to a subject's disease and may be measured by gauging significant events in the course of the disease, wherein the first event may be diagnosis and the subsequent event may be proliferation, metastasis, etc.
  • kinase assays to determine inhibition of phosphorylation effects can be performed according to well known methods in the art.
  • assays for determining GSK-3 kinase activity are well known in the art (see, for example, the publications described herein and incorporated by reference in their entirety).
  • Phosphorylation of a substrate of GSK-3 can be detected using a labelled phosphate group, such as the use of the radioactive label 32 P present as the ATP source in the buffer.
  • antibodies specific for the phosphorylated products of GSK-3 catalytic activity can be used to detect activity.
  • the assays are easily amenable to high through-put technologies using robotics and automated processes.
  • compositions described herein can be used in a variety of diagnostic, prognostic, and therapeutic applications,
  • One aspect of the present invention relates to screening assays, including non-cell based assays.
  • the invention relates to assays for screening test agents which bind to, or modulate the biological activity of, at least one biomarker listed in Tables 2-6.
  • a method for identifying such an agent entails determining the ability of the agent to modulate, e.g. inhibit, the at least one biomarker listed in Tables 2-6.
  • an assay is a cell-free or cell-based assay, comprising contacting at least one biomarker listed in Tables 2-6, with a test agent, and determining the ability of the test agent to modulate (e.g. inhibit) the enzymatic activity of the biomarker, such as by measuring direct binding of substrates or by measuring indirect parameters as described below.
  • biomarker protein in a direct binding assay, can be coupled with a radioisotope or enzymatic label such that binding can be determined by detecting the labeled protein or molecule in a complex.
  • the targets can be labeled with 125 1, 35 S, 14 C, or 3 H, either directly or indirectly, and the radioisotope detected by direct counting of radioemmission or by scintillation counting.
  • the targets can be enzymatically labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product.
  • Determining the interaction between biomarker and substrate can also be accomplished using standard binding or enzymatic analysis assays.
  • Binding of a test agent to a target can be accomplished in any vessel suitable for containing the reactants.
  • vessels include microtiter plates, test tubes, and micro-centrifuge tubes.
  • Immobilized forms of the antibodies of the present invention can also include antibodies bound to a solid phase like a porous, microporous (with an average pore diameter less than about one micron) or macroporous (with an average pore diameter of more than about 10 microns) material, such as a membrane, cellulose,
  • nitrocellulose, or glass fibers such as that made of agarose or polyacrylamide or latex; or a surface of a dish, plate, or well, such as one made of polystyrene.
  • determining the ability of the agent to modulate the interaction between the biomarker and a substrate or a biomarker metabolite and its natural binding partner can be accomplished by determining the ability of the test agent to modulate the activity of a polypeptide or other product that functions downstream or upstream of its position within the pathway (e.g., feedback loops).
  • the present invention further pertains to novel agents identified by the above- described screening assays. Accordingly, it is within the scope of this invention to further use an agent identified as described herein in an appropriate animal model.
  • an agent identified as described herein can be used in an animal model to determine the efficacy, toxicity, or side effects of treatment with such an agent.
  • an antibody identified as described herein can be used in an animal model to determine the mechanism of action of such an agent.
  • the present invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, and monitoring clinical trials are used for prognostic (predictive) purposes to thereby treat an individual prophylactically.
  • diagnostic assays for determining the amount and/or activity level of a biomarker listed in Tables 2-6 in the context of a biological sample (e.g., blood, serum, cells, or tissue) .
  • Such assays can be used for prognostic or predictive purpose to thereby prophylactically treat an individual prior to the onset or after recurrence of a disorder characterized by or associated with biomarker polypeptide, nucleic acid expression or activity.
  • any method can use one or more (e.g., combinations) of biomarkers listed in Tables 2-6.
  • Another aspect of the present invention pertains to monitoring the influence of agents (e.g. , drugs, compounds, and small nucleic acid-based molecules) on the expression or activity of a biomarker listed in Tables 2-6.
  • agents e.g. , drugs, compounds, and small nucleic acid-based molecules
  • a computer system can also store and manipulate data generated by the methods of the present invention which comprises a plurality of biomarker signal changes/profiles which can be used by a computer system in implementing the methods of this invention.
  • a computer system receives biomarker expression data; (ii) stores the data; and (iii) compares the data in any number of ways described herein (e.g. , analysis relative to appropriate controls) to determine the state of informative biomarkers from a sample.
  • a computer system (i) compares the determined expression biomarker level to a threshold value; and (ii) outputs an indication of whether said biomarker level is significantly modulated (e.g., above or below) the threshold value, or a phenotype based on said indication.
  • such computer systems are also considered part of the present invention.
  • Numerous types of computer systems can be used to implement the analytic methods of this invention according to knowledge possessed by a skilled artisan in the bioinformatics and/or computer arts.
  • Several software components can be loaded into memory during operation of such a computer system.
  • the software components can comprise both software components that are standard in the art and components that are special to the present invention (e.g., dCHIP software described in Lin et al. (2004) Bioinformatics 20, 1233-1240; radial basis machine learning algorithms (RBM) known in the art).
  • dCHIP software described in Lin et al. (2004) Bioinformatics 20, 1233-1240
  • RBM radial basis machine learning algorithms
  • the methods of the invention can also be programmed or modeled in mathematical software packages that allow symbolic entry of equations and high-level specification of processing, including specific algorithms to be used, thereby freeing a user of the need to procedurally program individual equations and algorithms.
  • Such packages include, e.g.,
  • the computer comprises a database for storage of biomarker data.
  • biomarker data can be accessed and used to perform comparisons of interest at a later point in time.
  • biomarker expression profiles of a sample derived from the blood or tissue of a subject and/or profiles generated from population-based distributions of informative loci of interest in relevant populations of the same species can be stored and later compared to that of a sample derived from the blood or tissue of the subject .
  • other, alternative program structures and computer systems will be readily apparent to the skilled artisan. Such alternative systems, which do not depart from the above described computer system and programs structures either in spirit or in scope, are therefore intended to be comprehended within the accompanying claims.
  • the present invention provides, in part, methods, systems, and code for accurately classifying whether a biological sample is associated with a condition that is likely to respond to a therapy.
  • the present invention is useful for classifying a sample (e.g., from a subject) as associated with or at risk for responding to or not responding to a therapy using a statistical algorithm and/or empirical data (e.g., the amount or activity of a biomarker listed in Table s 2-6).
  • An exemplary method for detecting the amount or activity of a biomarker listed in Table s 2-6, and thus useful for classifying whether a sample is likely or unlikely to respond to a therapy involves obtaining a biological sample from a test subject and contacting the biological sample with an agent.
  • the statistical algorithm is a single learning statistical classifier system.
  • a single learning statistical classifier system can be used to classify a sample as a based upon a prediction or probability value and the presence or level of the biomarker.
  • the use of a single learning statistical classifier system typically classifies the sample as, for example, a likely anti-immune checkpoint inhibitor therapy responder or progressor sample with a sensitivity, specificity, positive predictive value, negative predictive value, and/or overall accuracy of at least about 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
  • learning statistical classifier systems include a machine learning algorithmic technique capable of adapting to complex data sets (e.g., panel of markers of interest) and making decisions based upon such data sets.
  • a single learning statistical classifier system such as a classification tree (e.g., random forest) is used.
  • a combination of 2, 3, 4, 5, 6, 7, 8, 9, 10, or more learning statistical classifier systems are used, preferably in tandem.
  • learning statistical classifier systems include, but are not limited to, those using inductive learning (e.g., decision/classification trees such as random forests, classification and regression trees (C&RT), boosted trees, etc.), Probably Approximately Correct (PAC) learning, connectionist learning (e.g.
  • inductive learning e.g., decision/classification trees such as random forests, classification and regression trees (C&RT), boosted trees, etc.
  • PAC Probably Approximately Correct
  • neural networks NN
  • artificial neural networks ANN
  • neuro fuzzy networks NNN
  • network structures perceptrons such as multi-layer perceptrons, multi-layer feed-forward networks, applications of neural networks, Bayesian learning in belief networks, etc.
  • reinforcement learning e.g., passive learning in a known environment such as naive learning, adaptive dynamic learning, and temporal difference learning, passive learning in an unknown environment, active learning in an unknown environment, learning action-value functions, applications of reinforcement learning, etc.
  • genetic algorithms and evolutionary programming e.g., passive learning in a known environment such as naive learning, adaptive dynamic learning, and temporal difference learning, passive learning in an unknown environment, active learning in an unknown environment, learning action-value functions, applications of reinforcement learning, etc.
  • the method of the present invention further comprises sending the sample classification results to a clinician, e.g., an oncologist.
  • a clinician e.g., an oncologist.
  • diagnosis of a subject is followed by administering to the individual a therapeutically effective amount of a defined treatment based upon the diagnosis.
  • the methods further involve obtaining a control biological sample (e.g., biological sample from a subject who does not have a condition. d. Prognostic Assays
  • the diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk that are likely or unlikely to be responsive to a therapy.
  • the assays described herein such as the preceding diagnostic assays or the following assays, can be utilized to identify a subject having or at risk of developing a disorder associated with a misregulation of the amount or activity of at least one biomarker described in Tables 2-6.
  • the prognostic assays can be utilized to identify a subject having or at risk for developing a disorder associated with a misregulation of the at least one biomarker described in Table 2-6.
  • the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, polypeptide, peptide, nucleic acid, small molecule, or other drug candidate) to treat a disease or disorder associated with the aberrant biomarker expression or activity.
  • an agent e.g., an agonist, antagonist, peptidomimetic, polypeptide, peptide, nucleic acid, small molecule, or other drug candidate
  • the compositions described herein (including dual binding antibodies and derivatives and conjugates thereof) can be used in a variety of in vitro and in vivo therapeutic applications using the formulations and/or combinations described herein. VII. Pharmaceutical Compositions
  • the present invention provides pharmaceutically acceptable compositions which comprise a therapeutically-effective amount of an agent that modulates biomarker expression and/or activity ⁇ e.g., GSK-3P activity), one or more anti-immune checkpoint inhibitors, or a combination thereof, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents.
  • an agent that modulates biomarker expression and/or activity e.g., GSK-3P activity
  • one or more anti-immune checkpoint inhibitors e.g., a combination thereof
  • compositions of the present invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, boluses, powders, granules, pastes; (2) parenteral administration, for example, by subcutaneous, intramuscular or intravenous injection as, for example, a sterile solution or suspension; (3) topical application, for example, as a cream, ointment or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; or (5) aerosol, for example, as an aqueous aerosol, liposomal preparation or solid particles containing the compound.
  • oral administration for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, boluses, powders, granules, pastes
  • parenteral administration for example, by subcutaneous, intramuscular or intravenous injection
  • therapeutically-effective amount means that amount of an agent that modulates biomarker expression and/or activity, or expression and/or activity of the complex, or composition comprising an agent that modulates biomarker expression and/or activity, or expression and/or activity of the complex, which is effective for producing some desired therapeutic effect at a reasonable benefit/risk ratio.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those agents, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication,
  • phrases "pharmaceutically-acceptable carrier” as used herein means a
  • composition or vehicle such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject chemical from one organ, or portion of the body, to another organ, or portion of the body.
  • a liquid or solid filler such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject chemical from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject.
  • materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydrox
  • salts refers to the relatively non-toxic, inorganic and organic acid addition salts of the agents that modulates biomarker expression and/or activity, or expression and/or activity of the complex encompassed by the invention. These salts can be prepared in situ during the final isolation and purification of the agents, or by separately reacting a purified agent in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed.
  • Representative salts include the
  • the agents useful in the methods of the present invention may contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically- acceptable salts with pharmaceutically-acceptable bases.
  • pharmaceutically- acceptable salts refers to the relatively non-toxic, inorganic and organic base addition salts of agents that modulates biomarker expression and/or activity, or expression and/or activity of the complex.
  • salts can likewise be prepared in situ during the final isolation and purification of the agents, or by separately reacting the purified agent in its free acid form with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically-acceptable metal cation, with ammonia, or with a pharmaceutically- acceptable organic primary, secondary or tertiary amine.
  • a suitable base such as the hydroxide, carbonate or bicarbonate of a pharmaceutically-acceptable metal cation, with ammonia, or with a pharmaceutically- acceptable organic primary, secondary or tertiary amine.
  • Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like.
  • Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like (see, for example, Berge et ah, supra).
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), le
  • Formulations useful in the methods of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal, aerosol and/or parenteral administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration.
  • the amount of active ingredient, which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, most preferably from about 10 per cent to about 30 per cent.
  • Methods of preparing these formulations or compositions include the step of bringing into association an agent that modulates biomarker expression and/or activity, with the carrier and, optionally, one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing into association an agent with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • Formulations suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of an agent as an active ingredient.
  • a compound may also be administered as a bolus, electuary or paste.
  • solid dosage forms for oral administration capsules, tablets, pills, dragees, powders, granules and the like
  • the active ingredient is mixed with one or more
  • pharmaceutically-acceptable carriers such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, acetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a
  • compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface- active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered peptide or peptidomimetic moistened with an inert liquid diluent.
  • Tablets, and other solid dosage forms may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions, which can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • opacifying agents include polymeric substances and waxes.
  • the active ingredient can also be in microencapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1 ,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emuls
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • Suspensions in addition to the active agent may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • Formulations for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more agents with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
  • suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
  • Formulations which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration of an agent that modulates (e.g., inhibits) biomarker expression and/or activity include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active component may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
  • the ointments, pastes, creams and gels may contain, in addition to an agent, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to an agent that modulates (e.g. , inhibits) biomarker expression and/or activity, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • the agent that modulates (e.g., inhibits) biomarker expression and/or activity can be alternatively administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation or solid particles containing the compound. A nonaqueous (e.g., fluorocarbon propellant) suspension could be used. Sonic nebulizers are preferred because they minimize exposing the agent to shear, which can result in degradation of the compound.
  • an aqueous aerosol is made by formulating an aqueous solution or suspension of the agent together with conventional pharmaceutically acceptable carriers and stabilizers.
  • the carriers and stabilizers vary with the requirements of the particular compound, but typically include nonionic surfactants (Tweens, Pluronics, or polyethylene glycol), innocuous proteins like serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars or sugar alcohols.
  • Aerosols generally are prepared from isotonic solutions.
  • Transdermal patches have the added advantage of providing controlled delivery of an agent to the body.
  • dosage forms can be made by dissolving or dispersing the agent in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the agent
  • the rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the peptidomimetic in a polymer matrix or gel.
  • Ophthalmic formulations are also contemplated as being within the scope of this invention.
  • compositions of this invention suitable for parenteral administration comprise one or more agents in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
  • adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
  • Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride
  • a liquid suspension of crystalline or amorphous material having poor water solubility The rate of absorption of the drug then depends upon its rate of dissolution, which, in turn, may depend upon crystal size and crystalline form.
  • delayed absorption of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
  • Injectable depot forms are made by forming microencapsule matrices of an agent that modulates biomarker expression and/or activity, in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions, which are compatible with body tissue.
  • the agents of the present invention When the agents of the present invention are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be determined by the methods of the present invention so as to obtain an amount of the active ingredient, which is effective to achieve the desired therapeutic response for a particular subject, composition, and mode of administration, without being toxic to the subject.
  • kits for detecting and/or modulating biomarkers described herein may also include instructional materials disclosing or describing the use of the kit or an antibody of the disclosed invention in a method of the disclosed invention as provided herein.
  • a kit may also include additional components to facilitate the particular application for which the kit is designed.
  • a kit may additionally contain means of detecting the label (e.g. , enzyme substrates for enzymatic labels, filter sets to detect fluorescent labels, appropriate secondary labels such as a sheep anti-mouse-HRP, etc.) and reagents necessary for controls (e.g., control biological samples or metabolite standards).
  • a kit may additionally include buffers and other reagents recognized for use in a method of the disclosed invention. Non-limiting examples include agents to reduce non-specific binding, such as a carrier protein or a detergent.
  • Example 1 Materials and Methods for Examples 2-8
  • a canine pacing model was used as described by Chakir et al. (Chakir, K., et al.
  • HFd ys and HF sync were atrially tachypaced for the entire 6 weeks, while CRT animals were subjected first to three weeks of atrial tachypacing, followed by three weeks of biventricular
  • V3A3 model was RV paced for the first 3 weeks, and then received 3 weeks of atrial tachypacing, while the AVA model was atrially paced atrially for the entire period excepting a two week period in the middle when it received right ventricular pacing.
  • a subset of dogs were placed under anesthesia and a 7F bioptome (Cordis Corporation) was used to collect 5-7 biopsies from the endocardium near the right ventricular apex, under fluoroscopic guidance.
  • Table 1 shows echocardiography (at 3 and 6 weeks) and hemodynamic (at sacrifice) indices in the dog models.
  • DI dyssynchrony index
  • LVEDV left ventricular end-diastolic volume
  • LVESV left ventricular end-systolic volume
  • SV stroke volume
  • LVEF left ventricular ejection fraction
  • HR heart rate
  • LVPes left ventricular end-systolic pressure
  • LVPed left ventricular end-diastolic pressure
  • SBP systolic blood pressure
  • DBP diastolic blood pressure
  • MAP mean arterial pressure
  • dP/dt max maximum rate of change of the pressure waveform
  • IP isovolumic pressure. Italicized values indicates P ⁇ 0.05 vs. Control/Baseline value. *, P ⁇ 0.05 vs. HFdys, ⁇ , P ⁇ 0.05 vs. the 3 week value in
  • Tissue from the endocardium layer of the left ventricular lateral wall was flash frozen in liquid nitrogen and stored at -80 °C. Subsequently, the tissue was homogenized in isolation solution, in the presence of 0.3% Triton X-100, and protease and phosphatase inhibitors. Isolation solution contained (in mM): 5.5 Na 2 ATP, 7.11 MgCl 2 , 2 EGTA, 108.01 KC1, 8.91 KOH, 10 imidazole, 10 DTT. Myocytes were then washed in the absence of Triton X-100 to remove the detergent.
  • Myocytes were glued, using silicone, to the tips of 150 ⁇ diameter minutia pins attached to a force transducer and motor arm (Aurora Scientific Inc., Aurora, ON, Canada). Sarcomere length was monitored by video camera (Imperx, Boca Raton, FL) and calculated by the High-speed Video Sarcomere Length Program (Aurora Scientific Inc.). Myocytes were maintained at a sarcomere length of 2.1 ⁇ . Myocytes were kept in relaxing buffer containing (in mM): 5.95 Na 2 ATP, 6.41 MgCl 2 , 10 EGTA, 100 BES, 10
  • GSK-3P glycogen synthase kinase-3P
  • PKA PKA (0.125 units/mL, 20 minutes, Sigma- Aldrich
  • Akt Akt
  • Tissue samples were minced into 2-4 mm pieces then mechanically homogenized three times at low speed (1,000 rpm, 3 sec), and re-suspended in relaxing solution containing Triton X-100 overnight at 4 °C. All solutions contained protease inhibitors (Sigma-Aldrich).
  • Multicellular skinned fragments (0.1-0.25 mm wide; 1-2.5 mm long) were attached to aluminum T-clips and mounted to arms extending from a force transducer (World Precision Instruments) and a high-speed length controller (Aurora Scientific Inc). Sarcomere length was set to 2.3 um by laser diffraction. The isolated muscle was then exposed to a range of calcium solutions while generated tension and consumed ATP were measured simultaneously during the contraction. ATP consumption was measured as previously described using a UV coupled optical absorbance method 3 . Following each contraction, calibration steps were performed by step-wise injection of 250 pmol of ADP. Activating solution contained (in mM): 20
  • Trabeculae were isolated from the right ventricular free wall and skinned overnight in the presence of 1% Triton X-100 (to remove all membranous structures), and protease (Sigma- Aldrich, St. Louis, MO) and phosphatase inhibitors (Roche Diagnostics, Indianapolis, IN) at 4 °C. Trabeculae were attached via a "basket and hook” technique 4 , to a force transducer (SI Heidelberg) and stationary hook. The muscle was stretched to produce 5 mN » mm ⁇ 2 of passive tension (corresponding to maximum twitch force). Force was measured as bath solution Ca 2+ was increased from 0 to saturating levels (46.8 ⁇ ), obtained by proportional mixing of activating and relaxing solutions.
  • Relaxing solution contained (in mM): 10 K 2 H 2 EGTA, 5.45 MgCl 2 , 5.5 Na 2 ATP, 15 Na 2 CrP, 80 KCl, 25 HEPES, 5 DTT, protease and phosphatase inhibitors.
  • Activating solution contained (in mM): 10 Ca 2+ EGTA, 5.15 MgCl 2 , 5.5 Na 2 ATP, 15 Na 2 CrP, 80 KCl, 25 HEPES, 5 DTT, protease and phosphatase inhibitors. All buffers were adjusted to a pH of 7.2. Trabeculae were completely activated at the beginning and end of each experiment, and if there was greater than 10% rundown, the experiment was discarded.
  • a second force-Ca 2+ relation was determined after exposure to protein phosphatase 1 (PP1, 25,000 mU/mL for 60 minutes, New England Biolabs, Ipswich, MA).
  • tissue was homogenized, using a glass dounce homogenizer, in ice-cold standard rigor buffer (SRB) and 1% Triton X-100.
  • SRB contained (in mM): 75 KCl, 10 imidazole, 2 MgCl 2 , 2 EGTA, 1 NaN 3 , plus protease and phosphatase inhibitors. The sample was then centrifuged at 16,000 g for 1 minute at 4 °C.
  • Tissue samples were prepared as above, biopsies were prepared using a 0.1 mL dounce micro-homogenizer (Radnoti LLC, Monrovia, CA), in 40 cell lysis buffer (Cell Signaling, Danvers, MA), and further solubilized by adding 1 % SDS. Samples were ran on 4-12% precast Bis-Tris gels (Invitrogen), transferred to nitrocellulose membranes, and blotted using custom MyBPC antibodies against phospho Ser-273, Ser-282, Ser-302 (Sadayappan et al. (2009) Circulation.
  • Myofilament-enriched HF dys and CRT samples were labeled with cyanine (Cy) 3 or Cy5, and a mixture of the two (as an internal control) labeled with Cy2 (0.4 iL dye/ 50 ⁇ g protein). All three samples were then isoelectrically focused on an IEF strip (GE Healthcare Biosciences, Piscataway, NJ). The IEF instrument was run with the following steps: 10 hours active rehydration at 50V, 1 hour rapid ramp to 250V, 1 hour rapid ramp to 500V, 1 hour rapid ramp to 1000V, 4 hours normal ramp to 10,000V, 50,000 V'hours at 10,000V.
  • the strips were alkylated and reduced, and then separated by weight on a large-format (18 cm) 10% Bis-Tris resolving gel in MES buffer. After fixing and washing the gel, it was scanned on a Typhoon scanner (GE Healthcare). Image analysis was performed by REDFIN (Ludesi, Malmo, Sweden).
  • Labeled relax buffer was prepared by combining relax buffer with.
  • the labeled relax buffer contained a 1 : 1 ratio of unlabeled and 18 0 labeled ATP.
  • the samples were incubated at 30 °C for 30 minutes.
  • myo filament-enriched HFd ys and CRT samples (1 mg protein in 8M urea, 1% SDS) were reduced, alkylated, then digested overnight in solution using trypsin (Promega, Madison, WI) (100 ⁇ g/1.0 mg protein). The reaction was stopped with 10% TFA, and the samples were desalted with Oasis HLB cartridges (Waters, Milford, MA). Strong cation exchange (SCX) columns were prepared using SCX bulk media (Nest Group, Southborough, MA). The sample was split into two parts: 60% was run through the SCX columns (1/3 collected as flow through, 1/3 eluted with SCX buffer plus 40 mM KC1, and 1/3 eluted with SCX buffer plus 150 mM KC1). These three fractions, along with the 40% not run through the SCX column, were then individually desalted.
  • trypsin Promega, Madison, WI
  • SCX Strong cation exchange
  • Ti0 2 titanium oxide beads
  • each fraction 4 per sample, 6 samples per group
  • the Ti0 2 beads were then washed, and the sample eluted in 10%> ACN/3%> NH 4 OH.
  • Samples were then dried down and resuspended in 70% ACN/1 % FA for LC-MS/MS analysis.
  • Phospho-peptides were analyzed on an Agilent 1200 nano-LC system (Agilent, Santa Clara, CA) connected to an LTQ-Orbitrap mass spectrometer (Thermo Scientific, Waltham, MA), equipped with a nanoelectrospray ion source.
  • the peptides were separated on a C 18 RP- HPLC column (75 ⁇ x 10 cm self-packed with 5 urn, 200 A Magic CI 8; Michrom Bio Resources, Auburn, CA) at a flow rate of 300 nL/min. Each fraction (4 per sample) was run separately to maximize the number of peptides acquired.
  • Each MSI scan was followed by col lision induced dissociation of the 5 most abundant precursor ions.
  • Mono-isotopic precursor selection was enabled and dynamic exclusion was enabled with a repeat count of 2, repeat duration of 30 seconds and exclusion duration of 60 seconds. Only MSI signals exceeding 1000 counts triggered the MS2 scans and +1 and unassigned charge states were not selected for MS2 analysis.
  • a second set of samples was prepared for the GSK-3P treatment.
  • Myofilament enriched samples isolated myofibrils
  • the sample from each dog was split into two equal parts, one incubated in labeled relax buffer, and the second incubated in labeled relax buffer plus 4 ⁇ g of GSK-3p.
  • Labeled relax buffer was prepared by combining relax buffer with ATP ⁇ - ⁇ 18 0 (Cambridge Isotope Laboratories, Inc., Andover, MA).
  • the labeled relax buffer contained a 1 : 1 ratio of unlabeled and 18 0 labeled ATP (20 ⁇ of each, per sample). The samples were incubated at 30 °C for 30 minutes. Post treatment, samples were prepared, run on the mass spectrometer, and analyzed in the same manner as above, with one exception: searching also included an additional variable PTM: labeled phosphorylation that includes an additional 6.01 Da shift due to the 18 0-labeled phosphate group.
  • CRT involved direct ventricular myocardial stimulation that might have triggered improved myofibrillar function.
  • CRT cardiac free-wall pacing
  • F max normalized to myocyte cross sectional area, CSA
  • CSA myofibrillar proteins
  • troponin I 27
  • isoform switching e.g. troponin T
  • Example 5 Ca 2+ -sensitization is mediated by myofilament phosphorylation
  • Figure 8A displays example force-Ca 2+ data from RV trabeculae prior to and following incubation with the phosphatase PP1. Dephosphorylation did not change F max in any group
  • Ca 2+ sensitivity is regulated by phosphorylation of the regulatory thin filament proteins troponin I (Tnl), troponin T (TnT), myosin binding protein C (MyBPC), and myosin light chain 2 (MLC2).
  • Tnl troponin I
  • TnT troponin T
  • MyBPC myosin binding protein C
  • MLC2 myosin light chain 2
  • Table 2 shows myofilament phosphorylation sites that are known to modulate calcium sensitivity.
  • the table provides the direction of a change in calcium sensitivity due to phosphorylation, the direction that phosphorylation is modified by heart failure, and how this in turn impacts calcium sensitivity.
  • TnT phosphorylation at T206 (38) PLC
  • S278/ T287 (39) Rho-kinase II
  • Table 3 shows a selection of phosphorylation sites identified by mass spectrometry, which were not significantly different between HFd ys and CRT. As stated in the online methods, the protein probability threshold was set at 95%; however, MLC2, included in this table, had a 93% protein probability. Loc. Prob., localization probability as calculated by Scaffold PTM (vl .1.3, Proteome Software) using the A-score algorithm.
  • Example 6 Z-Disk and M-Band Proteins are phosphorylated by CRT; Role of GSKSfi Given that the known phospho-protein targets did not appear to be involved, mass spectrometry was used to broadly interrogate the myofibrillar sub-proteome. A total of 1187 phosphorylation sites on 166 myofilament and myo filament-associated proteins were detected, and CRT significantly altered 33 of these sites. Based on the PPl results, residues exhibiting increased phosphorylation with CRT were assessed, yielding 15 sites on 13 proteins (Table 4 and Figures 12-26). Figure 27 summarizes this analysis.
  • Table 4 shows myofilament phosphorylation sites increased by CRT. The peptide sequence is shown, where the phosphorylated site is indicated in lowercase. Possible kinases were determined using the Human Protein Reference Database and NetphosK. Loc Prob,
  • GSK-3P we performed a second mass spectrometry experiment.
  • the myofilament subproteome obtained from HFd ys dogs was incubated with 18 0 labeled ATP in the absence or presence of recombinant active GSK-3P for 30 minutes at 30°C and prepared for proteomic analysis.
  • Table 5 lists the phosphorylated amino acid residues targeted by GSK-3P that were also found to increase with CRT.
  • Four of the seven GSK-3P sites revealed by in silico analysis were confirmed, and four additional sites altered by CRT but not predicted by bio- informatics were revealed as GSK-3P targets as well. This supports the notion that GSK-3P converts a subset of the HFd ys phospho-proteome into one similar to CRT.
  • Akt is a recognized upstream kinase modulator of GSK-3P
  • myocytes incubated with Akt exhibited no change in F max or EC 50 in either group ( Figures 29A-29B), supporting an alternative pathway.
  • Akt treatment did not detectably increase phosphorylation of any proteins in the myofilament fraction (Figure 29C), it did in whole cell lysate (serine 9 on GSK-3P, Figures 29D-29E).
  • GSK-3P has several myofilament targets that have been determined to be detectable in the blood, such as plasma (Table 6), when the myocardium is damaged. Phosphorylation of these sites is increased by CRT, in concordance with re-activation of GSK-3p. These phosphorylation sites thus represent biomarkers, correlating with myocardial GSK-3P activity and cardiac dyssynchrony. Table 6
  • Table 6 shows that GSK-3P myofilament phosphorylation targets are detectable in plasma.
  • Table 6 The results in Table 6 were generated from proteins identified in plasma samples from the PaxDB website. Specifically, the dataset where generated from data uploaded to pride and the integrated dataset is as follows: 1. Human plasma, Lee, PRIDE,ID:8653-8672', 2 .Human plasma, non-alkylated, Qian,MCP,2005' (weighting 100%), and 3. Human plasma,
  • GSK-3P In order to determine the extent that GSK-3P is involved in dyssynchrony mechano- transduction, it is necessary to use genetic modifications to block its inactivation or remove it entirely. Unfortunately, genetic tools in the dog are mostly limited to gene therapy, and even this turns out to be extremely difficult in this species. Thus, a mouse model was developed where the desired GSK-3P genetically engineered lines exist. For example, an inducible GSK-3P knock-out mouse line and a constitutive ly active GSK-3P model are available.
  • the developed pacemaker system utilizes a custom designed micro-pacing unit ( Figure 31 A), bi-polar implantable lead ( Figure 3 IB), a connector at the back of the mouse's neck, a micro-commutator built into the top of the cage (allowing complete rotational freedom), and a power source attached to the outside of each cage. Cardiac dyssynchrony can be induced by single site pacing of the right ventricle (RV).
  • RV right ventricle
  • mice have been successfully paced dyssynchronously for 2 weeks and longer, which has been confirmed by ECG (Figure 31C) and echo (Figure 3 ID).
  • ECG leads can be implanted and passed through the commutator for easy and frequent confirmation of pacing capture.
  • RV pacing reduced systolic function (%FS) over the course of the pacing protocol ( Figure 3 IE).
  • SWATH improves throughput and sensitivity .
  • SWATH is a new MS approach that allows the quantification of tens of thousands of peptides generated from the tryptic digestion of the proteome and phospho-proteome in every single sample.
  • the platform is stable, robust, fast, reproducible and sensitive. It is particularly useful in situations where post-translational modifications must be tracked over various models and treatments as is proposed below.
  • Example 10 Determining whether GSK-3fi inactivation is necessary and sufficient for dyssynchrony-induced myofilament calcium desensitization
  • GSK-3P is inactivated by cardiac dyssynchrony and this depresses calcium sensitivity.
  • Paced mouse recapitulate the phenotype observed in the dog described in Examples 1- 8.
  • Cardiac dyssynchrony via RV pacing (rate just above sinus at 700 bpm x 10 days) exhibits calcium desensitization compared to a lead implant without pacing ( Figure 32A).
  • a GSK-3P myocyte targeted knock-out model (combines GSK-3P flox'd mouse with MerCreMer-tamoxifen inducible Cre coupled to aMHC promoter) is obtained. Data are assessed after 4 weeks of tamoxifen treatment to be free from any acute myocardial depression that can occur with this gene-targeting system. As shown in Figure 32C, myocytes from these hearts in which GSK-3P is suppressed show a right shift of the force-Ca 2+ dependence, consistent with a desensitization of the myofilament to Ca 2+ . This supports the hypothesis based on the canine results described above where a loss of function analysis was not feasible.
  • an embodiment of the pacemaker system consists of an implantable lead, the externalized connector, a micro-commutator (electrical swivel), a micro pacing unit, and power source.
  • the bi-polar lead consists of two metal rings (positive, ground), 1.5 mm in diameter, attached (but isolated from each other) using a biocompatible epoxy. Each ring is attached to 5 cm of 36 AWG wire. The two wires terminate in a 4-pin 0.050" pitch male connector.
  • the connector attaches to a female connector at the bottom of the flying lead micro-commutator, which is the smallest and lowest friction commutator available (see Dragonfly Research and Design, Inc.).
  • the micro-commutator is attached to the lid of the mouse cage.
  • the commutator connects to the custom pacing unit.
  • the units are 9.5 mm in diameter, have a depth of 3.2 mm, and are coated in medical grade silicone.
  • the chip is connected to a battery holder with 3 AA batteries (4.5 volts). Both the pacing chip and the batteries attach to the mouse cage lid.
  • a mouse is anesthetized with etomidate, intubated, and ventilated. Anesthesia is maintained with inhaled isoflurane. A midline incision exposes the chest wall and abdomen. The right ventricular free wall is exposed by right anterolateral thoracotomy. The pacing lead is placed against the RV free wall and attached using 7-0 prolene suture. The lead is connected to an external stimulator, and ECG used to determine pacing voltage threshold. The threshold typically increases 1.5- to 2-fold over the first week, at which point it is relatively stable. Thus, the lead is repositioned if the threshold is over 2 V (the battery generates 4.5 V).
  • the lead connector is tunneled to the mouse's back, and externalized on the back of the neck so the mouse cannot chew through it.
  • a purse string knot keeps the wound closed around the connector, and a small amount of superglue keeps the connector in place.
  • the chest is closed using 6-0 Prolene and negative pressure in the thorax is restored by removal of air by a chest tube attached to a syringe. The surgery lasts ⁇ 30 minutes and is associated with less than 25% mortality.
  • the pacemaker is turned on after a one-week recovery period. During pacing, the animal cages are returned to the animal facility, which reduces stress and avoids weight loss problems during the protocol.
  • the pacing protocol can be applied to four groups.
  • GSK-3P knock-out mice (GSK-SP ⁇ /Cre) treated with tamoxifen for three weeks, are used.
  • mice will be given tamoxifen (20 mg/kg IP), dissolved in 30% ethanol in sterile PBS at a final concentration of 4 mg/rnL.
  • tamoxifen (20 mg/kg IP)
  • tamoxifen dissolved in 30% ethanol in sterile PBS at a final concentration of 4 mg/rnL.
  • GSK-3p fl/fl /Cre mice treated with tamoxifen saw a 77% average reduction in GSK-3P expression (Woulfe, K.C., et al., (2010) Circ Res 106(10): 1635-45); the incomplete knock-out is due in part to its persistence in non-myocytes.
  • controls for group 1 are mice with inducible Cre lacking the flox'd gene and GSK-3p fl/fl /-, both treated with tamoxifen for the same duration as group 1. For both groups, tamoxifen is administered before the pacemaker implant surgery.
  • a constitutively active knock-in model of GSK-3P with an S9A substitution, GSK-3p S9A , are used. This mouse shows some minor developmental differences from wild-type, but a normal cardiac phenotype.
  • controls for group 3 are the wild-type littermate controls of the GSK-3p S9A mice (GSK-3p WT ).
  • Intact heart function is assessed in vivo by trans-thoracic echocardiography. Mice are sedated (isoflurane) to facilitate pacing manipulation while images are obtained
  • Skinned myocytes are prepared by homogenizing LV myocardium in isolation solution plus protease and phosphatase inhibitors and Triton X-100. Triton is a detergent that removes the membrane and leaves the myofilament.
  • the myocyte is glued to two minutia pins using silicone.
  • the myocyte is moved to relaxing solution (0 Ca 2+ ), and sarcomere length is set to 2.1 ⁇ (detected by a high-speed camera).
  • the myocyte is then exposed to sub- max Ca 2+ concentrations achieved by mixing Relaxing and Activating solutions .
  • Myocyte width and depth are recorded, to normalize data to cross-sectional area.
  • GSK-3P activity can also be assessed.
  • Tissue samples are treated to obtain a myofilament-enriched sample as described in Arrell et al. in the presence of protease and phosphatase inhibitors (Arrell, D.K., et al., (2001) Circ Res. 89(6):480-7).
  • protease and phosphatase inhibitors Arrell, D.K., et al., (2001) Circ Res. 89(6):480-7.
  • "everything else” cytosolic, mitochondria, membrane, etc.
  • Antibodies against total GSK-3P (mouse) and p-S9 (rabbit) can be used simultaneously, using Licor IR secondary antibodies.
  • the membrane is imaged on an Odyssey system (Licor), allowing simultaneous quantitation of phosphor- and total GSK-3P, the ratio of which represents GSK-3P activity.
  • the baseline myofilament function and GSK-3P activity is determined in one or more of the four groups. In knock-out mice, this can be done before and after exogenous GSK-3P treatment (as described in Kirk, J.A., et al., (2014) Journal of
  • pacing While pacing, ECG readings are obtained every other day. Pacing can be confirmed by QRS morphology and heart rate equal to pacing frequency (700). If capture is lost, the mouse is excluded from the study. If capture is intermittent, the commutator is connected to an external stimulator, offering output voltages above 4.5 V. The mice are echoed three times (day 3, 7, and 14). The protocol lasts 14 days, but can be extended if needed. The heart is removed, washed in ice-cold PBS, and dissected to obtain the relevant regions. Each heart is optionally snap frozen in liquid N 2 and stored at -80 °C until needed for GSK-3P activity assay, skinned myocytes, or proteomics described further herein. An embodiment of the groups and protocols is shown in Figure 33.
  • Data analysis includes 2-way ANOVA (mouse group vs. treatment) myofilament function parameters (F max , EC50, and 3 ⁇ 4 obtained by fitting to Hill Equation using non-linear regression), and myofilament and cytoplasmic GSK-3P activity (p-S9/total GSK-3P).
  • a three-way repeated measures ANOVA is necessary (group, treatment, time). The variance and magnitude of change of these parameters in the canine models described in examples 1-8 are used to identify the number of mice needed.
  • GSK-3P treatments pre and post
  • data are compared via paired t-test.
  • GSK-3P deactivation in dyssynchrony may mediate myofilament calcium
  • mice can exhibit compensatory signaling cascades that could interfere with our findings.
  • GSK-3P can be knocked-down immediately before the experiment.
  • Another useful aspect of pharmacological interventions is that they may be translational, since a small molecule drug could be used in humans to treat heart failure or dyssynchrony.
  • pressure-volume loops can be conducted in the mice. Capturing PV loops in mice is not a survival surgery, so it can be done only at sacrifice. The procedure provides very sensitive indices of in vivo chamber level cardiac function.
  • Western blot is also a measure of GSK-3P activity. This correlates with activity determined by p-S9, but may be more sensitive in some cases, and represents a possible alternative. If this is done, GSK-3P is isolated from both myofilament and cytosolic fractions to determine the activity in each pool independently.
  • mice Another set of mice is subjected to the same tamoxifen treatment, pacemaker implant, recovery, and two-week pacing protocol as described, although this set would not be sacrificed after two weeks.
  • the pacemaker is turned off at the end of week two, and the mice is returned to synchronous contraction for two additional weeks. Echocardiography and ECG is performed one additional time, two weeks after cessation of pacing, immediately before sacrifice. Tissue is collected, myofilament function assessed, and GSK-3P activity measured as described above (see Figure 33 for a schematic of the protocol). Data analysis is conducted as described above, although since such analysis involves comparison to both baseline and dyssynchronous time points, ANCOVA is used.
  • mice When the mice are resynchronized (allowed to return to synchrony), the control groups recover, showing calcium re-sensitization as observed in the canine models.
  • the GSK-3p S9A mice are protected from desensitization, so there is no further recovery ( Figure 34). In knockout mice, since they cannot re-activate GSK-3P, there is no recovery of calcium sensitivity.
  • the pacemaker has the capacity for dual chamber pacing, to deliver true bi-ventricular CRT. If resynchronization via pacing cessation doesn't show recovery, an RV and LV lead is implanted, and RV pacing is used for the first two weeks, and Bi-V pacing is used for the second two weeks.
  • Example 12 Determining whether GSK-3fi localizes to the myofilament and is uncoupled from Akt
  • GSK-3P is inactive when phosphorylated at serine 9. While many kinases are phosphorylate GSK-3P at this residue, Akt is the most common. In dyssynchrony, GSK-3P phosphorylation increases, but Akt activity drops, with the reverse occurring in CRT.
  • FIG. 35 A shows GSK-3P is present in myofilament-enriched samples (using two enrichment protocols, triton and In- Sequence; Kane, L.A., et al, (2007) Methods Mol Biol 357: 87-90).
  • active GSK-3P is present in the myofilament in an alternating pattern with the Z-disk ( Figure 35B).
  • Figure 35C When phosphorylated at serine 9 (inactive form), this localization is lost ( Figure 35C).
  • GSK-3P can be determined to be localized in the myofilament using fluorescence confocal, immuno-electron microscopy, and determine its binding partner(s) using, for example, yeast-2 hybrid techniques ( Figure 35D).
  • GSK-3P Activating GSK-3P in dyssynchrony is believed to be beneficial and is believed to preserve myofilament function.
  • GSK-3P also increases hypertrophic signaling and its activation would likely have off target effects.
  • maintaining activation of a myofilament pool of GSK-3P is a useful therapy.
  • determining the kinase responsible for de-activating the myofilament pool of GSK-3P is useful. It is believed that two likely candidates are p90 rsk and PKCa, since dyssynchrony and CRT (in the dog) altered activity of both in directions commensurate with GSK-3P activity ( Figure 36A).
  • Akt has no detectable myofilament targets, and has no effect on myofilament function (Figure 36B)
  • p90 rsk and PKCa target myofilament proteins, and PKCa localizes to the myofilament.
  • PKCa and p90 rsk are also implicated in mechano-transduction signaling.
  • Identifying this upstream kinase 1) clarifies the mechanism(s) of mechanical/electrical sensing and signaling in the myocyte, 2) indicates other targets and consequences of cardiac dyssynchrony and resynchronization, and 3) reveals small molecule targets. Thus, important aspects of GSK-3P's regulation in the cardiac myocytes are identified.
  • confocal imaging and antibodies are used in, for example, control dog myocytes.
  • Such analyses is also applied to other canine models, dyssynchronous HF, synchronous HF, and CRT. The results will confirm that localization changes in response to various pacing modalities.
  • Confocal imaging is also done in the mouse models described above (e.g., GSK-3P knock-out, GSK-3p S9A , baseline, dyssynchronous pacing, resynchronized, and the like).
  • yeast two-hybrid is used to identify the critical proteins to which GSK-3P bind.
  • GSK-3P and GSK-3p S9E phospho-mimetic S ⁇ E, deactivated are used as bait proteins to probe a human heart library of expressed proteins for interacting protein partners, using, for example, Clontech's Matchmaker Gold Y2H System. Plasmids for these bait proteins are available.
  • Two-Hybrid techniques are similar to those used to identify kinase- myofilament interactions, such as AMPK-Tnl (Oliveira, S.M., et al., (2012) Circ Res
  • Data is analyzed in a number of ways. For example, some analyses involve quantitative line scans to determine myofilament localization from confocal data. In immuno- EM, many random images are collected, and the number and location of the gold-particle labeled GSK-3P is compared across groups via 1-way ANOVA. The Y2H data reveal a list of the putative interactors with the bait, either active or inactive GSK-3P, or specific amino acid sections thereof.
  • GSK-3P will be localized to the sarcomere and this localization will not change with pacing (control vs. dyssynchrony vs. CRT), but instead be affected by GSK- 3P's phosphorylation status.
  • the amount of active GSK-3P changes based on pacing, but the active GSK-3P has the same localization.
  • the data described herein indicate that GSK- 3 ⁇ binds to a thick or thin filament protein, or possibly an M-band protein.
  • the immuno- electron microscopy experiments confirm that, and narrow down exactly where GSK-3P appears.
  • the Y2H indicates the particular proteins to which GSK-3P bind.
  • the S9E GSK-3P bait disrupts this binding.
  • GSK-3P regulates calcium-handling proteins. Phosphorylation levels of calcium handling proteins are then analyzed via Western blots or mass spectrometry techniques described herein.
  • any lack of possible Y2H interactions between GSK-3P and a myofilament protein could result from: 1) GSK-3P is localized to the myofilament via a secondary binding partner (such as AKAP220), or 2) the myofilament protein GSK-3P interacts with is not currently known or included in the heart protein library. In either case, broader libraries are optionally used. If GSK-3P binds to multiple myofilament proteins, competition experiments are performed in order to determine the more likely binding partner, or to test the ability of GSK-3P to form a complex in the myofilament.
  • a secondary binding partner such as AKAP220
  • the kinase responsible phosphorylating and thus inactivating the myofilament pool of GSK-3P is identified.
  • confirmation of whether Akt is unable to de-activate the myofilament GSK-3P is determined.
  • the activity of PKCa and p90 rsk is assessed from the mouse models of dyssynchrony and resynchronization using antibody-based approaches.
  • the kinases are inhibited and/or activated in myocytes from the dog and mice models.
  • Myocytes are isolated, as previously described in both canine (Chakir, K., et al, (2009) Circulation 119(9): 1231-40.) and mouse (Zhang, M., et al, (2012) Circulation 126(8):942-51.) models.
  • Upstream kinases are optionally inhibited using pharmacological inhibitors, such as chelerythrine chloride (general PKC), GO 6976 (PKCa and PKCp specific), LY 333531 hydrochloride (PKCp specific), and BI-D1870 (p90 rsk specific [80]).
  • pharmacological inhibitors such as chelerythrine chloride (general PKC), GO 6976 (PKCa and PKCp specific), LY 333531 hydrochloride (PKCp specific), and BI-D1870 (p90 rsk specific [80]).
  • Akt inhibition is also observed via treatment with Akt Inhibitor XI (Millipore) that does not inhibit any other upstream kinases.
  • Kinase activity is optionally increased by trans fecting with active Akt, PKC, or p90 rsk .
  • cells are fixed, stained with total GSK-3P, phosphor-serine 9 GSK-3P, and a- actinin, and observed using a confocal fluorescence microscope.
  • a second pool of cells is myofilament enriched (the cytosolic fraction will also be kept and tested), and changes in GSK-3P phosphorylation are assessed using standard Western blots.
  • Data analysis for some experiments involves GSK-3P localization (via confocal imaging and line scans), and GSK-3P activity in the myo filament-enriched and myo filament-depleted samples.
  • One-way ANOVA is used to compare these data across the multiple groups.
  • PKCa or p90 rsk act as a de-activator of myofilament GSK-3P in the dog, and they likely follow the same pattern in the mouse models (increased in dyssynchrony and decreased in resynchronization). Whichever one is primarily responsible increases myo filament-bound GSK-3P phosphorylation, and possibly disrupts the localization pattern. Either inhibition or activation of Akt, on the other hand has no effect on GSK-3P de-activation with dyssynchrony. In mice with S9A, the myocytes are not disrupted by treatment with kinase.
  • kinases are also analyzed, such as other PKC isozymes and p70, if neither PKCa nor p90 rsk interact with myofilament GSK-3p.
  • siRNA is used to inhibit kinases if pharmacological inhibition is not specific enough.
  • Bio-informatics identifies a subset of Z-disk and M-band proteins as GSK-3P targets, which are confirmed in vitro (Table 4).
  • High-value targets include obscurin (S4809), actin- binding LIM protein 1 (abLIMl, S421), and filamin-C (S2228).
  • Obscurin is present in both the Z-disk, where it binds to titin, and the M-band, where it binds to myomesin.
  • abLIMl is also present in the Z-disk, and phosphorylation at S421 is in the region thought to bind to actin.
  • Filamin-C is an actin-binding protein, localized within the Z-disk.
  • phosphatase inhibitors Whether similar targets are identified in the mouse model as in the dog, and/or identify the subset shared by both species, is determined.
  • deleting GSK-3P provides a means to definitively test the link between the sarcomere protein phosphorylation changes and the kinase that was not previously possible in the canine model.
  • MS mass spectrometry
  • SWATH is based on data-independent acquisition of all observable peptides present in every sample. Based on peptide fragmentation databases that are unique to a particular sample type, thousands of proteins and their modified forms are tracked in every subsequent experimental sample analyzed. Since SWATH provides complete and permanent recording of all peptides in a sample, it is particularly useful when analyzing proteins containing modifiable residues. Thus, it combines the high-throughput of shotgun techniques, and the reproducibility and sensitivity of single/multiple reaction monitoring (SRM/MRM). A 5600 Triple TOF MS is optionally used for the data collection.
  • SRM/MRM single/multiple reaction monitoring
  • GSK-3p fl/fl /Cre without tamoxifen treatment is compared to those receiving the tamoxifen treatment with Cre but not flox'd GSK-3P since the tamoxifen +MCM model can impact heart function itself.
  • the difference between groups represents GSK-3P targeted phosphorylation.
  • the gradual knock-out and subsequent stabilization period (3-4 weeks) may result in compensatory changes in other kinases.
  • myofilament samples from GSK-3P knock-out mice are optionally treated with active GSK-3P to determine if this reverses the difference.
  • Data analysis involves quantification of peak area (performed in duplicate) of phospho-peptides between groups.
  • SWATH provides sensitive phospho data without the need for TiO phospho- enrichment, meaning that the phospho form can be normalized to the un-phosphorylated form, providing a sensitive metric.
  • primary culture of adult rabbit myocytes is infected with adenoviruses coding for obscurin WT , obscurin S4809E , obscurin S4809A , abLIMl WT , abLIMl S421E , abLIMl S421A , filamin-C WT , filamin-C S2228E , filamin-C S2228A
  • the viral expression vectors are optionally tagged, such as with a fluorescent tag (GFP, red-cherry), and tagged vectors optionally serve as negative controls.
  • the myocytes are then prepared for myofilament function experiments as described above. A striated expression pattern is used to identify cells incorporating the mutant protein, and those are used to assess myofilament function.
  • the phospho-mimetic mutant displays an increase in calcium sensitivity, while the phospho-silenced exhibits a decrease in calcium sensitivity. Moreover, both mutants block the calcium sensitizing effect of GSK-3P treatment, or at least reduce it - depending on the amount of expression of mutant protein.
  • sites are identified that have other effects on myofilament function.
  • other possible functional experiments include: stretch-release experiments to measure cross-bridge turnover dynamics (k ti ); sinusoidal stretch to measure crossbridge stiffness; and in vitro ATPase activity (Kirk, J.A., et ah, (2009) Circ Res 105(12): 1232-9.). In addition to finding the single residue that mediates the
  • Biomarker Actin binding LIM protein 1 (AbLIMl)
  • Biomarker LIM domain only protein 7 (LMO-7)
  • Biomarker LIM domain-binding protein 3 (LDB3/Cypher)
  • Biomarker Striated muscle preferentially expressed kinase (SPEG) mRNA Sequence

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cell Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Cardiology (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to methods for identifying, assessing, preventing, and treating cardiac disorders (e.g., ventricular dyssynchrony in humans), wherein alterations in the amount and/or activity of one or more biomarkers selected from GSK-3β and phosphorylation targets thereof is associated with cardiac disorder status and indicates amenability to treatment or prevention by modulating the amount and/or activity of such biomarkers.

Description

METHODS OF IDENTIFYING, ASSESSING,
PREVENTING, AND TREATING CARDIAC
DISORDERS USING GSK-3B AND
PHOSPHORYLATION TARGETS THEREOF
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application No. 61/883,677, filed on September 27, 2013, and U.S. Provisional Application No. 61/919,100, filed on December 20, 2013, the entire contents of said applications are incorporated by reference herein in their entirety.
GOVERNMENT INTEREST
This invention was made with government support under Grants NIH P01-HL077180 and T32-HL007227 awarded by the National Institutes of Health. The U.S. government has certain rights in the invention. This statement is included solely to comply with 37 C.F.R. § 401.14(a)(f)(4) and should not be taken as an assertion or admission that the application discloses and/or claims only one invention.
BACKGROUND
For the past 30 years or more, the quest to develop treatments that directly improve systolic performance of the failing heart while also conferring long-term survival benefits, has been met with disappointment. Agents that stimulate the βΐ -adrenergic receptor or enhance cyclic- AMP {e.g. , PDE3 inhibitors) work in the near-term, but have worsened chronic outcomes. This has led some to question whether safe and effective systolic therapy for the failing heart is an oxymoron.
Approximately 30% of heart failure patients develop ventricular dyssynchrony, which significantly worsens morbidity and mortality. Dyssynchrony, which is caused by conduction delays, can be treated with cardiac resynchronization therapy (CRT), but diagnosing the disease can be difficult. CRT restores coordinate contraction in heart failure patients who also have ventricular dyssynchrony due to conduction abnormalities. Its improvement of systolic function occurs within one beat, and is associated with negligible or even reduced myocardial oxygen consumption, indicating improved chamber efficiency. While dyssynchrony is an independent risk factor in patients with heart failure (HF), successful CRT reduces mortality perhaps to levels even below those of HF patients without dyssynchrony. The effects of CRT were initially attributed to its impact on chamber mechano- energetics. However, recent studies have revealed many changes at the cellular and molecular level as well, and these present a unique profile for CRT unmatched by other heart failure treatments to date. For example, marked heterogeneity of regional gene expression is reversed, the cytoprotective Akt pathway is stimulated and is coupled with reduced apoptosis, calcium handling and associated ultrastructure of the sarcoplasmic reticulum improve, mitochondrial efficiency and ATP synthase activity are enhanced via a redox switch, ion channel function regulating the action potential is improved, and resting as well as β- adrenergic stimulated myocyte function are markedly restored by a mechanism involving regulators of G-protein signaling. While CRT substantially improves resting myocyte function throughout the ventricle, this it is not consistently matched by increases in the calcium transient; suggesting CRT may also enhance myofilament responses to calcium. This is intriguing, as small molecule HF therapies that target the myofilament are of growing interest.
Current state-of-the-art techniques for diagnosing cardiac dyssynchrony involve electrical measurements of the heart (via ECG), or imaging techniques. These techniques are inadequate, however, as 30-50% of patients diagnosed with dyssynchrony fail to respond to CRT. Thus, there is a clear need in the art to develop more robust predictors of ventricular dyssynchrony and, in particular, to determine who will be a non-responder and whether there is a unique intervention for this disease subgroup. In addition, without an understanding of the molecular mechanisms by which CRT can enhance myofilament function responsible for contraction, pharmacologic or other modalities of mimicking the effects of CRT cannot be developed. Accordingly, there is also a need in the art to identify the molecular mechanisms by which CRT enhances contractile function. SUMMARY
In one aspect, the disclosure provides a method of determining whether a subject is afflicted with a cardiac disorder, the method comprising: a) determining the level of expression or level of activity of one or more biomarkers selected from glycogen synthase kinase 3β (GSK-3P) and the phosphorylation targets of GSK-3P in a biological sample obtained from the subject; b) determining the level of expression or level of activity of the one or more biomarkers in a control sample; and c) comparing the level of expression or level of activity of said one or more biomarkers detected in steps a) and b). A significant decrease in the level of expression or level of activity of the one or more biomarkers in the subject sample relative to the control level of expression or level of activity of the one or more biomarkers indicates that the subject is afflicted with the cardiac disorder.
In another aspect, the disclosure features a method of determining whether a subject afflicted with a cardiac disorder or at risk for developing a cardiac disorder would benefit from cardiac resynchronization therapy (CRT) or enhanced activity of GSK-3P, which method comprises: a) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P in a biological sample obtained from the subject; b) determining the level of expression or level of activity of the one or more biomarkers in a control sample; and c) comparing the level of expression or level of activity of said one or more biomarkers detected in steps a) and b), optionally wherein between the first point in time and the subsequent point in time, the subject has undergone treatment for the cardiac disorder; wherein a significant decrease in the level of expression or level of activity of the one or more biomarkers in the subject sample relative to the control level of expression or level of activity of the one or more biomarkers indicates that the subject afflicted with the cardiac disorder or at risk for developing the cardiac disorder would benefit from CRT or enhanced activity of GSK-3p.
In another aspect, the disclosure features a method for monitoring the progression of a cardiac disorder in a subject. The method comprises: a) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P in a biological sample obtained from the subject at a first point; b) repeating step a) at a subsequent point in time; and c) comparing the level of expression or level of activity of said one or more biomarkers detected in steps a) and b) to monitor the progression of the cardiac disorder.
In another aspect, the disclosure features a method for stratifying subjects afflicted with a cardiac disorder according to predicted clinical outcome of treatment with CRT or one or more GSK-3P modulators, optionally wherein the predicted clinical outcome is (a) restored mechanical cardiac synchrony, (b) decreased morbidity, or (c) increased survival time resulting from treatment with one or more modulators of GSK-3p. The method comprises: a) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P, in a biological sample obtained from the subject; b) determining the level of expression or level of activity of the one or more biomarkers in a control sample; and c) comparing the level of expression or level of activity of said one or more biomarkers detected in steps a) and b); wherein a significant modulation, optionally a decrease, in the level of expression or level of activity of the one or more biomarkers in the subject sample relative to the control level of expression or level of activity of the one or more biomarkers predicts the clinical outcome of the patient to treatment with CRT or one or more GSK-3P modulators.
In some embodiments, any of the methods described herein can comprise determining the level of expression of the one or more biomarkers in each sample. The level of expression may be determined by contacting each sample, or one or more nucleic acids derived from each sample, with one or more nucleic acid arrays or one or more nucleic acid probes. The level of expression may be determined by performing qPCR on each sample or by performing qPCR on one or more nucleic acids derived from each sample.
In some embodiments, any of the methods described herein can comprise determining the level of activity of the one or more biomarkers in each sample. The level of activity may be determined by analyzing one or more biomarkers in each sample by mass spectroscopy. The level of activity may be determined by contacting each sample with an antibody that specifically binds to the one or more biomarkers.
In some embodiments, any of the methods described herein can comprise treating the subject with a therapeutic agent that specifically modulates the level of expression or level of activity of the one or more biomarkers. The method may comprise treating the subject with one or more modulators of GSK-3p. The modulator may increase the expression level of GSK-3P or increases the phosphorylation state of GSK-3p.
In some embodiments, the methods described herein may comprise treating the subject in need thereof with a therapeutically effective amount of a therapeutic agent that specifically modulates the level of expression or level of activity of the one or more biomarkers. For example, the method may comprise treating the subject with one or more modulators of GSK- 3β. The modulator may increase the expression level of GSK-3P or increase the
phosphorylation state of GSK-3p.
In yet another aspect, the disclosure features a method of determining the efficacy of a test compound for treating a cardiac disorder in a subject. The method comprises: a) exposing a first sample obtained from a subject to a test compound b) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P in the first sample , optionally wherein the sample is analyzed in vivo, ex vivo, or in vitro; c) determining the level of expression or level of activity of the one or more biomarkers in a second sample obtained from the subject, wherein the second sample is not exposed to the test compound; and d) comparing the level of expression or level of activity of the one or more biomarkers in the first and second samples, wherein the test compound has efficacy for treating a cardiac disorder if the level of expression or level of activity is greater in the first sample than in the second sample.
In yet another aspect, the disclosure features a method of determining the efficacy of a therapy for treating a cardiac disorder in a subject. The method comprises: a) obtaining a first sample from a subject prior to providing at least a portion of the therapy to the subject; b) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P in the first sample, optionally wherein the sample or therapy is analyzed in vivo, ex vivo, or in vitro; c) obtaining a second sample from the subject following provision of the portion of the therapy; d) determining the level of expression or level of activity of the one or more biomarkers in the second sample; and e) comparing the level of expression or level of activity of the one or more biomarkers in the first and second samples, wherein the therapy has efficacy for treating a cardiac disorder if the level of expression or level of activity is greater in the second sample than in the first sample.
In another aspect, the disclosure features a method for identifying a compound which treats a cardiac disorder, the method comprising: a) contacting one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P with a test compound, optionally wherein the contacting occurs in vivo, ex vivo, or in vitro; and b) determining the effect of the test compound on the level of expression or level of activity of the one or more biomarkers to thereby identify a compound which treats the cardiac disorder.
In some embodiments, the one or more biomarkers may be expressed on or in a cell. The cells may be isolated from an animal model of the cardiac disorder. In some
embodiments, the cells are from a subject afflicted with the cardiac disorder.
In another aspect, the disclosure features a method for treating a cardiac disorder, the method comprising contacting a cell with an agent that modulates, optionally increases, the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P to thereby treat the cardiac disorder. The step of contacting may occur in vivo, ex vivo, or in vitro. The method may further comprise contacting the cell with an additional agent that treats the cardiac disorder.
In some embodiments of any of the methods described herein, the control may be determined from a sample from a subject not afflicted with the cardiac disorder.
In some embodiments of any of the methods described herein, the sample may consist of or comprises body fluid, cells, cell lines, histological slides, paraffin embedded tissue, fresh frozen tissue, fresh tissue, biopsies, blood, plasma, serum, buccal scrape, saliva, cerebrospinal fluid, urine, stool, mucus, or bone marrow, obtained from the subject. For example, the sample may comprise body fluid, and the body fluid may be selected from amniotic fluid, aqueous humor, bile, blood and blood plasma, cerebrospinal fluid, cerumen and earwax, cowper's fluid or pre-ejaculatory fluid, chyle, chyme, stool, female ejaculate, interstitial fluid, intracellular fluid, lymph, menses, breast milk, mucus, pleural fluid, peritoneal fluid, pus, saliva, sebum, semen, serum, sweat, synovial fluid, tears, urine, vaginal lubrication, vitreous humor, and vomit. In some embodiments, the sample is blood.
The expression level of the one or more biomarkers may be assessed by detecting the presence in the samples of a polynucleotide molecule encoding the biomarker or a portion of said polynucleotide molecule. The polynucleotide molecule may be a mR A, cDNA, or functional variants or fragments thereof and, optionally, wherein the step of detecting further comprises amplifying the polynucleotide molecule.
The expression level of the one or more biomarkers may be assessed by annealing a nucleic acid probe with the sample of the polynucleotide encoding the one or more biomarkers or a portion of said polynucleotide molecule under stringent hybridization conditions.
In some embodiments, the expression level of the biomarker is assessed by detecting the presence in the samples, whether phosphorylated, unphosphorylated, or both, of a protein of the biomarker, a polypeptide, or protein fragment thereof comprising said protein. The presence of the protein, polypeptide or protein fragment thereof may be detected using a reagent which specifically binds with said protein, polypeptide or protein fragment thereof, optionally wherein the reagent is selected from an antibody, an antibody derivative, and an antibody fragment.
The activity level of the biomarker may be assessed by determining the magnitude of modulation of the activity or expression level of downstream targets of the one or more biomarkers.
In some embodiments, determining the level of expression or level of activity of the one or more biomarkers comprises determining the phosphorylation state of GSK-3P or one or more phosphorylation targets of GSK-3p. The phosphorylation state may be determined, for example, either by mass spectroscopy or by using an antibody that specifically binds to the phosphorylated form of the protein.
The phosphorylation targets of GSK-3P may be myofilament phosphorylation targets. For example, the myofilament phosphorylation targets may be selected from the group listed in Tables 2-5. In some embodiments, the phosphorylation targets of GSK-3P are selected from Tnl, MyBPC, TnT, MLC2, cMyBP-C, cTnl, a-Tropomyosin, titin, obscurin, Actin binding LIM protein 1 (AbLIMl), Tensin-1, Thyroid hormone receptor-associated protein 3 (THRAP-3), Nestin, Sorbin and SH3 domain-containing protein 2 (SORB-2), LIM domain only protein 7 (LMO-7), LIM domain-binding protein 3 (LDB3/Cypher), Striated muscle preferentially expressed kinase (SPEG), Filamin-C, Myotilin 1, and Leiomodin-2. The phosphorylation targets GSK-3P may be selected from titin, obscurin, AbLIMl, Tensin-1, THRAP-3, Nestin, SORB-2, LMO-7, LDB3, SPEG, Filamin-C, Myotilin 1, and Leiomodin 2.
In some embodiments, the cardiac disorder is heart failure or ventricular
dyssynchrony.
In some embodiments, the subject is a mammal, optionally a human.
In some embodiments of any of the methods described herein, the expression level, level of activity, or phosphorylation status of at least two (e.g., at least three, four, five, six, seven, or eight) biomarkers is determined.
In some embodiments, any of the methods described herein can further include generating a risk score related to the expression level, level of activity, or phosphorylation status (e.g., level of phosphorylation) of any of the biomarkers described herein. The risk score can be used, e.g., to classify a subject as one in need of treatment, one at risk for developing a cardiac disorder, or one who has benefited from therapy.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows a timeline of pacing protocols in each of the canine models: Control, dyssynchronous heart failure (HFdys), cardiac resynchronization therapy (CRT), synchronous heart failure (HFsync), V3A3 (3 weeks RV pacing to induce dyssynchrony, 3 weeks atrial pacing to restore synchrony), and AVA (synchronous heart failure with two weeks of induced transient dyssynchrony via RV pacing). The HFdys and CRT models receive left bundle branch block via radio-frequency ablation. After a one-week recovery period from the pacemaker implant, each pacing protocol lasts 6 weeks before the animal is sacrificed.
Figures 2A-2G show that CRT globally restores myofilament function. (A) Force- calcium data and fitted curves for myocytes from the left ventricular lateral wall for Con (Control, filled circles, n = 15 myocytes from 6 dogs), HFdys (dyssynchronous heart failure, open triangles, n = 16 myocytes from 6 dogs) and CRT (cardiac resynchronization therapy, gray squares, n = 16 myocytes from 7 dogs). Inset: force normalized to maximum calcium- activated force (Fmax) to visualize the change in calcium sensitivity. (B) Fmax is decreased by -50% in HFdys, and restored by CRT. HFdys causes desensitization to calcium (increase in EC50), but is restored to Control levels with CRT. (C) Force-calcium data and fitted curves for right ventricular trabeculae muscles for Control (filled circles, n = 14 muscles from 8 dogs), HFdys (open triangles, n = 24 muscles from 13 dogs), and CRT (gray squares, n = 18 muscles from 10 dogs). Inset: force normalized to Fmax to visualize the change in calcium sensitivity.
(D) Fmax and EC50 were altered in the right ventricle to a similar extent as in the left ventricle.
(E) Tension cost, the ratio of ATPase activity to developed tension, was reduced in both heart failure models, and not recovered by CRT. (F) Cross-bridge turnover dynamics (kto) were also reduced in the HF models, and not restored by CRT. (G) Lastly, stiffness was not affected in any of the groups. Values are mean ± s.e.m. *, P < 0.05 vs. Control, #, P < 0.05 vs. HFdys by one-way ANOVA.
Figures 3A-3D show that skinned myocytes from the left anterior septum exhibited the same functional changes as those from the LV lateral wall. (A, B) Force-calcium data and curves, showing (A) actual force and (B) normalized force. (C) Compared to Con (n = S myocytes from 3 dogs), maximum force was reduced in HFdys (n = 7 myocytes from 3 dogs) and restored by CRT (n = S myocytes from 3 dogs). (D) HFdys was desensitized to calcium, but CRT restored the value to near the Con value. *, P < 0.05 vs. Con. #, P < 0.05 vs HFdys.
Figures 4A-4D show that always synchronous heart failure (HFsync) exhibited reduced myofilament function. (A, B) Force-calcium data and curves, showing (A) actual force and (B) normalized force. Con and HFdys skinned myocytes shown for comparison to HFsync (n = 15 myocytes from 5 dogs). (C) Maximum force was reduced similarly in both HFdys and HFsync. (D) HFsync exhibited a hyper-sensitization to calcium. *, P < 0.05 vs Con. #, P < 0.05
VS HFdys-
Figures 5A-5D show that myofilament function can be improved via a brief period of induced dyssynchrony. (A, B) Both maximum force and calcium sensitivity are improved in the HFsync model when the middle two weeks of the pacing protocol is made dyssynchronous (AVA model). (C, D). When dyssynchrony is induced via right ventricular pacing (instead of radio frequency ablation of the left bundle branch), and resynchronization is restored by moving the pacing site to the atrium (V3A3), the effects of CRT are recapitulated. (C) Dyssynchrony caused a decrease in maximum force (Fmax), which was increased by V3A3.
(D) Dyssynchrony desensitized the myofilament response to calcium (increase in EC50) which was also reversed in the V3A3 model. *, P < 0.05 vs HFsync or HFdys.
Figures 6A-6E show that there were no changes in protein degradation or isoforms between Control, HFdys, and CRT. (A) High sensitivity silver stain of myofilament enriched samples and the gray-intensity for the each group. There were no differences in any bands between the three groups (n = 3 per group). (B) Lower sensitivity coomassie stain was also used to more accurately quantify the higher abundant proteins. The lack of changes was also confirmed with antibodies specific to (C) Tnl, (D) TnT, and (E) MLC2, all of which showed secondary bands (representing 1-10% of the total) were was unchanged in HFdys or CRT (n = 3 per group).
Figures 7A-7E shows that the apparent decrease in Fmax is due to myocyte
hypertrophy. (A) Skinned myocytes from Con, HFdys, and CRT. (B) Cross-sectional area (calculated by measuring both width and depth and assuming an elliptical shape) was significantly increased in HFdys, but restored to normal levels in CRT. (C) The un-biased approach also showed that HFdys myocytes were hypertrophied, and this was reversed by CRT (n = 150 myocytes from 3 dogs, per group). (D) Fmax is typically normalized to CSA, but if it is not, the myocytes generate the same amount of force between the groups, indicating that the force producing capacity of the myocytes is not altered, they are just bigger. (E) There was no difference in trabeculae muscle size, despite the fact that a decrease in Fmax was also observed. This suggests that there are fewer myocytes in HFdys in a constant area of myocardium.
Values are mean ± s.e.m. *, P < 0.05 vs. Control, #, P < 0.05 vs. HFdys by one-way ANOVA.
Figures 8A-8F show that calcium sensitization is mediated by phosphorylation, but not at the known targets. (A) Representative force-calcium curves for Con, HFdys, and CRT before protein phosphatase- 1 (PP1) and post treatment. (B) While there was no change in Fmax, PP1 caused a desensitization in the Con (n = 6) and CRT (n =5) groups, but had no effect on HFdys (n = 5). (C) Top, phos-Tag gel blotted for troponin I (Tnl). Bottom, there was an increase in the un-phosphorylated form, and a decrease in the bi-phosphorylated form of Tnl in the HFdys, and CRT groups (n = 3), indicating overall de -phosphorylation of Tnl in heart failure. (D) Top, western blot with antibodies against 3 phosphorylation sites on myosin binding protein C (MyBPC). Bottom, both HFdys and CRT showed decreased phosphorylation at S273 and S282 (n = 3). (E) Top, western blot against S 15 phosphorylation on myosin light chain 2 (MLC2) and phospho-serine motif sites on MLC2. Bottom, HFdys (n = 4) and CRT (n = 4) were decreased from Con (n = 3), but no different from each other at S I 5, and there was no change in overall serine phosphorylation. (F) Top, phospho-motif antibodies for serine and threonine sites and total troponin T (TnT). Bottom, there was very little phosphorylation of TnT in the 3 groups, with no differences between them (n = 3). Values are mean ± s.e.m. *, P < 0.05 vs. Control by one-way ANOVA. Figures 9A-9B show the effect of PPl in all dog models. (A) The change in Fmax in each of the dog models in response to PPl treatment. None of the groups showed any change in Fmax post-treatment. (B) As presented in Figure 5B, Con and CRT each experienced desensitization to calcium in response to PPl treatment, while there was no effect in HFdys. Here, the other groups (HFsync, V3A3, and AVA) also exhibited calcium desensitization in response to PPl treatment. This suggests that the benefits of CRT are reversed similarly in the CRT variants (V3A3 and AVA). (†, P < 0.05 vs. 0. *, P < 0.05 vs. Con).
Figure 10 shows that the phos-Tag gel (Figure 8C) showed that overall Tnl phosphorylation was decreased in HFdys and CRT, but could not show whether the PKA or PKC sites specifically were decreasing. Antibodies specific to the S22/S23 PKA sites indicate that the PKA-phosphorylated form of Tnl decreased in both HFdys and CRT (n = 4 per group), confirming the data from the Phos-Tag gel. As PKA phosphorylation was unchanged between HFdys and CRT, as was total phosphorylation, it suggests that there was also no change in any of the other highly phosphorylated sites. *, P < 0.05 vs. Con by one-way ANOVA.
Figure 11 shows that, in order to confirm there were no phosphorylation changes among the most abundant myofilament proteins between HFdys and CRT, 2D-DiGE gels were performed. The two images represent the HFdys (top) and CRT (bottom) channels that were differentially labeled with Cy5 and Cy3, respectively, and run simultaneously on the same gel. The two channels have been independently presented here for clarity. Analysis by Ludesi REDFIN confirmed that there were no phosphorylation changes (n = 4 per group). The boxes indicate a selection of myofilament proteins. Note: troponin I is not represented on these 2D gels because it has a very high pi.
Figures 12A-12C show mass spectrometry (MS) data for phosphorylation at Titin (N2BA isoform) S9901. (A) MS spectra for the peptide containing S9901. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site.
Figures 13A-13C show mass spectrometry (MS) data for phosphorylation at Obscurin S4809. (A) MS spectra for the peptide containing S4809. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified
phosphorylation site.
Figures 14A-14C show mass spectrometry (MS) data for phosphorylation at ABLIM1 S421. (A) MS spectra for the peptide containing S421. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site. Figures 15A-15C show mass spectrometry (MS) data for phosphorylation at Tensin-1 S1274. (A) MS spectra for the peptide containing S1274. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site.
Figures 16A-16C show mass spectrometry (MS) data for phosphorylation at TRAP3
S244. (A) MS spectra for the peptide containing S244. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site.
Figures 17A-17C show mass spectrometry (MS) data for phosphorylation at Nestin S 1062. (A) MS spectra for the peptide containing S 1062. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site.
Figures 18A-18C show mass spectrometry (MS) data for phosphorylation at SORB2 S40. (A) MS spectra for the peptide containing S40. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site.
Figures 19A-19C show mass spectrometry (MS) data for phosphorylation at SORB2 S231. (A) MS spectra for the peptide containing S231. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site.
Figures 20A-20C show mass spectrometry (MS) data for phosphorylation at SORB2 S307. (A) MS spectra for the peptide containing S307. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site.
Figures 21A-21C show mass spectrometry (MS) data for phosphorylation at LM07
SI 346. (A) MS spectra for the peptide containing SI 346. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site.
Figures 22A-22C show mass spectrometry (MS) data for phosphorylation at LDB3 (Cypher/Zasp) S507. (A) MS spectra for the peptide containing S507. (B) Fragmentation
Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site.
Figures 23A-23C show mass spectrometry (MS) data for phosphorylation at SPEG S2039. (A) MS spectra for the peptide containing SS2039. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site.
Figures 24A-24C show mass spectrometry (MS) data for phosphorylation at Filamin- C S2228. (A) MS spectra for the peptide containing S2228. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified
phosphorylation site.
Figures 25A-25C show mass spectrometry (MS) data for phosphorylation at Myotilin S231. (A) MS spectra for the peptide containing S231. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site.
Figures 26A-26C show mass spectrometry (MS) data for phosphorylation at
Leiomodin-2 S513. (A) MS spectra for the peptide containing S513. (B) Fragmentation Table. (C) Known domains and protein binding regions shown with location of the identified phosphorylation site.
Figures 27A-27C show increased phosphorylation at GSK-3P sites in CRT mediated the calcium sensitizing effect. (A) Schematic of the cardiac sarcomere showing Z-disk (vertical lines), titin (labeled; diagonal lines), thick filament (branched structures), M-line (middle/center ), and thin filament (horizontal lines). The proteins (and sites) that were identified as increased with CRT are shown, with their best-known localization. *, matches GSK-3P consensus sequence. (B) Representative force-calcium relationships in Con (top), HFdys (middle) and CRT (bottom) before (solid line) and after GSK-3P treatment (dashed line). (C) There was a calcium sensitizing effect of GSK-3P in HFdys (n = 8), bringing it to the same level as Con and CRT. GSK-3P had no effect on Con (n = 5) or CRT myocytes (n = 8). Values are mean ± s.e.m. *, P < 0.05 vs. Con baseline value.
Figures 28A-28B show the effect of PKA treatment on HFdys and CRT myocytes. (A)
There was a small, but significant, increase in Fmax in response to PKA treatment, in both HFdys and CRT. (B) It is known that PKA has a desensitizing effect on the myofilament, and this was observed equally in both models. *, P < 0.05 vs. pre-treatment; #, P < 0.05 vs. HFdys with corresponding treatment; via two-way ANOVA. The interaction term was not significant for either panel.
Figures 29A-29E show the effect of Akt treatment on HFdys and CRT myocytes. (A and B) Akt treatment had no effect on either Fmax or EC50 in either group. (C) Akt did not induce any detectable phosphorylation of myofilament targets, by Pro-Q Diamond phospho- stain. (D) Akt treatment did induce phosphorylation of known targets in both groups, as shown by phosphorylation of serine 9 on GSK-3P (cytosolic fraction). (E) There was a statistically significant increase in GSK-3P serine 9 phosphorylation after Akt treatment. There was a borderline significant higher phosphorylation in HFdys compared to CRT (P = 0.07). *, P < 0.05 vs. pre-treatment; #, P < 0.05 vs. HFdys with corresponding treatment; via two-way ANOVA.
Figures 30A-30E show the time course of function and phosphorylation during dyssynchrony and heart failure. (A) Endocardial biopsies taken at the 3-week time point (n, Con: 8, HFdys 3 weeks: 11, HFsync 3 weeks: 5, HFdys 6 weeks: 17, HFsync 6 weeks: 18 myocytes) showed that Fmax decreased steadily over the entire 6-week protocol. (B) Calcium sensitivity was altered in HFsync by 3 weeks, with no changes between 3 and 6 weeks.
However, HFdys was unaltered at 3 weeks, but desensitized between 3 and 6 weeks. (C) When the sensitizing temporal effect of decreased Tnl phosphorylation is added to the desensitizing temporal effects of decreased GSK-3P activity, it recapitulates the observed phenotype in panel B. (D) S22/S23 Tnl phosphorylation was decreased in both HFdys and HFsync by 3 weeks, with no additional decrease between 3 and 6 weeks (n = 5 per time point). (E) Phospho GSK-3P was unchanged in HFsync over the 6 weeks, but linearly increased in HFdys, P < 0.01 via multivariate regression analysis. *, P < 0.05 vs. Con. #, P < 0.05 vs. HFsync at 3 weeks.
Figures 31A-31E show the details of an embodiment of a mouse cardiac pacemaker. Figures 31A-31B show a mouse pacemaker chip (Figure 31 A) and a pacing lead (Figure 3 IB). Figure 31C shows an ECG confirming capture of cardiac pacing data. Figure 3 ID shows an echo showing dyssynchrony with RV pacing. Figure 3 IE shows that RV pacing decreases systolic function.
Figures 32A-32C show results of cardiac paced mice. Cardiac dyssynchrony was induced in mice using a custom RV pacing lead and pacemaker system. Figure 32A shows that RV pacing, causing dyssynchrony, caused calcium desensitization compared to those mice with an implanted pacing lead, but no pacing. There was no effect on maximum force, however, with RV pacing. This replicates what was observed in the canine model of dyssynchrony, which resulted in reduced calcium sensitivity, as a consequence of reduced phosphorylation of GSK-3P's myofilament targets. Figure 32B shows that with RV pacing, there was an increase in the ratio of phospho-serine 9 GSK-3P to total GSK-3P, indicating kinase de-activation in dyssynchrony. That both GSK-3P de-activation and calcium desensitization occurred in the mouse model with dyssynchrony but no heart failure, supports that these phenotypes are specific to cardiac dyssynchrony. Figure 32C shows that genetic knock-out of GSK-3P in a mouse results in myofilament Ca2+ desensitization. Skinned myocytes from inducible cardiomyocyte-specific GSK-3P knockout mice (Woulfe, Circ Res, 2010) were compared to myocytes from mice expressing WT GSK-3p. There was a significant reduction in calcium sensitivity, with no change in maximum force when GSK-3P was knocked out. This agrees with the canine data showing that GSK-3P deactivation in cardiac dyssynchrony results in calcium desensitization, directly implicating GSK-3p.
Figure 33 shows an embodiment of a pacing protocol.
Figure 34 shows the expected results from the pacing protocol shown in Figure 33 and related experiments described herein.
Figure 35A-35D show GSK-3P expression results. Figure 36A shows that GSK-3P is present in myo filament-enriched samples. Figure 36B shows GSK-3P myofilament localization. Figure 36C shows that this localization is with GSK-3P de-activation. Figure 36D shows cardiac myofilament electron microscopy (EM).
Figure 36A-36B show biological relationships between GSK-3P, PKCa, p90rsk, and Akt. Figure 36A shows increased PKCa and p90rsk activity with HFdys. Figure 36B shows that Akt has no effect on myofilament function.
Figure 37 shows a schematic diagram of GSK-3P targets with known binding domains.
DETAILED DESCRIPTION
Overview
The invention relates, at least in part, to the discovery that the kinase glycogen synthase kinase 3β (GSK-3P) is specifically de-activated in ventricular dyssynchrony. This de-activation occurs via its phosphorylation at serine 9. This de-activation is reversed in response to CRT, and does not occur in synchronous heart failure, indicating that it is a specific biomarker for cardiac dyssynchrony. While a biomarker for GSK-3P activation would require a cardiac biopsy, GSK-3P has several myofilament targets, which have been discovered herein to be detectable in the blood when the myocardium is damaged.
Phosphorylation of these sites is increased by CRT, in concordance with re-activation of GSK-3p. These phosphorylation sites therefore represent biomarkers correlating with myocardial GSK-3P activity and cardiac dyssynchrony. In some embodiments of the invention, these phosphorylation sites are detected with very high sensitivity using multiple reaction monitoring (MRM). In addition to a diagnostic biomarker, GSK-3P activity and the myofilament phosphorylation targets represent a useful tool for drug screening. Allowing for in vitro testing of therapeutics to replace CRT, or translate CRT to a more diverse patient population. Knowing the phosphorylation status of one or more of these proteins (e.g. , in biopsies or in circulating blood) provides molecular insights into whether an individual will be a responder to therapy.
Accordingly, the detection, quantification, and/or use of target phosphorylation sites described herein in tissue or circulating cells or proteins as indication of the level of GSK-3P activity in the heart, and thus, whether a person will be responsive or not to CRT therapy and/or will responding after implantation of device mediating CRT therapy. As described herein, embodiments of the present invention can make use of well-known reagents and techniques for determining the status of a biomarker described herein (e.g. , publicly available antibody, mass spectrometry, nucleic acid probe, and similar reagents). Moreover, routine adaptions of the methods described herein, such as assaying the presence of phosphorylation or quantitative increase or decrease in levels over time, over different courses of therapy, in therapeutic agent screening protocols, and the like, are contemplated. For example, assays for screening test agents to determine whether they activate the GSK-3P pathway and mimic CRT cellular contractile effects are contemplated.
I. Definitions
The articles "a" and "an" are used herein to refer to one or to more than one (i.e. to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element.
The amount of a biomarker in a subject is "significantly" higher or lower than the normal amount of the biomarker, if the amount of the biomarker is greater or less,
respectively, than the normal level by an amount greater than the standard error of the assay employed to assess amount, and preferably at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 300%, 350%, 400%, 500%, 600%, 700%, 800%, 900%, 1000% or than that amount. Alternately, the amount of the biomarker in the subject can be considered "significantly" higher or lower than the normal amount if the amount is at least about two, and preferably at least about three, four, or five times, higher or lower, respectively, than the normal amount of the biomarker. Such "significance" can also be applied to any other measured parameter described herein, such as for expression, inhibition, phosphorylation, and the like.
The term "altered level of expression" of a biomarker refers to an expression level or copy number of the biomarker in a test sample, that is greater or less than the standard error of the assay employed to assess expression or copy number, and is preferably at least twice, and more preferably three, four, five or ten or more times the expression level or copy number of the biomarker in a control sample (e.g., a sample from a healthy subject) and preferably, the average expression level or copy number of the biomarker in several control samples. The altered level of expression is greater or less than the standard error of the assay employed to assess expression or copy number, and is preferably at least twice, and more preferably three, four, five or ten or more times the expression level or copy number of the biomarker in a control sample (e.g., sample from a healthy subjects not having the associated disease) and preferably, the average expression level or copy number of the biomarker in several control samples. This also applies to changes in the level of phosphorylation or phosphorylation status of a biomarker protein.
The term "altered activity" of a biomarker refers to an activity of the biomarker which is increased or decreased in a disease state as compared to the activity of the biomarker in a normal, control sample. Altered activity of the biomarker may be the result of, for example, altered expression of the biomarker, altered phosphorylation the biomarker, or, e.g., an altered interaction with other proteins involved in the same or different pathway as the biomarker or altered interaction with transcriptional activators or inhibitors.
Unless otherwise specified here within, the terms "antibody" and "antibodies" broadly encompass naturally-occurring forms of antibodies (e.g. IgG, IgA, IgM, IgE) and recombinant antibodies such as single-chain antibodies, chimeric and humanized antibodies and multi- specific antibodies, as well as fragments and derivatives of all of the foregoing, which fragments and derivatives have at least an antigenic binding site. Antibody derivatives may comprise a protein or chemical moiety conjugated to an antibody.
Antibodies may be polyclonal or monoclonal; xenogeneic, allogeneic, or syngeneic; or modified forms thereof (e.g. humanized, chimeric, etc.). Antibodies may also be fully human. Preferably, antibodies of the invention bind specifically or substantially specifically to a biomarker polypeptide or fragment thereof. The terms "monoclonal antibodies" and
"monoclonal antibody composition," as used herein, refer to a population of antibody polypeptides that contain only one species of an antigen binding site capable of
immunoreacting with a particular epitope of an antigen, whereas the term "polyclonal antibodies" and "polyclonal antibody composition" refer to a population of antibody polypeptides that contain multiple species of antigen binding sites capable of interacting with a particular antigen. A monoclonal antibody composition typically displays a single binding affinity for a particular antigen with which it immunoreacts. The term "biomarker" refers to a measurable entity of the present invention that has been determined to be predictive of a cardiac response. Biomarkers can include, without limitation, nucleic acids, proteins, and metabolites, particularly those shown in Tables 2-6.
The term "body fluid" refers to fluids that are excreted or secreted from the body as well as fluids that are normally not (e.g., bronchoalveolar lavage fiuid, amniotic fluid, aqueous humor, bile, blood and blood plasma, cerebrospinal fluid, cerumen and earwax, cowper's f uid or pre-ejaculatory fluid, chyle, chyme, stool, female ejaculate, interstitial fluid, intracellular f uid, lymph, menses, breast milk, mucus, pleural f uid, pus, saliva, sebum, semen, serum, sweat, synovial fluid, tears, urine, vaginal lubrication, vitreous humor, vomit).
The term "complementary" refers to the broad concept of sequence complementarity between regions of two nucleic acid strands or between two regions of the same nucleic acid strand. It is known that an adenine residue of a first nucleic acid region is capable of forming specific hydrogen bonds ("base pairing") with a residue of a second nucleic acid region which is antiparallel to the first region if the residue is thymine or uracil. Similarly, it is known that a cytosine residue of a first nucleic acid strand is capable of base pairing with a residue of a second nucleic acid strand which is antiparallel to the first strand if the residue is guanine. A first region of a nucleic acid is complementary to a second region of the same or a different nucleic acid if, when the two regions are arranged in an antiparallel fashion, at least one nucleotide residue of the first region is capable of base pairing with a residue of the second region. Preferably, the first region comprises a first portion and the second region comprises a second portion, whereby, when the first and second portions are arranged in an antiparallel fashion, at least about 50%, and preferably at least about 75%, at least about 90%, or at least about 95%) of the nucleotide residues of the first portion are capable of base pairing with nucleotide residues in the second portion. More preferably, all nucleotide residues of the first portion are capable of base pairing with nucleotide residues in the second portion.
The term "control" refers to any reference standard suitable to provide a comparison to the expression products in the test sample. In one embodiment, the control comprises obtaining a "control sample" from which expression product levels are detected and compared to the expression product levels from the test sample. Such a control sample may comprise any suitable sample, including but not limited to a sample from a control patient (can be stored sample or previous sample measurement) with a known outcome; normal tissue, blood, or cells isolated from a subject, such as a normal patient or the patient with a known condition, cultured primary cells/tissues isolated from a subject such as a normal subject or the patient with a known condition, a tissue or cell sample isolated from a normal subject, or primary cells/blood/tissues obtained from a depository. In another embodiment, the control may comprise a reference standard expression product level from any suitable source, including but not limited to housekeeping genes, an expression product level range from normal tissue (or other previously analyzed control sample), a previously determined expression product level range within a test sample from a group of patients, or a set of patients with a certain outcome (for example, survival for one, two, three, four years, etc.) or receiving a certain treatment (for example, standard of care therapy). It will be understood by those of skill in the art that such control samples and reference standard expression product levels can be used in combination as controls in the methods of the present invention. In one embodiment, the control may comprise normal cells/blood/tissue. In another preferred embodiment, the control may comprise an expression level for a set of patients, such as a set of patients with a known condition, or for a set of patients receiving a certain treatment, or for a set of patients with one outcome versus another outcome. In the former case, the specific expression product level of each patient can be assigned to a percentile level of expression, or expressed as either higher or lower than the mean or average of the reference standard expression level. In another preferred embodiment, the control may comprise normal cells, blood, or tissue from patients treated with therapy. In another embodiment, the control may also comprise a measured value for example, average level of expression of a particular gene in a population compared to the level of expression of a housekeeping gene in the same population. Such a population may comprise normal subjects, patients who have not undergone any treatment (i.e., treatment naive), or patients undergoing standard of care therapy. In another preferred embodiment, the control comprises a ratio transformation of expression product levels, including but not limited to determining a ratio of expression product levels of two genes in the test sample and comparing it to any suitable ratio of the same two genes in a reference standard; determining expression product levels of the two or more genes in the test sample and determining a difference in expression product levels in any suitable control; and determining expression product levels of the two or more genes in the test sample, normalizing their expression to expression of housekeeping genes in the test sample, and comparing to any suitable control. In particularly preferred embodiments, the control comprises a control sample which is of the same lineage and/or type as the test sample. In another embodiment, the control may comprise expression product levels grouped as percentiles within or based on a set of patient samples, such as all patients with a certain condition. In one embodiment a control expression product level is established wherein higher or lower levels of expression product relative to, for instance, a particular percentile, are used as the basis for predicting outcome. In another preferred embodiment, a control expression product level is established using expression product levels from control patients with a known outcome, and the expression product levels from the test sample are compared to the control expression product level as the basis for predicting outcome. As demonstrated by the data below, the methods of the invention are not limited to use of a specific cut-point in comparing the level of expression product in the test sample to the control.
The term "expression signature" or "signature" refers to a group of two or more coordinately expressed biomarkers. For example, the genes, proteins, metabolites, and the like making up this signature may be expressed in a specific cell lineage or during a particular biological response. Expression data and gene expression levels can be stored on computer readable media, e.g., the computer readable medium used in conjunction with a microarray or chip reading device, or in conjunction with a qPCR detection system. Such expression data can be manipulated to generate expression signatures.
The term "interaction", when referring to an interaction between two molecules, refers to the physical contact (e.g., binding) of the molecules with one another. An interaction may cause phosphorylation or Dephosphorylation of a molecule. Generally, such an interaction results in an activity (which produces a biological effect) of one or both of said molecules.
An "isolated protein" refers to a protein that is substantially free of other proteins, cellular material, separation medium, and culture medium when isolated from cells or produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized. An "isolated" or "purified" protein or biologically active portion thereof is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the antibody, polypeptide, peptide or fusion protein is derived, or substantially free from chemical precursors or other chemicals when chemically synthesized. The language "substantially free of cellular material" includes preparations of a biomarker polypeptide or fragment thereof, in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly produced. In one embodiment, the language "substantially free of cellular material" includes preparations of a biomarker protein or fragment thereof, having less than about 30% (by dry weight) of non-biomarker protein (also referred to herein as a "contaminating protein"), more preferably less than about 20% of non-biomarker protein, still more preferably less than about 10% of non-biomarker protein, and most preferably less than about 5% non-biomarker protein. When antibody, polypeptide, peptide or fusion protein or fragment thereof, e.g., a biologically active fragment thereof, is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the protein preparation.
The "normal" level of expression of a biomarker is the level of expression of the biomarker in cells of a subject, e.g., a healthy human patient. An "over-expression" or "significantly higher level of expression" of a biomarker refers to an expression level in a test sample that is greater than the standard error of the assay employed to assess expression, and is preferably at least twice, and more preferably 2.1 , 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20 times or more higher than the expression activity or level of the biomarker in a control sample (e.g. , sample from a healthy subject not having the biomarker associated disease) and preferably, the average expression level of the biomarker in several control samples. A "significantly lower level of expression" of a biomarker refers to an expression level in a test sample that is at least twice, and more preferably 2.1 , 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20 times or more lower than the expression level of the biomarker in a control sample (e.g. , sample from a healthy subject not having the biomarker associated disease) and preferably, the average expression level of the biomarker in several control samples.
An "over-expression" or "significantly higher level of expression" of a biomarker refers to an expression level in a test sample that is greater than the standard error of the assay employed to assess expression, and is preferably at least twice, and more preferably 2.1 , 2.2,
2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20 times or more higher than the expression activity or level of the biomarker in a control sample (e.g., sample from a healthy subject not having the biomarker associated disease) and preferably, the average expression level of the biomarker in several control samples. A "significantly lower level of expression" of a biomarker refers to an expression level in a test sample that is at least twice, and more preferably 2.1 , 2.2, 2.3,
2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20 times or more lower than the expression level of the biomarker in a control sample (e.g., sample from a healthy subject not having the biomarker associated disease) and preferably, the average expression level of the biomarker in several control samples. This also applies to changes in the level of phosphorylation or phosphorylation status of a biomarker protein. The terms "prevent," "preventing," "prevention," "prophylactic treatment," and the like refer to reducing the probability of developing a disease, disorder, or condition in a subject, who does not have, but is at risk of or susceptible to developing a disease, disorder, or condition.
The term "probe" refers to any molecule which is capable of selectively binding to a specifically intended target molecule, for example, a nucleotide transcript or protein encoded by or corresponding to a biomarker nucleic acid. Probes can be either synthesized by one skilled in the art, or derived from appropriate biological preparations. For purposes of detection of the target molecule, probes may be specifically designed to be labeled, as described herein. Examples of molecules that can be utilized as probes include, but are not limited to, R A, DNA, proteins, antibodies, and organic molecules.
The term "sample" used for detecting or determining the presence or level of at least one biomarker is typically whole blood, plasma, serum, saliva, urine, stool (e.g., feces), tears, and any other bodily fluid (e.g., as described above under the definition of "body fluids"), or a tissue sample (e.g., biopsy) such as a surgical resection tissue. In certain instances, the method of the present invention further comprises obtaining the sample from the individual prior to detecting or determining the presence or level of at least one marker in the sample.
The term "subject" refers to any animal, mammal or human. The term "subject" is interchangeable with "patient."
The term "therapeutic effect" refers to a local or systemic effect in animals, particularly mammals, and more particularly humans, caused by a pharmacologically active substance. The term thus means any substance intended for use in the diagnosis, cure, mitigation, treatment or prevention of disease or in the enhancement of desirable physical or mental development and conditions in an animal or human. The phrase "therapeutically- effective amount" means that amount of such a substance that produces some desired local or systemic effect at a reasonable benefit/risk ratio applicable to any treatment. In certain embodiments, a therapeutically effective amount of a compound will depend on its therapeutic index, solubility, and the like. For example, certain compounds discovered by the methods of the present invention may be administered in a sufficient amount to produce a reasonable benefit/risk ratio applicable to such treatment.
The terms "therapeutically-effective amount" and "effective amount" as used herein means that amount of a compound, material, or composition comprising a compound of the present invention which is effective for producing some desired therapeutic effect in at least a sub -population of cells in an animal at a reasonable benefit/risk ratio applicable to any medical treatment.
A "transcribed polynucleotide" or "nucleotide transcript" is a polynucleotide (e.g. an mRNA, hnRNA, a cDNA, or an analog of such RNA or cDNA) which is complementary to or homologous with all or a portion of a mature mRNA made by transcription of a biomarker nucleic acid and normal post-transcriptional processing (e.g. splicing), if any, of the RNA transcript, and reverse transcription of the RNA transcript.
II. Sequences
There is a known and definite correspondence between the amino acid sequence of a particular protein and the nucleotide sequences that can code for the protein, as defined by the genetic code (shown below). Likewise, there is a known and definite correspondence between the nucleotide sequence of a particular nucleic acid and the amino acid sequence encoded by that nucleic acid, as defined by the genetic code.
GENETIC CODE
Alanine (Ala, A) GCA, GCC, GCG, GCT
Arginine (Arg, R) AGA, ACG, CGA, CGC, CGG, CGT
Asparagine (Asn, N) AAC, AAT
Aspartic acid (Asp, D) GAC, GAT
Cysteine (Cys, C) TGC, TGT
Glutamic acid (Glu, E) GAA, GAG
Glutamine (Gin, Q) CAA, CAG
Glycine (Gly, G) GGA, GGC, GGG, GGT
Histidine (His, H) CAC, CAT
Isoleucine (He, I) ATA, ATC, ATT
Leucine (Leu, L) CTA, CTC, CTG, CTT, TTA, TTG
Lysine (Lys, K) AAA, AAG
Methionine (Met, M) ATG
Phenylalanine (Phe, F) TTC, TTT
Proline (Pro, P) CCA, CCC, CCG, CCT
Serine (Ser, S) AGC, AGT, TCA, TCC, TCG, TCT
Threonine (Thr, T) ACA, ACC, ACG, ACT
Tryptophan (Trp, W) TGG Tyrosine (Tyr, Y) TAC, TAT
Valine (Val, V) GTA, GTC, GTG, GTT
Termination signal (end) TAA, TAG, TGA
An important and well known feature of the genetic code is its redundancy, whereby, for most of the amino acids used to make proteins, more than one coding nucleotide triplet may be employed (illustrated above). Therefore, a number of different nucleotide sequences may code for a given amino acid sequence. Such nucleotide sequences are considered functionally equivalent since they result in the production of the same amino acid sequence in all organisms (although certain organisms may translate some sequences more efficiently than they do others). Moreover, occasionally, a methylated variant of a purine or pyrimidine may be found in a given nucleotide sequence. Such methylations do not affect the coding relationship between the trinucleotide codon and the corresponding amino acid.
In view of the foregoing, the nucleotide sequence of a DNA or R A encoding a biomarker nucleic acid (or any portion thereof) can be used to derive the polypeptide amino acid sequence, using the genetic code to translate the DNA or RNA into an amino acid sequence. Likewise, for polypeptide amino acid sequence, corresponding nucleotide sequences that can encode the polypeptide can be deduced from the genetic code (which, because of its redundancy, will produce multiple nucleic acid sequences for any given amino acid sequence). Thus, description and/or disclosure herein of a nucleotide sequence which encodes a polypeptide should be considered to also include description and/or disclosure of the amino acid sequence encoded by the nucleotide sequence. Similarly, description and/or disclosure of a polypeptide amino acid sequence herein should be considered to also include description and/or disclosure of all possible nucleotide sequences that can encode the amino acid sequence.
Finally, nucleic acid and amino acid sequence information for the loci and biomarkers of the present invention (e.g., biomarkers listed in Tables 2-6) are well known in the art and readily available on publicly available databases, such as the National Center for
Biotechnology Information (NCBI). For example, exemplary nucleic acid and amino acid sequences derived from publicly available sequence databases are provided below.
III. Sample Collection, Preparation and Separation
In some embodiments, biomarker amount and/or activity measurement(s) in a sample from a subject is compared to a predetermined control (standard) sample. The control sample can be from the same subject or from a different subject. The control sample can be a combination of samples from several different subjects. In some embodiments, the biomarker amount and/or activity measurement(s) from a subject is compared to a pre-determined level. This pre-determined level is typically obtained from normal samples. As described herein, a "pre-determined" biomarker amount and/or activity measurement(s) may be a biomarker amount and/or activity measurement(s) used to, by way of example only, evaluate a subject that may be selected for treatment, evaluate a response to a treatment. A pre-determined biomarker amount and/or activity measurement(s) may be determined in populations of patients with or without a condition. The pre-determined biomarker amount and/or activity measurement(s) can be a single number, equally applicable to every patient, or the pre- determined biomarker amount and/or activity measurement(s) can vary according to specific subpopulations of patients. Age, weight, height, and other factors of a subject may affect the pre-determined biomarker amount and/or activity measurement(s) of the individual.
Furthermore, the pre-determined biomarker amount and/or activity can be determined for each subject individually. In one embodiment, the amounts determined and/or compared in a method described herein are based on absolute measurements. In another embodiment, the amounts determined and/or compared in a method described herein are based on relative measurements, such as ratios (e.g., expression and/or activity of biomarkers to that of wild type biomarkers and expression and/or activity of a biomarker of interest normalized to that of a housekeeping gene).
The pre-determined biomarker amount and/or activity measurement(s) can be any suitable standard. For example, the pre-determined biomarker amount and/or activity measurement(s) can be obtained from the same or a different human for whom a patient selection is being assessed. In one embodiment, the pre-determined biomarker amount and/or activity measurement(s) can be obtained from a previous assessment of the same patient. In such a manner, the progress of the selection of the patient can be monitored over time. In addition, the control can be obtained from an assessment of another human or multiple humans, e.g., selected groups of humans, if the subject is a human. In such a manner, the extent of the selection of the human for whom selection is being assessed can be compared to suitable other humans, e.g., other humans who are in a similar situation to the human of interest, such as those suffering from similar or the same condition(s) and/or of the same ethnic group.
In some embodiments of the present invention, the change of biomarker amount and/or activity measurement(s) from the pre-determined level is about 0.5 fold, about 1.0 fold, about 1.5 fold, about 2.0 fold, about 2.5 fold, about 3.0 fold, about 3.5 fold, about 4.0 fold, about 4.5 fold, or about 5.0 fold or greater. In some embodiments, the fold change is less than about 1, less than about 5, less than about 10, less than about 20, less than about 30, less than about 40, or less than about 50. In other embodiments, the fold change in biomarker amount and/or activity measurement(s) compared to a predetermined level is more than about 1 , more than about 5, more than about 10, more than about 20, more than about 30, more than about 40, or more than about 50.
Biological samples can be collected from a variety of sources from a patient including a body fluid sample, cell sample, or a tissue sample comprising nucleic acids and/or proteins. "Body fluids" refer to fluids that are excreted or secreted from the body as well as fluids that are normally not (e.g. , bronchoalevolar lavage fluid, amniotic fluid, aqueous humor, bile, blood and blood plasma, cerebrospinal fluid, cerumen and earwax, cowper's fluid or pre- ejaculatory fluid, chyle, chyme, stool, female ejaculate, interstitial fluid, intracellular fluid, lymph, menses, breast milk, mucus, pleural fluid, pus, saliva, sebum, semen, serum, sweat, synovial fluid, tears, urine, vaginal lubrication, vitreous humor, vomit). In an embodiment, the subject and/or control sample is selected from the group consisting of cells, cell lines, histological slides, paraffin embedded tissues, biopsies, whole blood, nipple aspirate, serum, plasma, buccal scrape, saliva, cerebrospinal fluid, urine, stool, and bone marrow. In one embodiment, the sample is blood, serum, plasma, or urine. In another embodiment, the sample is serum.
The samples can be collected from individuals repeatedly over a longitudinal period of time (e.g., once or more on the order of days, weeks, months, annually, biannually, etc.). Obtaining numerous samples from an individual over a period of time can be used to verify results from earlier detections and/or to identify an alteration in biological pattern as a result of, for example, disease progression, drug treatment, etc. For example, subject samples can be taken and monitored every month, every two months, or combinations of one, two, or three month intervals according to the invention. In addition, the biomarker amount and/or activity measurements of the subject obtained over time can be conveniently compared with each other, as well as with those of normal controls during the monitoring period, thereby providing the subject's own values, as an internal, or personal, control for long-term monitoring.
Sample preparation and separation can involve any of the procedures, depending on the type of sample collected and/or analysis of biomarker measurement(s). Such procedures include, by way of example only, concentration, dilution, adjustment of pH, removal of high abundance polypeptides (e.g., albumin, gamma globulin, and transferrin, etc.), addition of preservatives and calibrants, addition of protease inhibitors, addition of denaturants, desalting of samples, concentration of sample proteins, extraction and purification of lipids.
The sample preparation can also isolate molecules that are bound in non-covalent complexes to other protein (e.g., carrier proteins). This process may isolate those molecules bound to a specific carrier protein (e.g., albumin), or use a more general process, such as the release of bound molecules from all carrier proteins via protein denaturation, for example using an acid, followed by removal of the carrier proteins.
Removal of undesired proteins (e.g. , high abundance, uninformative, or undetectable proteins) from a sample can be achieved using high affinity reagents, high molecular weight filters, ultracentrifugation and/or electrodialysis. High affinity reagents include antibodies or other reagents (e.g., aptamers) that selectively bind to high abundance proteins. Sample preparation could also include ion exchange chromatography, metal ion affinity
chromatography, gel filtration, hydrophobic chromatography, chromatofocusing, adsorption chromatography, isoelectric focusing and related techniques. Molecular weight filters include membranes that separate molecules on the basis of size and molecular weight. Such filters may further employ reverse osmosis, nanofiltration, ultrafiltration and microfiltration.
Ultracentrifugation is a method for removing undesired polypeptides from a sample. Ultracentrifugation is the centrifugation of a sample at about 15,000-60,000 rpm while monitoring with an optical system the sedimentation (or lack thereof) of particles.
Electrodialysis is a procedure which uses an electromembrane or semipermable membrane in a process in which ions are transported through semi-permeable membranes from one solution to another under the influence of a potential gradient. Since the membranes used in electrodialysis may have the ability to selectively transport ions having positive or negative charge, reject ions of the opposite charge, or to allow species to migrate through a
semipermable membrane based on size and charge, it renders electrodialysis useful for concentration, removal, or separation of electrolytes.
Separation and purification in the present invention may include any procedure known in the art, such as capillary electrophoresis (e.g. , in capillary or on-chip) or chromatography (e.g., in capillary, column or on a chip). Electrophoresis is a method which can be used to separate ionic molecules under the influence of an electric field. Electrophoresis can be conducted in a gel, capillary, or in a microchannel on a chip. Examples of gels used for electrophoresis include starch, acrylamide, polyethylene oxides, agarose, or combinations thereof. A gel can be modified by its cross-linking, addition of detergents, or denaturants, immobilization of enzymes or antibodies (affinity electrophoresis) or substrates (zymography) and incorporation of a pH gradient. Examples of capillaries used for electrophoresis include capillaries that interface with an electrospray.
Capillary electrophoresis (CE) is preferred for separating complex hydrophilic molecules and highly charged solutes. CE technology can also be implemented on microfluidic chips. Depending on the types of capillary and buffers used, CE can be further segmented into separation techniques such as capillary zone electrophoresis (CZE), capillary isoelectric focusing (CIEF), capillary isotachophoresis (cITP) and capillary
electrochromatography (CEC). An embodiment to couple CE techniques to electrospray ionization involves the use of volatile solutions, for example, aqueous mixtures containing a volatile acid and/or base and an organic such as an alcohol or acetonitrile.
Capillary isotachophoresis (cITP) is a technique in which the analytes move through the capillary at a constant speed but are nevertheless separated by their respective mobilities. Capillary zone electrophoresis (CZE), also known as free-solution CE (FSCE), is based on differences in the electrophoretic mobility of the species, determined by the charge on the molecule, and the factional resistance the molecule encounters during migration which is often directly proportional to the size of the molecule. Capillary isoelectric focusing (CIEF) allows weakly-ionizable amphoteric molecules, to be separated by electrophoresis in a pH gradient. CEC is a hybrid technique between traditional high performance liquid
chromatography (HPLC) and CE.
Separation and purification techniques used in the present invention include any chromatography procedures known in the art. Chromatography can be based on the differential adsorption and elution of certain analytes or partitioning of analytes between mobile and stationary phases. Different examples of chromatography include, but not limited to, liquid chromatography (LC), gas chromatography (GC), high performance liquid chromatography (HPLC), etc.
IV. Biomarker Nucleic Acids and Polypeptides
One aspect of the invention pertains to the use of isolated nucleic acid molecules that correspond to biomarker nucleic acids that encode a biomarker polypeptide or a portion of such a polypeptide. As used herein, the term "nucleic acid molecule" is intended to include DNA molecules (e.g., cDNA or genomic DNA) and RNA molecules (e.g., mRNA) and analogs of the DNA or RNA generated using nucleotide analogs. The nucleic acid molecule can be single-stranded or double-stranded, but preferably is double-stranded DNA. An "isolated" nucleic acid molecule is one which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid molecule. Preferably, an "isolated" nucleic acid molecule is free of sequences (preferably protein-encoding sequences) which naturally flank the nucleic acid (i.e., sequences located at the 5' and 3' ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived. For example, in various embodiments, the isolated nucleic acid molecule can contain less than about 5 kB, 4 kB, 3 kB, 2 kB, 1 kB, 0.5 kB or 0.1 kB of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived. Moreover, an "isolated" nucleic acid molecule, such as a cDNA molecule, can be substantially free of other cellular material or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized.
A biomarker nucleic acid molecule of the present invention can be isolated using standard molecular biology techniques and the sequence information in the database records described herein. Using all or a portion of such nucleic acid sequences, nucleic acid molecules of the invention can be isolated using standard hybridization and cloning techniques (e.g., as described in Sambrook et ah, ed., Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989).
A nucleic acid molecule of the invention can be amplified using cDNA, mRNA, or genomic DNA as a template and appropriate oligonucleotide primers according to standard PCR amplification techniques. The nucleic acid molecules so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis. Furthermore,
oligonucleotides corresponding to all or a portion of a nucleic acid molecule of the invention can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
Moreover, a nucleic acid molecule of the invention can comprise only a portion of a nucleic acid sequence, wherein the full length nucleic acid sequence comprises a marker of the invention or which encodes a polypeptide corresponding to a marker of the invention. Such nucleic acid molecules can be used, for example, as a probe or primer. The
probe/primer typically is used as one or more substantially purified oligonucleotides. The oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 7, preferably about 15, more preferably about 25, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, or 400 or more consecutive nucleotides of a biomarker nucleic acid sequence. Probes based on the sequence of a biomarker nucleic acid molecule can be used to detect transcripts or genomic sequences corresponding to one or more markers of the invention. The probe comprises a label group attached thereto, e.g., a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor.
A biomarker nucleic acid molecules that differ, due to degeneracy of the genetic code, from the nucleotide sequence of nucleic acid molecules encoding a protein which corresponds to the biomarker, and thus encode the same protein, are also contemplated.
In addition, it will be appreciated by those skilled in the art that DNA sequence polymorphisms that lead to changes in the amino acid sequence can exist within a population (e.g., the human population). Such genetic polymorphisms can exist among individuals within a population due to natural allelic variation. An allele is one of a group of genes which occur alternatively at a given genetic locus. In addition, it will be appreciated that DNA polymorphisms that affect RNA expression levels can also exist that may affect the overall expression level of that gene (e.g., by affecting regulation or degradation).
The term "allele," which is used interchangeably herein with "allelic variant," refers to alternative forms of a gene or portions thereof. Alleles occupy the same locus or position on homologous chromosomes. When a subject has two identical alleles of a gene, the subject is said to be homozygous for the gene or allele. When a subject has two different alleles of a gene, the subject is said to be heterozygous for the gene or allele. For example, biomarker alleles can differ from each other in a single nucleotide, or several nucleotides, and can include substitutions, deletions, and insertions of nucleotides. An allele of a gene can also be a form of a gene containing one or more mutations.
The term "allelic variant of a polymorphic region of gene" or "allelic variant", used interchangeably herein, refers to an alternative form of a gene having one of several possible nucleotide sequences found in that region of the gene in the population. As used herein, allelic variant is meant to encompass functional allelic variants, non-functional allelic variants, SNPs, mutations and polymorphisms.
The term "single nucleotide polymorphism" (SNP) refers to a polymorphic site occupied by a single nucleotide, which is the site of variation between allelic sequences. The site is usually preceded by and followed by highly conserved sequences of the allele (e.g. , sequences that vary in less than 1/100 or 1/1000 members of a population). A SNP usually arises due to substitution of one nucleotide for another at the polymorphic site. SNPs can also arise from a deletion of a nucleotide or an insertion of a nucleotide relative to a reference allele. Typically the polymorphic site is occupied by a base other than the reference base. For example, where the reference allele contains the base "T" (thymidine) at the polymorphic site, the altered allele can contain a "C" (cytidine), "G" (guanine), or "A" (adenine) at the polymorphic site. SNP's may occur in protein-coding nucleic acid sequences, in which case they may give rise to a defective or otherwise variant protein, or genetic disease. Such a SNP may alter the coding sequence of the gene and therefore specify another amino acid (a "missense" SNP) or a SNP may introduce a stop codon (a "nonsense" SNP). When a SNP does not alter the amino acid sequence of a protein, the SNP is called "silent." SNP's may also occur in noncoding regions of the nucleotide sequence. This may result in defective protein expression, e.g., as a result of alternative spicing, or it may have no effect on the function of the protein.
As used herein, the terms "gene" and "recombinant gene" refer to nucleic acid molecules comprising an open reading frame encoding a polypeptide corresponding to a marker of the invention. Such natural allelic variations can typically result in 1-5% variance in the nucleotide sequence of a given gene. Alternative alleles can be identified by
sequencing the gene of interest in a number of different individuals. This can be readily carried out by using hybridization probes to identify the same genetic locus in a variety of individuals. Any and all such nucleotide variations and resulting amino acid polymorphisms or variations that are the result of natural allelic variation and that do not alter the functional activity are intended to be within the scope of the invention.
In another embodiment, a biomarker nucleic acid molecule is at least 7, 15, 20, 25, 30, 40, 60, 80, 100, 150, 200, 250, 300, 350, 400, 450, 550, 650, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2200, 2400, 2600, 2800, 3000, 3500, 4000, 4500, or more nucleotides in length and hybridizes under stringent conditions to a nucleic acid molecule corresponding to a marker of the invention or to a nucleic acid molecule encoding a protein corresponding to a marker of the invention. As used herein, the term "hybridizes under stringent conditions" is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 60% (65%>, 70%>, 75%, 80%, preferably 85%) identical to each other typically remain hybridized to each other. Such stringent conditions are known to those skilled in the art and can be found in sections 6.3.1-6.3.6 of Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989). A preferred, non-limiting example of stringent hybridization conditions are hybridization in 6X sodium chloride/sodium citrate (SSC) at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 50- 65°C.
In addition to naturally-occurring allelic variants of a nucleic acid molecule of the invention that can exist in the population, the skilled artisan will further appreciate that sequence changes can be introduced by mutation thereby leading to changes in the amino acid sequence of the encoded protein, without altering the biological activity of the protein encoded thereby. For example, one can make nucleotide substitutions leading to amino acid substitutions at "non-essential" amino acid residues. A "non-essential" amino acid residue is a residue that can be altered from the wild-type sequence without altering the biological activity, whereas an "essential" amino acid residue is required for biological activity. For example, amino acid residues that are not conserved or only semi-conserved among homologs of various species may be non-essential for activity and thus would be likely targets for alteration. Alternatively, amino acid residues that are conserved among the homologs of various species (e.g. , murine and human) may be essential for activity and thus would not be likely targets for alteration.
Accordingly, another aspect of the invention pertains to nucleic acid molecules encoding a polypeptide of the invention that contain changes in amino acid residues that are not essential for activity. Such polypeptides differ in amino acid sequence from the naturally- occurring proteins which correspond to the markers of the invention, yet retain biological activity. In one embodiment, a biomarker protein has an amino acid sequence that is at least about 40% identical, 50%, 60%, 70%, 75%, 80%, 83%, 85%, 87.5%, 90%, 91%, 92%, 93%, 94%), 95%), 96%), 97%), 98%>, 99% or identical to the amino acid sequence of a biomarker protein described herein.
An isolated nucleic acid molecule encoding a variant protein can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of nucleic acids of the invention, such that one or more amino acid residue substitutions, additions, or deletions are introduced into the encoded protein. Mutations can be introduced by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis. Preferably, conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues. A "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), non-polar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Alternatively, mutations can be introduced randomly along all or part of the coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for biological activity to identify mutants that retain activity. Following mutagenesis, the encoded protein can be expressed recombinantly and the activity of the protein can be determined.
In some embodiments, the present invention further contemplates the use of anti- biomarker antisense nucleic acid molecules, i.e., molecules which are complementary to a sense nucleic acid of the invention, e.g., complementary to the coding strand of a double- stranded cDNA molecule corresponding to a marker of the invention or complementary to an mR A sequence corresponding to a marker of the invention. Accordingly, an antisense nucleic acid molecule of the invention can hydrogen bond to (i.e. anneal with) a sense nucleic acid of the invention. The antisense nucleic acid can be complementary to an entire coding strand, or to only a portion thereof, e.g. , all or part of the protein coding region (or open reading frame). An antisense nucleic acid molecule can also be antisense to all or part of a non-coding region of the coding strand of a nucleotide sequence encoding a polypeptide of the invention. The non-coding regions ("5' and 3' untranslated regions") are the 5' and 3' sequences which flank the coding region and are not translated into amino acids.
An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 or more nucleotides in length. An antisense nucleic acid can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used. Examples of modified nucleotides which can be used to generate the antisense nucleic acid include 5-fluorouracil, 5- bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5- (carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5- carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6- isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2- methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7- methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D- mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6- isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2- thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5- oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3- N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine. Alternatively, the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been sub-cloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).
The antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mR A and/or genomic DNA encoding a polypeptide corresponding to a selected marker of the invention to thereby inhibit expression of the marker, e.g., by inhibiting transcription and/or translation. The hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule which binds to DNA duplexes, through specific interactions in the major groove of the double helix. Examples of a route of administration of antisense nucleic acid molecules of the invention includes direct injection at a tissue site or infusion of the antisense nucleic acid into a blood- or bone marrow- associated body fluid. Alternatively, antisense nucleic acid molecules can be modified to target selected cells and then administered systemically. For example, for systemic administration, antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface, e.g., by linking the antisense nucleic acid molecules to peptides or antibodies which bind to cell surface receptors or antigens. The antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient intracellular concentrations of the antisense molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II or pol III promoter are preferred.
In various embodiments, the nucleic acid molecules of the present invention can be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g., the stability, hybridization, or solubility of the molecule. For example, the deoxyribose phosphate backbone of the nucleic acid molecules can be modified to generate peptide nucleic acid molecules (see Hyrup et ah, 1996, Bioorganic & Medicinal Chemistry 4(1): 5-23). As used herein, the terms "peptide nucleic acids" or "PNAs" refer to nucleic acid mimics, e.g., DNA mimics, in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained. The neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength. The synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described in Hyrup et al. (1996), supra; Perry-O'Keefe et al. (1996) Proc. Natl. Acad. Sci. USA 93: 14670-675.
PNAs can be used in therapeutic and diagnostic applications. For example, PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, e.g., inducing transcription or translation arrest or inhibiting replication. PNAs can also be used, e.g. , in the analysis of single base pair mutations in a gene by, e.g. , PNA directed PCR clamping; as artificial restriction enzymes when used in combination with other enzymes, e.g., SI nucleases (Hyrup (1996), supra; or as probes or primers for DNA sequence and hybridization (Hyrup, 1996, supra; Perry-O'Keefe et al, 1996, Proc. Natl. Acad. Sci. USA 93: 14670-675).
In another embodiment, PNAs can be modified, e.g. , to enhance their stability or cellular uptake, by attaching lipophilic or other helper groups to PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art. For example, PNA-DNA chimeras can be generated which can combine the advantageous properties of PNA and DNA. Such chimeras allow DNA recognition enzymes, e.g., RNASE H and DNA polymerases, to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity. PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleobases, and orientation (Hyrup, 1996, supra). The synthesis of PNA- DNA chimeras can be performed as described in Hyrup (1996), supra, and Finn et al. (1996) Nucleic Acids Res. 24(17):3357-63. For example, a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry and modified nucleoside analogs. Compounds such as 5'-(4-methoxytrityl)amino-5'-deoxy-thymidine phosphoramidite can be used as a link between the PNA and the 5' end of DNA (Mag et al, 1989, Nucleic Acids Res. 17:5973-88). PNA monomers are then coupled in a step-wise manner to produce a chimeric molecule with a 5' PNA segment and a 3' DNA segment (Finn et al, 1996, Nucleic Acids Res. 24(17):3357-63). Alternatively, chimeric molecules can be synthesized with a 5' DNA segment and a 3' PNA segment (Peterser et al, 1975, Bioorganic Med. Chem. Lett. 5: 1119- 11124).
An "isolated" or "purified" protein or biologically active portion thereof is
substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the protein is derived, or substantially free of chemical precursors or other chemicals when chemically synthesized. The language "substantially free of cellular material" includes preparations of protein in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly produced. Thus, protein that is substantially free of cellular material includes preparations of protein having less than about 30%, 20%, 10%>, or 5% (by dry weight) of heterologous protein (also referred to herein as a "contaminating protein"). When the protein or biologically active portion thereof is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%>, 10%>, or 5% of the volume of the protein preparation. When the protein is produced by chemical synthesis, it is preferably substantially free of chemical precursors or other chemicals, i.e., it is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein. Accordingly such preparations of the protein have less than about 30%>, 20%>, 10%>, 5% (by dry weight) of chemical precursors or compounds other than the polypeptide of interest.
Biologically active portions of a biomarker polypeptide include polypeptides comprising amino acid sequences sufficiently identical to or derived from a biomarker protein amino acid sequence described herein, but which includes fewer amino acids than the full length protein, and exhibit at least one activity of the corresponding full-length protein.
Typically, biologically active portions comprise a domain or motif with at least one activity of the corresponding protein. A biologically active portion of a protein of the invention can be a polypeptide which is, for example, 10, 25, 50, 100 or more amino acids in length. Moreover, other biologically active portions, in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the functional activities of the native form of a polypeptide of the invention.
Preferred polypeptides have an amino acid sequence of a biomarker protein encoded by a nucleic acid molecule described herein. Other useful proteins are substantially identical (e.g., at least about 40%, preferably 50%, 60%, 70%, 75%, 80%, 83%, 85%, 88%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) to one of these sequences and retain the functional activity of the protein of the corresponding naturally-occurring protein yet differ in amino acid sequence due to natural allelic variation or mutagenesis.
To determine the percent identity of two amino acid sequences or of two nucleic acids, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % identity = # of identical positions/total # of positions (e.g., overlapping positions) xlOO). In one embodiment the two sequences are the same length.
The determination of percent identity between two sequences can be accomplished using a mathematical algorithm. A preferred, non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul (1990) Proc. Natl. Acad. Sci. USA 87:2264-2268, modified as in Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5877. Such an algorithm is incorporated into the
NBLAST and XBLAST programs of Altschul, et al. (1990) J. Mol. Biol. 215:403-410.
BLAST nucleotide searches can be performed with the NBLAST program, score = 100, wordlength = 12 to obtain nucleotide sequences homologous to a nucleic acid molecules of the invention. BLAST protein searches can be performed with the XBLAST program, score = 50, wordlength = 3 to obtain amino acid sequences homologous to a protein molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25:3389-3402.
Alternatively, PSI-Blast can be used to perform an iterated search which detects distant relationships between molecules. When utilizing BLAST, Gapped BLAST, and PSI-Blast programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used. See http://www.ncbi.nlm.nih.gov. Another preferred, non- limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, (1988) Comput Appl Biosci, 4: 11-7. Such an algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a
PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used. Yet another useful algorithm for identifying regions of local sequence similarity and alignment is the FASTA algorithm as described in Pearson and Lipman (1988) Proc. Natl. Acad. Sci. USA 85:2444-2448. When using the FASTA algorithm for comparing nucleotide or amino acid sequences, a PAM120 weight residue table can, for example, be used with a k- tuple value of 2.
The percent identity between two sequences can be determined using techniques similar to those described above, with or without allowing gaps. In calculating percent identity, only exact matches are counted.
The invention also provides chimeric or fusion proteins corresponding to a biomarker protein. As used herein, a "chimeric protein" or "fusion protein" comprises all or part (preferably a biologically active part) of a polypeptide corresponding to a marker of the invention operably linked to a heterologous polypeptide (i.e., a polypeptide other than the polypeptide corresponding to the marker). Within the fusion protein, the term "operably linked" is intended to indicate that the polypeptide of the invention and the heterologous polypeptide are fused in- frame to each other. The heterologous polypeptide can be fused to the amino-terminus or the carboxyl-terminus of the polypeptide of the invention.
One useful fusion protein is a GST fusion protein in which a polypeptide
corresponding to a marker of the invention is fused to the carboxyl terminus of GST sequences. Such fusion proteins can facilitate the purification of a recombinant polypeptide of the invention.
In another embodiment, the fusion protein contains a heterologous signal sequence, immunoglobulin fusion protein, toxin, or other useful protein sequence. Chimeric and fusion proteins of the invention can be produced by standard recombinant DNA techniques. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and re-amplified to generate a chimeric gene sequence (see, e.g., Ausubel et ah, supra). Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide). A nucleic acid encoding a polypeptide of the invention can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the polypeptide of the invention.
A signal sequence can be used to facilitate secretion and isolation of the secreted protein or other proteins of interest. Signal sequences are typically characterized by a core of hydrophobic amino acids which are generally cleaved from the mature protein during secretion in one or more cleavage events. Such signal peptides contain processing sites that allow cleavage of the signal sequence from the mature proteins as they pass through the secretory pathway. Thus, the invention pertains to the described polypeptides having a signal sequence, as well as to polypeptides from which the signal sequence has been proteolytically cleaved (i.e., the cleavage products). In one embodiment, a nucleic acid sequence encoding a signal sequence can be operably linked in an expression vector to a protein of interest, such as a protein which is ordinarily not secreted or is otherwise difficult to isolate. The signal sequence directs secretion of the protein, such as from a eukaryotic host into which the expression vector is transformed, and the signal sequence is subsequently or concurrently cleaved. The protein can then be readily purified from the extracellular medium by art recognized methods. Alternatively, the signal sequence can be linked to the protein of interest using a sequence which facilitates purification, such as with a GST domain.
The present invention also pertains to variants of the biomarker polypeptides described herein. Such variants have an altered amino acid sequence which can function as either agonists (mimetics) or as antagonists. Variants can be generated by mutagenesis, e.g., discrete point mutation or truncation. An agonist can retain substantially the same, or a subset, of the biological activities of the naturally occurring form of the protein. An antagonist of a protein can inhibit one or more of the activities of the naturally occurring form of the protein by, for example, competitively binding to a downstream or upstream member of a cellular signaling cascade which includes the protein of interest. Thus, specific biological effects can be elicited by treatment with a variant of limited function. Treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein can have fewer side effects in a subject relative to treatment with the naturally occurring form of the protein.
Variants of a biomarker protein which function as either agonists (mimetics) or as antagonists can be identified by screening combinatorial libraries of mutants, e.g., truncation mutants, of the protein of the invention for agonist or antagonist activity. In one embodiment, a variegated library of variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library. A variegated library of variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential protein sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins {e.g., for phage display). There are a variety of methods which can be used to produce libraries of potential variants of the polypeptides of the invention from a degenerate oligonucleotide sequence.
Methods for synthesizing degenerate oligonucleotides are known in the art (see, e.g., Narang,
1983, Tetrahedron 39:3; Itakura et al, 1984, Annu. Rev. Biochem. 53:323; Itakura et al,
1984, Science 198: 1056; Ike et al, 1983 Nucleic Acid Res. 11 :477).
In addition, libraries of fragments of the coding sequence of a polypeptide
corresponding to a marker of the invention can be used to generate a variegated population of polypeptides for screening and subsequent selection of variants. For example, a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of the coding sequence of interest with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA to form double stranded DNA which can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with S 1 nuclease, and ligating the resulting fragment library into an expression vector. By this method, an expression library can be derived which encodes amino terminal and internal fragments of various sizes of the protein of interest.
Several techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA libraries for gene products having a selected property. The most widely used techniques, which are amenable to high throughput analysis, for screening large gene libraries typically include cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates isolation of the vector encoding the gene whose product was detected. Recursive ensemble mutagenesis (REM), a technique which enhances the frequency of functional mutants in the libraries, can be used in combination with the screening assays to identify variants of a protein of the invention (Arkin and Yourvan, 1992, Proc. Natl. Acad. Sci. USA 59:7811-7815; Delgrave et al, 1993, Protein Engineering 6(3):327- 331).
The production and use of biomarker nucleic acid and/or biomarker polypeptide molecules described herein can be facilitated by using standard recombinant techniques. In some embodiments, such techniques use vectors, preferably expression vectors, containing a nucleic acid encoding a biomarker polypeptide or a portion of such a polypeptide. As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated. Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced {e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors {e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors, namely expression vectors, are capable of directing the expression of genes to which they are operably linked. In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids (vectors). However, the present invention is intended to include such other forms of expression vectors, such as viral vectors {e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions. The recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell. This means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which is operably linked to the nucleic acid sequence to be expressed. Within a recombinant expression vector, "operably linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell). The term "regulatory sequence" is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, Methods in Enzymology: Gene Expression
Technology vol.185, Academic Press, San Diego, CA (1991). Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in many types of host cell and those which direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, and the like. The expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein.
The recombinant expression vectors for use in the invention can be designed for expression of a polypeptide corresponding to a marker of the invention in prokaryotic (e.g. , E. coli) or eukaryotic cells (e.g., insect cells {using baculovirus expression vectors}, yeast cells or mammalian cells). Suitable host cells are discussed further in Goeddel, supra.
Alternatively, the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
Expression of proteins in prokaryotes is most often carried out in E. coli with vectors containing constitutive or inducible promoters directing the expression of either fusion or non- fusion proteins. Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein. Such fusion vectors typically serve three purposes: 1) to increase expression of recombinant protein; 2) to increase the solubility of the recombinant protein; and 3) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification. Often, in fusion expression vectors, a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein. Such enzymes, and their cognate recognition sequences, include Factor Xa, thrombin and enterokinase. Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith and Johnson, 1988, Gene 67:31-40), pMAL (New England Biolabs, Beverly, MA) and pRIT5 (Pharmacia, Piscataway, NJ) which fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein.
Examples of suitable inducible non-fusion E. coli expression vectors include pTrc (Amann et al, 1988, Gene 69:301-315) and pET l id (Studier et al, p. 60-89, In Gene Expression Technology: Methods in Enzymology vol.185, Academic Press, San Diego, CA, 1991). Target biomarker nucleic acid expression from the pTrc vector relies on host RNA polymerase transcription from a hybrid trp-lac fusion promoter. Target biomarker nucleic acid expression from the pET l id vector relies on transcription from a T7 gnlO-lac fusion promoter mediated by a co-expressed viral RNA polymerase (T7 gnl). This viral polymerase is supplied by host strains BL21 (DE3) or HMS174(DE3) from a resident prophage harboring a T7 gnl gene under the transcriptional control of the lacUV 5 promoter.
In another embodiment, the expression vector is a yeast expression vector. Examples of vectors for expression in yeast S. cerevisiae include pYepSecl (Baldari et al, 1987, EMBO J. 6:229-234), pMFa (Kurjan and Herskowitz, 1982, Cell 30:933-943), pJRY88 (Schultz et al, 1987, Gene 54:113-123), pYES2 (Invitrogen Corporation, San Diego, CA), and pPicZ (Invitrogen Corp, San Diego, CA).
Alternatively, the expression vector is a baculovirus expression vector. Baculovirus vectors available for expression of proteins in cultured insect cells (e.g. , Sf 9 cells) include the pAc series (Smith et al, 1983, Mol. Cell Biol. 3:2156-2165) and the pVL series (Lucklow and Summers, 1989, Virology 170:31-39).
In yet another embodiment, a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed, 1987, Nature 329:840) and pMT2PC (Kaufman et al, 1987, EMBO J. 6: 187-195). When used in mammalian cells, the expression vector's control functions are often provided by viral regulatory elements. For example, commonly used promoters are derived from polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40. For other suitable expression systems for both prokaryotic and eukaryotic cells see chapters 16 and 17 of Sambrook et al. , supra.
The invention further provides a recombinant expression vector comprising a DNA molecule cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operably linked to a regulatory sequence in a manner which allows for expression (by transcription of the DNA molecule) of an R A molecule which is antisense to the mR A encoding a polypeptide of the invention. Regulatory sequences operably linked to a nucleic acid cloned in the antisense orientation can be chosen which direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen which direct constitutive, tissue-specific or cell type specific expression of antisense RNA. The antisense expression vector can be in the form of a recombinant plasmid, phagemid, or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced. For a discussion of the regulation of gene expression using antisense genes (see Weintraub et ah, 1986, Trends in Genetics, Vol. 1(1)).
Another aspect of the invention pertains to host cells into which a recombinant expression vector of the invention has been introduced. The terms "host cell" and
"recombinant host cell" are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
A host cell can be any prokaryotic {e.g., E. coli) or eukaryotic cell (e.g., insect cells, yeast or mammalian cells).
Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques. As used herein, the terms "transformation" and "transfection" are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid into a host cell, including calcium phosphate or calcium chloride co- precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. {supra), and other laboratory manuals.
For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate the foreign DNA into their genome. In order to identify and select these integrants, a gene that encodes a selectable marker {e.g., for resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Preferred selectable markers include those which confer resistance to drugs, such as G418, hygromycin and methotrexate. Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die).
V. Analyzing Biomarkers
Biomarker nucleic acids and/or biomarker polypeptides can be analyzed according to the methods described herein and techniques known to the skilled artisan to identify such genetic or expression alterations useful for the present invention including, but not limited to, 1) an alteration in the level of a biomarker transcript or polypeptide, 2) a deletion or addition of one or more nucleotides from a biomarker gene, 4) a substitution of one or more nucleotides of a biomarker gene, 5) aberrant modification of a biomarker gene, such as an expression regulatory region, 6) an alteration in the phosphorylation status or level of phosphorylation of a biomarker protein described herein, and the like,
a. Methods for Determining Levels of Expression
Biomarker expression may be assessed by any of a wide variety of well known methods for detecting expression of a transcribed molecule or protein. Non-limiting examples of such methods include immunological methods for detection of secreted, cell-surface, cytoplasmic, or nuclear proteins, protein purification methods, protein function or activity assays, nucleic acid hybridization methods, nucleic acid reverse transcription methods, and nucleic acid amplification methods.
In some embodiments, activity of a particular gene is characterized by a measure of gene transcript (e.g. m NA), by a measure of the quantity of translated protein, or by a measure of gene product activity. Marker expression can be monitored in a variety of ways, including by detecting mRNA levels, protein levels, or protein activity, any of which can be measured using standard techniques. Detection can involve quantification of the level of gene expression (e.g., genomic DNA, cDNA, mRNA, protein, or enzyme activity), or,
alternatively, can be a qualitative assessment of the level of gene expression, in particular in comparison with a control level. The type of level being detected will be clear from the context.
In another embodiment, detecting or determining expression levels of a biomarker and functionally similar homologs thereof, including a fragment or genetic alteration thereof (e.g., in regulatory or promoter regions thereof) comprises detecting or determining RNA levels for the marker of interest. In one embodiment, one or more cells from the subject to be tested are obtained and RNA is isolated from the cells. In a preferred embodiment, a sample of breast tissue cells is obtained from the subject. In one embodiment, RNA is obtained from a single cell. For example, a cell can be isolated from a tissue sample by laser capture microdissection (LCM). Using this technique, a cell can be isolated from a tissue section, including a stained tissue section, thereby assuring that the desired cell is isolated (see, e.g., Bonner et al. (1997) Science 278: 1481 ; Emmert- Buck et al. (1996) Science 274:998; Fend et al. (1999) Am. J. Path. 154: 61 and Murakami et al. (2000) Kidney Int. 58: 1346). For example, Murakami et al, supra, describe isolation of a cell from a previously immunostained tissue section.
It is also be possible to obtain cells from a subject and culture the cells in vitro, such as to obtain a larger population of cells from which RNA can be extracted. Methods for establishing cultures of non-transformed cells, i.e., primary cell cultures, are known in the art.
When isolating RNA from tissue samples or cells from individuals, it may be important to prevent any further changes in gene expression after the tissue or cells has been removed from the subject. Changes in expression levels are known to change rapidly following perturbations, e.g., heat shock or activation with lipopolysaccharide (LPS) or other reagents. In addition, the RNA in the tissue and cells may quickly become degraded.
Accordingly, in a preferred embodiment, the tissue or cells obtained from a subject is snap frozen as soon as possible.
RNA can be extracted from the tissue sample by a variety of methods, e.g., the guanidium thiocyanate lysis followed by CsCl centrifugation (Chirgwin et al, 1979,
Biochemistry 18:5294-5299). RNA from single cells can be obtained as described in methods for preparing cDNA libraries from single cells, such as those described in Dulac, C. (1998) Curr. Top. Dev. Biol. 36, 245 and Jena et al. (1996) J. Immunol. Methods 190: 199. Care to avoid RNA degradation must be taken, e.g., by inclusion of RNAsin.
The RNA sample can then be enriched in particular species. In one embodiment, poly(A)+ RNA is isolated from the RNA sample. In general, such purification takes advantage of the poly-A tails on mRNA. In particular and as noted above, poly-T
oligonucleotides may be immobilized within on a solid support to serve as affinity ligands for mRNA. Kits for this purpose are commercially available, e.g. , the MessageMaker kit (Life Technologies, Grand Island, NY).
In an embodiment, the RNA population is enriched in marker sequences. Enrichment can be undertaken, e.g., by primer-specific cDNA synthesis, or multiple rounds of linear amplification based on cDNA synthesis and template-directed in vitro transcription (see, e.g. , Wang et al. (1989) PNAS 86, 9717; Dulac et al., supra, and Jena et al., supra). The population of RNA, enriched or not in particular species or sequences, can further be amplified. As defined herein, an "amplification process" is designed to strengthen, increase, or augment a molecule within the RNA. For example, where RNA is mRNA, an amplification process such as RT-PCR can be utilized to amplify the mRNA, such that a signal is detectable or detection is enhanced. Such an amplification process is beneficial particularly when the biological, tissue, or tumor sample is of a small size or volume.
Various amplification and detection methods can be used. For example, it is within the scope of the present invention to reverse transcribe mRNA into cDNA followed by polymerase chain reaction (RT-PCR); or, to use a single enzyme for both steps as described in U.S. Pat. No. 5,322,770, or reverse transcribe mRNA into cDNA followed by symmetric gap ligase chain reaction (RT-AGLCR) as described by R. L. Marshall, et al., PCR Methods and Applications 4: 80-84 (1994). Real time PCR may also be used.
Other known amplification methods which can be utilized herein include but are not limited to the so-called "NASBA" or "3SR" technique described in PNAS USA 87: 1874- 1878 (1990) and also described in Nature 350 (No. 6313): 91-92 (1991); Q-beta amplification as described in published European Patent Application (EPA) No. 4544610; strand displacement amplification (as described in G. T. Walker et al., Clin. Chem. 42: 9-13 (1996) and European Patent Application No. 684315; target mediated amplification, as described by PCT Publication W09322461; PCR; ligase chain reaction (LCR) {see, e.g., Wu and Wallace, Genomics 4, 560 (1989), Landegren et al., Science 241, 1077 (1988)); self-sustained sequence replication (SSR) {see, e.g., Guatelli et al, Proc. Nat. Acad. Sci. USA, 87, 1874 (1990)); and transcription amplification {see, e.g., Kwoh et al., Proc. Natl. Acad. Sci. USA 86, 1173 (1989)).
Many techniques are known in the state of the art for determining absolute and relative levels of gene expression, commonly used techniques suitable for use in the present invention include Northern analysis, RNase protection assays (RPA), microarrays and PCR-based techniques, such as quantitative PCR and differential display PCR. For example, Northern blotting involves running a preparation of RNA on a denaturing agarose gel, and transferring it to a suitable support, such as activated cellulose, nitrocellulose or glass or nylon
membranes. Radiolabeled cDNA or RNA is then hybridized to the preparation, washed and analyzed by autoradiography.
In situ hybridization visualization may also be employed, wherein a radioactively labeled antisense RNA probe is hybridized with a thin section of a biopsy sample, washed, cleaved with RNase and exposed to a sensitive emulsion for autoradiography. The samples may be stained with hematoxylin to demonstrate the histological composition of the sample, and dark field imaging with a suitable light filter shows the developed emulsion. Nonradioactive labels such as digoxigenin may also be used.
Alternatively, mRNA expression can be detected on a DNA array, chip or a microarray. Labeled nucleic acids of a test sample obtained from a subject may be hybridized to a solid surface comprising biomarker DNA. Positive hybridization signal is obtained with the sample containing biomarker transcripts. Methods of preparing DNA arrays and their use are well known in the art (see, e.g., U.S. Pat. Nos: 6,618,6796; 6,379,897; 6,664,377;
6,451,536; 548,257; U.S. 20030157485 and Schena et al. (1995) Science 20, 467-470;
Gerhold et al. (1999) Trends In Biochem. Sci. 24, 168-173; and Lennon et al. (2000) Drug Discovery Today 5, 59-65, which are herein incorporated by reference in their entirety). Serial Analysis of Gene Expression (SAGE) can also be performed (See for example U.S. Patent Application 20030215858).
To monitor mRNA levels, for example, mRNA is extracted from the biological sample to be tested, reverse transcribed, and fluorescently-labeled cDNA probes are generated. The microarrays capable of hybridizing to marker cDNA are then probed with the labeled cDNA probes, the slides scanned and fluorescence intensity measured. This intensity correlates with the hybridization intensity and expression levels.
Types of probes that can be used in the methods described herein include cDNA, riboprobes, synthetic oligonucleotides and genomic probes. The type of probe used will generally be dictated by the particular situation, such as riboprobes for in situ hybridization, and cDNA for Northern blotting, for example. In one embodiment, the probe is directed to nucleotide regions unique to the RNA. The probes may be as short as is required to differentially recognize marker mRNA transcripts, and may be as short as, for example, 15 bases; however, probes of at least 17, 18, 19 or 20 or more bases can be used. In one embodiment, the primers and probes hybridize specifically under stringent conditions to a DNA fragment having the nucleotide sequence corresponding to the marker. As herein used, the term "stringent conditions" means hybridization will occur only if there is at least 95% identity in nucleotide sequences. In another embodiment, hybridization under "stringent conditions" occurs when there is at least 97% identity between the sequences.
The form of labeling of the probes may be any that is appropriate, such as the use of radioisotopes, for example, 32P and 35S. Labeling with radioisotopes may be achieved, whether the probe is synthesized chemically or biologically, by the use of suitably labeled bases. In one embodiment, the biological sample contains polypeptide molecules from the test subject. Alternatively, the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject.
In another embodiment, the methods further involve obtaining a control biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting marker polypeptide, mRNA, genomic DNA, or fragments thereof, such that the presence of the marker polypeptide, mRNA, genomic DNA, or fragments thereof, is detected in the biological sample, and comparing the presence of the marker polypeptide, mRNA, genomic DNA, or fragments thereof, in the control sample with the presence of the marker polypeptide, mRNA, genomic DNA, or fragments thereof in the test sample. b. Methods for Determining Levels of Protein Expression and Levels of Protein Activity
The activity or level of a biomarker protein can be detected and/or quantified by detecting or quantifying the expressed polypeptide. The polypeptide can be detected and quantified by any of a number of means well known to those of skill in the art. Any method known in the art for detecting polypeptides can be used. Such methods include, but are not limited to, immunodiffusion, Immunoelectrophoresis, radioimmunoassay (RIA), enzyme- linked immunosorbent assays (ELISAs), immunofluorescent assays, Western blotting, binder- ligand assays, immunohistochemical techniques, agglutination, complement assays, high performance liquid chromatography (HPLC), thin layer chromatography (TLC),
hyperdiffusion chromatography, and the like (e.g., Basic and Clinical Immunology, Sites and Terr, eds., Appleton and Lange, Norwalk, Conn, pp 217-262, 1991 which is incorporated by reference). Preferred are binder-ligand immunoassay methods including reacting antibodies with an epitope or epitopes and competitively displacing a labeled polypeptide or derivative thereof.
For example, ELISA and RIA procedures may be conducted such that a desired biomarker protein standard is labeled (with a radioisotope such as 125I or 35S, or an assayable enzyme, such as horseradish peroxidase or alkaline phosphatase), and, together with the unlabelled sample, brought into contact with the corresponding antibody, whereon a second antibody is used to bind the first, and radioactivity or the immobilized enzyme assayed (competitive assay). Alternatively, the biomarker protein in the sample is allowed to react with the corresponding immobilized antibody, radioisotope- or enzyme-labeled anti- biomarker proteinantibody is allowed to react with the system, and radioactivity or the enzyme assayed (ELISA-sandwich assay). Other conventional methods may also be employed as suitable.
The above techniques may be conducted essentially as a "one-step" or "two-step" assay. A "one-step" assay involves contacting antigen with immobilized antibody and, without washing, contacting the mixture with labeled antibody. A "two-step" assay involves washing before contacting, the mixture with labeled antibody. Other conventional methods may also be employed as suitable.
In one embodiment, a method for measuring biomarker protein levels comprises the steps of: contacting a biological specimen with an antibody or variant (e.g., fragment) thereof which selectively binds the biomarker protein, and detecting whether said antibody or variant thereof is bound to said sample and thereby measuring the levels of the biomarker protein.
Enzymatic and radiolabeling of biomarker protein and/or the antibodies may be effected by conventional means. Such means will generally include covalent linking of the enzyme to the antigen or the antibody in question, such as by glutaraldehyde, specifically so as not to adversely affect the activity of the enzyme, by which is meant that the enzyme must still be capable of interacting with its substrate, although it is not necessary for all of the enzyme to be active, provided that enough remains active to permit the assay to be effected.
Indeed, some techniques for binding enzyme are non-specific (such as using formaldehyde), and will only yield a proportion of active enzyme.
It is usually desirable to immobilize one component of the assay system on a support, thereby allowing other components of the system to be brought into contact with the component and readily removed without laborious and time-consuming labor. It is possible for a second phase to be immobilized away from the first, but one phase is usually sufficient.
It is possible to immobilize the enzyme itself on a support, but if solid-phase enzyme is required, then this is generally best achieved by binding to antibody and affixing the antibody to a support, models and systems for which are well-known in the art. Simple polyethylene may provide a suitable support.
Enzymes employable for labeling are not particularly limited, but may be selected from the members of the oxidase group, for example. These catalyze production of hydrogen peroxide by reaction with their substrates, and glucose oxidase is often used for its good stability, ease of availability and cheapness, as well as the ready availability of its substrate
(glucose). Activity of the oxidase may be assayed by measuring the concentration of hydrogen peroxide formed after reaction of the enzyme-labeled antibody with the substrate under controlled conditions well-known in the art. Other techniques may be used to detect biomarker protein according to a practitioner's preference based upon the present disclosure. One such technique is Western blotting (Towbin et at., Proc. Nat. Acad. Sci. 76:4350 (1979)), wherein a suitably treated sample is run on an SDS-PAGE gel before being transferred to a solid support, such as a nitrocellulose filter. Anti-biomarker protein antibodies (unlabeled) are then brought into contact with the support and assayed by a secondary immunological reagent, such as labeled protein A or antiimmunoglobulin (suitable labels including 125I, horseradish peroxidase and alkaline phosphatase). Chromatographic detection may also be used.
Immunohistochemistry may be used to detect expression of biomarker protein, e.g., in a biopsy sample. A suitable antibody is brought into contact with, for example, a thin layer of cells, washed, and then contacted with a second, labeled antibody. Labeling may be by fluorescent markers, enzymes, such as peroxidase, avidin, or radiolabelling. The assay is scored visually, using microscopy.
Anti- biomarker protein antibodies, such as intrabodies, may also be used for imaging purposes, for example, to detect the presence of biomarker protein in cells and tissues of a subject. Suitable labels include radioisotopes, iodine (125I, 121I), carbon (14C), sulphur (35S), tritium ( 3 H), indium ( 112 In), and technetium ( 99 mTc), fluorescent labels, such as fluorescein and rhodamine, and biotin.
For in vivo imaging purposes, antibodies are not detectable, as such, from outside the body, and so must be labeled, or otherwise modified, to permit detection. Markers for this purpose may be any that do not substantially interfere with the antibody binding, but which allow external detection. Suitable markers may include those that may be detected by X- radiography, NMR or MRI. For X-radiographic techniques, suitable markers include any radioisotope that emits detectable radiation but that is not overtly harmful to the subject, such as barium or cesium, for example. Suitable markers for NMR and MRI generally include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the antibody by suitable labeling of nutrients for the relevant hybridoma, for example.
The size of the subject, and the imaging system used, will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of technetium-99. The labeled antibody or antibody fragment will then preferentially accumulate at the location of cells which contain biomarker protein. The labeled antibody or antibody fragment can then be detected using known techniques. Antibodies that may be used to detect biomarker protein include any antibody, whether natural or synthetic, full length or a fragment thereof, monoclonal or polyclonal, that binds sufficiently strongly and specifically to the biomarker protein to be detected. An antibody may have a IQ of at most about 10"6M, 10"7M, 10"8M, 10"9M, 10"10M, 10"UM, 10"12M. The phrase "specifically binds" refers to binding of, for example, an antibody to an epitope or antigen or antigenic determinant in such a manner that binding can be displaced or competed with a second preparation of identical or similar epitope, antigen or antigenic determinant. An antibody may bind preferentially to the biomarker protein relative to other proteins, such as related proteins.
Antibodies are commercially available or may be prepared according to methods known in the art.
Antibodies and derivatives thereof that may be used encompass polyclonal or monoclonal antibodies, chimeric, human, humanized, primatized (CDR-grafted), veneered or single-chain antibodies as well as functional fragments, i.e., biomarker protein binding fragments, of antibodies. For example, antibody fragments capable of binding to a biomarker protein or portions thereof, including, but not limited to, Fv, Fab, Fab' and F(ab') 2 fragments can be used. Such fragments can be produced by enzymatic cleavage or by recombinant techniques. For example, papain or pepsin cleavage can generate Fab or F(ab') 2 fragments, respectively. Other proteases with the requisite substrate specificity can also be used to generate Fab or F(ab') 2 fragments. Antibodies can also be produced in a variety of truncated forms using antibody genes in which one or more stop codons have been introduced upstream of the natural stop site. For example, a chimeric gene encoding a F(ab') 2 heavy chain portion can be designed to include DNA sequences encoding the CH, domain and hinge region of the heavy chain.
Synthetic and engineered antibodies are described in, e.g., Cabilly et al, U.S. Pat. No.
4,816,567 Cabilly et al, European Patent No. 0,125,023 Bl; Boss et al, U.S. Pat. No.
4,816,397; Boss et al, European Patent No. 0,120,694 Bl; Neuberger, M. S. et al, WO 86/01533; Neuberger, M. S. et al, European Patent No. 0,194,276 Bl; Winter, U.S. Pat. No. 5,225,539; Winter, European Patent No. 0,239,400 Bl; Queen et al., European Patent No. 0451216 Bl; and Padlan, E. A. et al, EP 0519596 Al . See also, Newman, R. et al,
BioTechnology, 10: 1455-1460 (1992), regarding primatized antibody, and Ladner et al, U.S. Pat. No. 4,946,778 and Bird, R. E. et al, Science, 242: 423-426 (1988)) regarding single- chain antibodies. Antibodies produced from a library, e.g., phage display library, may also be used. In some embodiments, agents that specifically bind to a biomarker protein other than antibodies are used, such as peptides. Peptides that specifically bind to a biomarker protein can be identified by any means known in the art. For example, specific peptide binders of a biomarker protein can be screened for using peptide phage display libraries. c. Methods for Determining Levels of Protein Phosphorylation
In some embodiments, the methods described herein can include determining the amount of serine 9 phosphorylated human GSK-3 or a corresponding phosphorylatable amino acid in a homolog of the human protein in the sample relative to said amount in the absence of the compound or at an earlier timepoint.
Phosphorylation is a biochemical reaction in which a phosphate group is added to Ser, Thr or Tyr residues of a protein and is catalyzed by protein kinase enzymes. Phosphorylation normally modifies the functions of target proteins, often causing activation. As part of the cell's homeostatic mechanisms, phosphorylation is only a transient process which is reversed by other enzyme called phosphatases. Therefore, protein phosphorylation levels change over time and can be evaluated in a number of well-known manners, including, for example, by immunological approaches. For example, the amount of serine 9 phosphorylated GSK-3 can be determined by an immunoassay using a reagent which specifically binds with serine 9 phosphorylated GSK-3P . Such an immunoassay comprises a number of well-known forms, including, without limitation, a radioimmunoassay, a Western blot assay, an
immunofluoresence assay, an enzyme immunoassay, an immunoprecipitation assay, a chemiluminescence assay, an immunohistochemical assay, a dot blot assay, or a slot blot assay. General techniques to be used in performing the various immunoassays noted above and other variations of the techniques, such as in situ proximity ligation assay (PLA), fluorescence polarization immunoassay (FPIA), fluorescence immunoassay (FIA), enzyme immunoassay (EIA), nephelometric inhibition immunoassay (NIA), enzyme linked immunosorbent assay (ELISA), and radioimmunoassay (RIA), ELISA, etc. alone or in combination or alternatively with NMR, MALDI-TOF, LC-MS/MS, are known to those of ordinary skill in the art.
In one embodiment, the enzyme immunoassay is a sandwich enzyme immunoassay using a capture antibody or fragment thereof which specifically binds with GSK-3 regardless of phosphorylation status and a detection antibody or fragment thereof which specifically binds with serine 9 phosphorylated form of the protein. Immunological reagents for identifying a protein, as well as phosphorylated forms of GSK-3 are well known in the art (e.g., Phospho-GSK-3 (ser9) antibody, Cell Signaling Technology®, catalogue number 9336 and the phospho-GSK-3 (ser9) antibody from abeam®, catalogue number ab75814). Methods for generating antibodies that bind to phospho-forms of proteins are well known in the art. Such antibody reagents (e.g. , monoclonal antibody) can be used to isolate and/or determine the amount of the respective proteins such as in a cellular lysate. Such reagents can also be used to monitor protein levels in a cell or tissue, e.g., white blood cells or lymphocytes, as part of a clinical testing procedure, e.g., in order to monitor an optimal dosage of an inhibitory agent. Detection can be facilitated by coupling (e.g., physically linking) the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials.
Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β- galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine,
dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include
1 25 luciferase, luciferin, and aequorin, and examples of suitable radioactive material include I, 13 11, 35S or 3H.
The screening assays described herein can be adapted to identify candidate/test compounds which modulate (e.g., stimulate or inhibit) the expression, activity, or
phosphorylation status of a protein, e.g., GSK-3P, or a protein target of the kinase. Examples of such target molecules or substrates include phosphorylated target proteins in the signaling pathway of GSK-3 .
In another embodiment, the disclosure provides assays for screening candidate/test compounds which interact with (e.g., bind to) GSK-3 protein. "Binding compound" shall refer to a binding composition, such as a small molecule, an antibody, a peptide, a peptide or non-peptide ligand, a protein, an oligonucleotide, an oligonucleotide analog, such as a peptide nucleic acid, a lectin, or any other molecular entity that is capable of specifically binding to a target protein or molecule or stable complex formation with an analyte of interest, such as a complex of proteins. "Binding moiety" means any molecule to which molecular tags can be directly or indirectly attached that is capable of specifically binding to an analyte. Binding moieties include, but are not limited to, antibodies, antibody binding compositions, peptides, proteins, nucleic acids and organic molecules having a molecular weight of up to about 1000 daltons and containing atoms selected from the group consisting of hydrogen, fluoride, carbon, oxygen, nitrogen, sulfur and phosphorus. Typically, the assays are cell-based assays. The cell, for example, can be of mammalian origin expressing GSK-3 .
In other embodiments, the assays are cell-free assays which include the steps of combining a GSK-3P protein or a biologically active portion thereof, and a candidate/test compound, e.g., under conditions which allow for interaction of {e.g., binding of) the candidate/test compound to the GSK-3 protein or portion thereof to form a complex, and detecting the formation of a complex, in which the ability of the candidate compound to interact with {e.g., bind to) the GSK-3 polypeptide or fragment thereof is indicated by the presence of the candidate compound in the complex. Formation of complexes between the GSK-3 protein and the candidate compound can be quantitated, for example, using standard immunoassays. Such analyses would identify test compounds as GSK-3 ligands.
To perform the above drug screening assays, it can be desirable to immobilize either
GSK-3 or its target molecules to facilitate separation of complexes from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay. Interaction {e.g., binding of) of GSK-3 to a target molecule, in the presence and absence of a candidate compound, can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtitre plates, test tubes, and micro-centrifuge tubes. In one embodiment, a fusion polypeptide can be provided which adds a domain that allows the polypeptide to be bound to a matrix. For example, glutathione-S-transferase/GSK-3 fusion polypeptides can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, Mo.) or glutathione derivatized microtitre plates, which are then combined with the cell lysates {e.g., 35 S-labeled) and the candidate compound, and the mixture incubated under conditions conducive to complex formation {e.g., at physiological conditions for salt and pH). Following incubation, the beads are washed to remove any unbound label, and the matrix immobilized and radiolabel determined directly, or in the supernatant after the complexes are dissociated. Alternatively, the complexes can be dissociated from the matrix, separated by SDS-PAGE, and the level of GSK-3 -binding polypeptide found in the bead fraction quantitated from the gel using standard electrophoretic techniques.
Other techniques for immobilizing polypeptides on matrices can also be used in the exemplary drug screening assays of the invention. For example, either GSK-3 or its target molecule can be immobilized utilizing conjugation of biotin and streptavidin. Biotinylated GSK-3 molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, 111.), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
Alternatively, antibodies reactive with GSK-3P, but which do not interfere with binding of the polypeptide to its target molecule can be derivatized to the wells of the plate, and GSK-3 trapped in the wells by antibody conjugation. As described above, preparations of a GSK-3 - binding polypeptide and a candidate compound are incubated in the GSK-3 -presenting wells of the plate, and the amount of complex trapped in the well can be quantitated. Methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies reactive with the GSK- 3β target molecule, or which are reactive with GSK-3 polypeptide and compete with the target molecule; as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the target molecule.
As used herein, "time course" shall refer to the amount of time between an initial event and a subsequent event. For example, with respect to a subject's disease progression, time course may relate to a subject's disease and may be measured by gauging significant events in the course of the disease, wherein the first event may be diagnosis and the subsequent event may be proliferation, metastasis, etc.
Once binding is confirmed, additional assays, such as kinase assays to determine inhibition of phosphorylation effects, can be performed according to well known methods in the art. For example, assays for determining GSK-3 kinase activity are well known in the art (see, for example, the publications described herein and incorporated by reference in their entirety). Phosphorylation of a substrate of GSK-3 can be detected using a labelled phosphate group, such as the use of the radioactive label 32P present as the ATP source in the buffer. Alternatively, antibodies specific for the phosphorylated products of GSK-3 catalytic activity can be used to detect activity. As will be apparent to those of ordinary skill in the art, the assays are easily amenable to high through-put technologies using robotics and automated processes.
Significant modulation of phosphorylation of serine 9 of GSK-3 can be assessed if the output under analysis is inhibited by 1.1-, 1.2-, 1.3-, 1.4-, 1.5-, 1.6-, 1.7-, 1.8-, 1.9-, 2.0-, 2.1-, 2.2-, 2.3-, 2.4-, 2.5-, 2.6-, 2.7-, 2.8-, 2.9-, 3.0-, 3.1-, 3.2-, 3.3-, 3.4-, 3.5-, 3.6-, 3.7-, 3.8-, 3.9-, 4.0-, 4.1-, 4.2-, 4.3-, 4.4-, 4.5-, 4.6-, 4.7-, 4.8-, 4.9-, 5.0-, 5.5-, 6.0, 6.5-, 7.0-, 7.5-, 8.0-, 8.5-, 9.0- 9.5-, 10-, 11-, 12-, 13-, 14-, 15-, 16-, 17-, 18-, 19-, 20-fold or more different (including any range inclusive), relative to a control.
VI. Further Uses and Methods of the Present Invention
The compositions described herein can be used in a variety of diagnostic, prognostic, and therapeutic applications,
a. Screening Methods
One aspect of the present invention relates to screening assays, including non-cell based assays.
In one embodiment, the invention relates to assays for screening test agents which bind to, or modulate the biological activity of, at least one biomarker listed in Tables 2-6. In one embodiment, a method for identifying such an agent entails determining the ability of the agent to modulate, e.g. inhibit, the at least one biomarker listed in Tables 2-6.
In one embodiment, an assay is a cell-free or cell-based assay, comprising contacting at least one biomarker listed in Tables 2-6, with a test agent, and determining the ability of the test agent to modulate (e.g. inhibit) the enzymatic activity of the biomarker, such as by measuring direct binding of substrates or by measuring indirect parameters as described below.
For example, in a direct binding assay, biomarker protein (or their respective target polypeptides or molecules) can be coupled with a radioisotope or enzymatic label such that binding can be determined by detecting the labeled protein or molecule in a complex. For example, the targets can be labeled with 1251, 35S, 14C, or 3H, either directly or indirectly, and the radioisotope detected by direct counting of radioemmission or by scintillation counting. Alternatively, the targets can be enzymatically labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product. Determining the interaction between biomarker and substrate can also be accomplished using standard binding or enzymatic analysis assays. In one or more embodiments of the above described assay methods, it may be desirable to immobilize polypeptides or molecules to facilitate separation of complexed from uncomplexed forms of one or both of the proteins or molecules, as well as to accommodate automation of the assay.
Binding of a test agent to a target can be accomplished in any vessel suitable for containing the reactants. Non-limiting examples of such vessels include microtiter plates, test tubes, and micro-centrifuge tubes. Immobilized forms of the antibodies of the present invention can also include antibodies bound to a solid phase like a porous, microporous (with an average pore diameter less than about one micron) or macroporous (with an average pore diameter of more than about 10 microns) material, such as a membrane, cellulose,
nitrocellulose, or glass fibers; a bead, such as that made of agarose or polyacrylamide or latex; or a surface of a dish, plate, or well, such as one made of polystyrene.
In an alternative embodiment, determining the ability of the agent to modulate the interaction between the biomarker and a substrate or a biomarker metabolite and its natural binding partner can be accomplished by determining the ability of the test agent to modulate the activity of a polypeptide or other product that functions downstream or upstream of its position within the pathway (e.g., feedback loops).
The present invention further pertains to novel agents identified by the above- described screening assays. Accordingly, it is within the scope of this invention to further use an agent identified as described herein in an appropriate animal model. For example, an agent identified as described herein can be used in an animal model to determine the efficacy, toxicity, or side effects of treatment with such an agent. Alternatively, an antibody identified as described herein can be used in an animal model to determine the mechanism of action of such an agent. b. Predictive Medicine
The present invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, and monitoring clinical trials are used for prognostic (predictive) purposes to thereby treat an individual prophylactically. Accordingly, one aspect of the present invention relates to diagnostic assays for determining the amount and/or activity level of a biomarker listed in Tables 2-6 in the context of a biological sample (e.g., blood, serum, cells, or tissue) . Such assays can be used for prognostic or predictive purpose to thereby prophylactically treat an individual prior to the onset or after recurrence of a disorder characterized by or associated with biomarker polypeptide, nucleic acid expression or activity. The skilled artisan will appreciate that any method can use one or more (e.g., combinations) of biomarkers listed in Tables 2-6.
Another aspect of the present invention pertains to monitoring the influence of agents (e.g. , drugs, compounds, and small nucleic acid-based molecules) on the expression or activity of a biomarker listed in Tables 2-6. These and other agents are described in further detail in the following sections. The skilled artisan will also appreciated that, in certain embodiments, the methods of the present invention implement a computer program and computer system. For example, a computer program can be used to perform the algorithms described herein. A computer system can also store and manipulate data generated by the methods of the present invention which comprises a plurality of biomarker signal changes/profiles which can be used by a computer system in implementing the methods of this invention. In certain embodiments, a computer system receives biomarker expression data; (ii) stores the data; and (iii) compares the data in any number of ways described herein (e.g. , analysis relative to appropriate controls) to determine the state of informative biomarkers from a sample. In other embodiments, a computer system (i) compares the determined expression biomarker level to a threshold value; and (ii) outputs an indication of whether said biomarker level is significantly modulated (e.g., above or below) the threshold value, or a phenotype based on said indication.
In certain embodiments, such computer systems are also considered part of the present invention. Numerous types of computer systems can be used to implement the analytic methods of this invention according to knowledge possessed by a skilled artisan in the bioinformatics and/or computer arts. Several software components can be loaded into memory during operation of such a computer system. The software components can comprise both software components that are standard in the art and components that are special to the present invention (e.g., dCHIP software described in Lin et al. (2004) Bioinformatics 20, 1233-1240; radial basis machine learning algorithms (RBM) known in the art).
The methods of the invention can also be programmed or modeled in mathematical software packages that allow symbolic entry of equations and high-level specification of processing, including specific algorithms to be used, thereby freeing a user of the need to procedurally program individual equations and algorithms. Such packages include, e.g.,
Matlab from Mathworks (Natick, Mass.), Mathematica from Wolfram Research (Champaign, 111.) or S-Plus from MathSoft (Seattle, Wash.).
In certain embodiments, the computer comprises a database for storage of biomarker data. Such stored profiles can be accessed and used to perform comparisons of interest at a later point in time. For example, biomarker expression profiles of a sample derived from the blood or tissue of a subject and/or profiles generated from population-based distributions of informative loci of interest in relevant populations of the same species can be stored and later compared to that of a sample derived from the blood or tissue of the subject . In addition to the exemplary program structures and computer systems described herein, other, alternative program structures and computer systems will be readily apparent to the skilled artisan. Such alternative systems, which do not depart from the above described computer system and programs structures either in spirit or in scope, are therefore intended to be comprehended within the accompanying claims. c. Diagnostic Assays
The present invention provides, in part, methods, systems, and code for accurately classifying whether a biological sample is associated with a condition that is likely to respond to a therapy. In some embodiments, the present invention is useful for classifying a sample (e.g., from a subject) as associated with or at risk for responding to or not responding to a therapy using a statistical algorithm and/or empirical data (e.g., the amount or activity of a biomarker listed in Table s 2-6).
An exemplary method for detecting the amount or activity of a biomarker listed in Table s 2-6, and thus useful for classifying whether a sample is likely or unlikely to respond to a therapy involves obtaining a biological sample from a test subject and contacting the biological sample with an agent.
In certain instances, the statistical algorithm is a single learning statistical classifier system. For example, a single learning statistical classifier system can be used to classify a sample as a based upon a prediction or probability value and the presence or level of the biomarker. The use of a single learning statistical classifier system typically classifies the sample as, for example, a likely anti-immune checkpoint inhibitor therapy responder or progressor sample with a sensitivity, specificity, positive predictive value, negative predictive value, and/or overall accuracy of at least about 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
Other suitable statistical algorithms are well known to those of skill in the art. For example, learning statistical classifier systems include a machine learning algorithmic technique capable of adapting to complex data sets (e.g., panel of markers of interest) and making decisions based upon such data sets. In some embodiments, a single learning statistical classifier system such as a classification tree (e.g., random forest) is used. In other embodiments, a combination of 2, 3, 4, 5, 6, 7, 8, 9, 10, or more learning statistical classifier systems are used, preferably in tandem. Examples of learning statistical classifier systems include, but are not limited to, those using inductive learning (e.g., decision/classification trees such as random forests, classification and regression trees (C&RT), boosted trees, etc.), Probably Approximately Correct (PAC) learning, connectionist learning (e.g. , neural networks (NN), artificial neural networks (ANN), neuro fuzzy networks (NFN), network structures, perceptrons such as multi-layer perceptrons, multi-layer feed-forward networks, applications of neural networks, Bayesian learning in belief networks, etc.), reinforcement learning (e.g., passive learning in a known environment such as naive learning, adaptive dynamic learning, and temporal difference learning, passive learning in an unknown environment, active learning in an unknown environment, learning action-value functions, applications of reinforcement learning, etc.), and genetic algorithms and evolutionary programming. Other learning statistical classifier systems include support vector machines (e.g., Kernel methods), multivariate adaptive regression splines (MARS), Levenberg- Marquardt algorithms, Gauss-Newton algorithms, mixtures of Gaussians, gradient descent algorithms, and learning vector quantization (LVQ). In certain embodiments, the method of the present invention further comprises sending the sample classification results to a clinician, e.g., an oncologist.
In another embodiment, the diagnosis of a subject is followed by administering to the individual a therapeutically effective amount of a defined treatment based upon the diagnosis.
In one embodiment, the methods further involve obtaining a control biological sample (e.g., biological sample from a subject who does not have a condition. d. Prognostic Assays
The diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk that are likely or unlikely to be responsive to a therapy. The assays described herein, such as the preceding diagnostic assays or the following assays, can be utilized to identify a subject having or at risk of developing a disorder associated with a misregulation of the amount or activity of at least one biomarker described in Tables 2-6. Alternatively, the prognostic assays can be utilized to identify a subject having or at risk for developing a disorder associated with a misregulation of the at least one biomarker described in Table 2-6. Furthermore, the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, polypeptide, peptide, nucleic acid, small molecule, or other drug candidate) to treat a disease or disorder associated with the aberrant biomarker expression or activity. e. Treatment Methods The compositions described herein (including dual binding antibodies and derivatives and conjugates thereof) can be used in a variety of in vitro and in vivo therapeutic applications using the formulations and/or combinations described herein. VII. Pharmaceutical Compositions
In another aspect, the present invention provides pharmaceutically acceptable compositions which comprise a therapeutically-effective amount of an agent that modulates biomarker expression and/or activity {e.g., GSK-3P activity), one or more anti-immune checkpoint inhibitors, or a combination thereof, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents. As described in detail below, the pharmaceutical compositions of the present invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, boluses, powders, granules, pastes; (2) parenteral administration, for example, by subcutaneous, intramuscular or intravenous injection as, for example, a sterile solution or suspension; (3) topical application, for example, as a cream, ointment or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; or (5) aerosol, for example, as an aqueous aerosol, liposomal preparation or solid particles containing the compound.
The phrase "therapeutically-effective amount" as used herein means that amount of an agent that modulates biomarker expression and/or activity, or expression and/or activity of the complex, or composition comprising an agent that modulates biomarker expression and/or activity, or expression and/or activity of the complex, which is effective for producing some desired therapeutic effect at a reasonable benefit/risk ratio.
The phrase "pharmaceutically acceptable" is employed herein to refer to those agents, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication,
commensurate with a reasonable benefit/risk ratio.
The phrase "pharmaceutically-acceptable carrier" as used herein means a
pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject chemical from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject. Some examples of materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations.
The term "pharmaceutically-acceptable salts" refers to the relatively non-toxic, inorganic and organic acid addition salts of the agents that modulates biomarker expression and/or activity, or expression and/or activity of the complex encompassed by the invention. These salts can be prepared in situ during the final isolation and purification of the agents, or by separately reacting a purified agent in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed. Representative salts include the
hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like (See, for example, Berge et al. (1977) "Pharmaceutical Salts", J. Pharm. Sci. 66: 1-19).
In other cases, the agents useful in the methods of the present invention may contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically- acceptable salts with pharmaceutically-acceptable bases. The term "pharmaceutically- acceptable salts" in these instances refers to the relatively non-toxic, inorganic and organic base addition salts of agents that modulates biomarker expression and/or activity, or expression and/or activity of the complex. These salts can likewise be prepared in situ during the final isolation and purification of the agents, or by separately reacting the purified agent in its free acid form with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically-acceptable metal cation, with ammonia, or with a pharmaceutically- acceptable organic primary, secondary or tertiary amine. Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like. Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like (see, for example, Berge et ah, supra).
Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
Examples of pharmaceutically-acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
Formulations useful in the methods of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal, aerosol and/or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingredient, which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, most preferably from about 10 per cent to about 30 per cent.
Methods of preparing these formulations or compositions include the step of bringing into association an agent that modulates biomarker expression and/or activity, with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association an agent with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
Formulations suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of an agent as an active ingredient. A compound may also be administered as a bolus, electuary or paste.
In solid dosage forms for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the active ingredient is mixed with one or more
pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, acetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface- active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered peptide or peptidomimetic moistened with an inert liquid diluent.
Tablets, and other solid dosage forms, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions, which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions, which can be used include polymeric substances and waxes. The active ingredient can also be in microencapsulated form, if appropriate, with one or more of the above-described excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1 ,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
Suspensions, in addition to the active agent may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
Formulations for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more agents with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
Formulations which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
Dosage forms for the topical or transdermal administration of an agent that modulates (e.g., inhibits) biomarker expression and/or activity include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active component may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which may be required. The ointments, pastes, creams and gels may contain, in addition to an agent, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
Powders and sprays can contain, in addition to an agent that modulates (e.g. , inhibits) biomarker expression and/or activity, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
The agent that modulates (e.g., inhibits) biomarker expression and/or activity, can be alternatively administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation or solid particles containing the compound. A nonaqueous (e.g., fluorocarbon propellant) suspension could be used. Sonic nebulizers are preferred because they minimize exposing the agent to shear, which can result in degradation of the compound.
Ordinarily, an aqueous aerosol is made by formulating an aqueous solution or suspension of the agent together with conventional pharmaceutically acceptable carriers and stabilizers. The carriers and stabilizers vary with the requirements of the particular compound, but typically include nonionic surfactants (Tweens, Pluronics, or polyethylene glycol), innocuous proteins like serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars or sugar alcohols. Aerosols generally are prepared from isotonic solutions.
Transdermal patches have the added advantage of providing controlled delivery of an agent to the body. Such dosage forms can be made by dissolving or dispersing the agent in the proper medium. Absorption enhancers can also be used to increase the flux of the
peptidomimetic across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the peptidomimetic in a polymer matrix or gel.
Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention.
Pharmaceutical compositions of this invention suitable for parenteral administration comprise one or more agents in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be
accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution, which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
Injectable depot forms are made by forming microencapsule matrices of an agent that modulates biomarker expression and/or activity, in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions, which are compatible with body tissue.
When the agents of the present invention are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier. Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be determined by the methods of the present invention so as to obtain an amount of the active ingredient, which is effective to achieve the desired therapeutic response for a particular subject, composition, and mode of administration, without being toxic to the subject.
VIII. KITS
The present invention also encompasses kits for detecting and/or modulating biomarkers described herein. A kit of the present invention may also include instructional materials disclosing or describing the use of the kit or an antibody of the disclosed invention in a method of the disclosed invention as provided herein. A kit may also include additional components to facilitate the particular application for which the kit is designed. For example, a kit may additionally contain means of detecting the label (e.g. , enzyme substrates for enzymatic labels, filter sets to detect fluorescent labels, appropriate secondary labels such as a sheep anti-mouse-HRP, etc.) and reagents necessary for controls (e.g., control biological samples or metabolite standards). A kit may additionally include buffers and other reagents recognized for use in a method of the disclosed invention. Non-limiting examples include agents to reduce non-specific binding, such as a carrier protein or a detergent.
The invention now being generally described, it will be more readily understood by reference to the following examples which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention.
EXEMPLIFICATION
Example 1: Materials and Methods for Examples 2-8
Canine Pacing Models
A canine pacing model was used as described by Chakir et al. (Chakir, K., et al.
(2011) Sci Transl Med 3: 100ral88.; Chakir, K., et al. (2009) Circulation 119: 1231-1240). All dogs except controls received a pacemaker implant (Medtronic). Dyssynchronous heart failure (HFdys) and CRT models both received left bundle branch radio-frequency ablation at time of pacemaker implant, while in 3 other models (HFsync, AVA, V3A3), the left bundle remained intact. The location and timing of pacing varied between the experimental models and is detailed in Figure 1. All dogs were provided 7-10 days for full recovery prior to initiating 6 weeks of tachypacing (200 bpm). The location of pacing varied by model: HFdys and HFsync were atrially tachypaced for the entire 6 weeks, while CRT animals were subjected first to three weeks of atrial tachypacing, followed by three weeks of biventricular
tachypacing (RV apex and LV lateral wall), the V3A3 model was RV paced for the first 3 weeks, and then received 3 weeks of atrial tachypacing, while the AVA model was atrially paced atrially for the entire period excepting a two week period in the middle when it received right ventricular pacing. After 3 weeks of pacing, a subset of dogs were placed under anesthesia and a 7F bioptome (Cordis Corporation) was used to collect 5-7 biopsies from the endocardium near the right ventricular apex, under fluoroscopic guidance.
At terminal study, animals were anesthetized with pentobarbital, hearts excised under ice-cold cardioplegia, and tissue prepared for analysis. Table 1 provides global
hemodynamics and echocardiographic data for each model - revealing the presence of cardiac failure at the chamber level compared with normal controls. All models display chamber dilation, systolic, and diastolic dysfunction, though the CRT model had improved systolic function (dP/dtmax/IP (IP - instantaneous developed pressure at time of dP/dtmax).
Table 1
Figure imgf000070_0001
Table 1 shows echocardiography (at 3 and 6 weeks) and hemodynamic (at sacrifice) indices in the dog models. DI, dyssynchrony index; LVEDV, left ventricular end-diastolic volume; LVESV, left ventricular end-systolic volume; SV, stroke volume; LVEF, left ventricular ejection fraction; HR, heart rate; LVPes, left ventricular end-systolic pressure; LVPed, left ventricular end-diastolic pressure; SBP, systolic blood pressure; DBP, diastolic blood pressure; MAP, mean arterial pressure; dP/dtmax, maximum rate of change of the pressure waveform; dP/d n minimum rate of change of the pressure waveform; IP, isovolumic pressure. Italicized values indicates P < 0.05 vs. Control/Baseline value. *, P < 0.05 vs. HFdys,†, P < 0.05 vs. the 3 week value in the same model.
Membrane Permeabilized Myocytes
Tissue from the endocardium layer of the left ventricular lateral wall was flash frozen in liquid nitrogen and stored at -80 °C. Subsequently, the tissue was homogenized in isolation solution, in the presence of 0.3% Triton X-100, and protease and phosphatase inhibitors. Isolation solution contained (in mM): 5.5 Na2ATP, 7.11 MgCl2, 2 EGTA, 108.01 KC1, 8.91 KOH, 10 imidazole, 10 DTT. Myocytes were then washed in the absence of Triton X-100 to remove the detergent. Myocytes were glued, using silicone, to the tips of 150 μιη diameter minutia pins attached to a force transducer and motor arm (Aurora Scientific Inc., Aurora, ON, Canada). Sarcomere length was monitored by video camera (Imperx, Boca Raton, FL) and calculated by the High-speed Video Sarcomere Length Program (Aurora Scientific Inc.). Myocytes were maintained at a sarcomere length of 2.1 μιη. Myocytes were kept in relaxing buffer containing (in mM): 5.95 Na2ATP, 6.41 MgCl2, 10 EGTA, 100 BES, 10
phosphocreatine, 50.25 potassium propionate, 10 DTT, protease and phosphatase inhibitors. Force was measured as the myocyte was exposed to increasing calcium, by combining relaxing and activating buffers in set rations. Activating buffer contained (in mM): 5.95 Na2ATP, 6.2 MgCl2, 10 Ca2+EGTA, 100 BES, 10 phosphocreatine, 29.98 potassium propionate, 10 DTT, protease and phosphatase inhibitors. All buffers were adjusted to a pH of 7.0. A complete activation of the myocyte occurred at the beginning and end of the experiment, and the myocyte discarded if there was >10% rundown. Force-Ca2+ data were fit to the Hill Equation: F = Fmax * Cah/(ECh 50 + Cah).
Subsets of myocytes were exposed to either glycogen synthase kinase-3P (GSK-3P, 10 μg/mL, 20 min, Sigma- Aldrich, St. Louis, MO), PKA (0.125 units/mL, 20 minutes, Sigma- Aldrich), or Akt (10 μg/mL, 20 min, Sigma- Aldrich). Force-calcium data was collected before and after the treatment.
Multicellular Fragments
Tissue samples were minced into 2-4 mm pieces then mechanically homogenized three times at low speed (1,000 rpm, 3 sec), and re-suspended in relaxing solution containing Triton X-100 overnight at 4 °C. All solutions contained protease inhibitors (Sigma-Aldrich).
Multicellular skinned fragments (0.1-0.25 mm wide; 1-2.5 mm long) were attached to aluminum T-clips and mounted to arms extending from a force transducer (World Precision Instruments) and a high-speed length controller (Aurora Scientific Inc). Sarcomere length was set to 2.3 um by laser diffraction. The isolated muscle was then exposed to a range of calcium solutions while generated tension and consumed ATP were measured simultaneously during the contraction. ATP consumption was measured as previously described using a UV coupled optical absorbance method3. Following each contraction, calibration steps were performed by step-wise injection of 250 pmol of ADP. Activating solution contained (in mM): 20
Ca2+EGTA, 1.55 potassium propionate, 6.59 magnesium chloride, 100 BES, 5 sodium azide, 1 DTT, 10 phosphoenolpyruvate, 0.01 oligomycin, 0.01 PMSF, and 0.01 A2P5, as well as protease inhibitor cocktail. Relaxing solution was identical except it contained (in mM) 20 EGTA, potassium propionate 21.2, and magnesium chloride 7.11. The pre-activating solution contained (in mM) 0.5 EGTA, 19.5 HDTA (Fluka), and 21.8 potassium propionate.
Furthermore, all solutions used in the ATPase assay contained 0.5 mg/mL pyruvate kinase and 0.05 mg/mL lactate dehydrogenase (Sigma).
High frequency muscle length perturbations (1%; 500 Hz) were applied continuously to measure stiffness (the relative number of attached cycling cross-bridges during tension development). The rate of force redevelopment following a release-restretch maneuver, ktr, was measured during a final contraction at maximum Ca2+. Only muscles that maintained greater than 80% maximal tension throughout the protocol were included for analysis.
Trabeculae Experiments
Trabeculae were isolated from the right ventricular free wall and skinned overnight in the presence of 1% Triton X-100 (to remove all membranous structures), and protease (Sigma- Aldrich, St. Louis, MO) and phosphatase inhibitors (Roche Diagnostics, Indianapolis, IN) at 4 °C. Trabeculae were attached via a "basket and hook" technique4, to a force transducer (SI Heidelberg) and stationary hook. The muscle was stretched to produce 5 mN»mm~2 of passive tension (corresponding to maximum twitch force). Force was measured as bath solution Ca2+ was increased from 0 to saturating levels (46.8 μΜ), obtained by proportional mixing of activating and relaxing solutions. Relaxing solution contained (in mM): 10 K2H2EGTA, 5.45 MgCl2, 5.5 Na2ATP, 15 Na2CrP, 80 KCl, 25 HEPES, 5 DTT, protease and phosphatase inhibitors. Activating solution contained (in mM): 10 Ca2+EGTA, 5.15 MgCl2, 5.5 Na2ATP, 15 Na2CrP, 80 KCl, 25 HEPES, 5 DTT, protease and phosphatase inhibitors. All buffers were adjusted to a pH of 7.2. Trabeculae were completely activated at the beginning and end of each experiment, and if there was greater than 10% rundown, the experiment was discarded.
In a subset of trabeculae, a second force-Ca2+ relation was determined after exposure to protein phosphatase 1 (PP1, 25,000 mU/mL for 60 minutes, New England Biolabs, Ipswich, MA).
Phos-tag gel
Tissue samples from Control, HFdys, and CRT hearts are treated to obtain a
myo filament-enriched protein sample as previously described in Arell et al. (2001) Circ. Res. 89:480-487 in the presence of protease and phosphatase inhibitors. Briefly, tissue was homogenized, using a glass dounce homogenizer, in ice-cold standard rigor buffer (SRB) and 1% Triton X-100. SRB contained (in mM): 75 KCl, 10 imidazole, 2 MgCl2, 2 EGTA, 1 NaN3, plus protease and phosphatase inhibitors. The sample was then centrifuged at 16,000 g for 1 minute at 4 °C. The supernatant was discarded, and the pellet was re-suspended in SRB without Triton X-100. This was repeated three times. The pellet was then solubilized in 8M urea and 4% CHAPS. The sample was centrifuged at maximum speed, room temperature, for 15 minutes, and the pellet was discarded. Myofilament-enriched samples were run on phos-Tag gels (Wako Pure Chemical Industries, Ltd., Chuo-ku, Osaka, Japan), transferred to a membrane and cardiac troponin I (Tnl) was detected with the 81-7 Tnl antibody (Spectral Diagnostics, Toronto, ON, Canada).
Western Blots
Tissue samples were prepared as above, biopsies were prepared using a 0.1 mL dounce micro-homogenizer (Radnoti LLC, Monrovia, CA), in 40
Figure imgf000073_0001
cell lysis buffer (Cell Signaling, Danvers, MA), and further solubilized by adding 1 % SDS. Samples were ran on 4-12% precast Bis-Tris gels (Invitrogen), transferred to nitrocellulose membranes, and blotted using custom MyBPC antibodies against phospho Ser-273, Ser-282, Ser-302 (Sadayappan et al. (2009) Circulation. 1 19: 1253-1262) and total, custom MLC2 phospho-antibodies against phospho Ser-15, phospho Tnl (Cell Signaling), total Tnl (Spectral Diagnostics), phospho- serine motif (Zymed Laboratories, San Francisco, CA), phospho-threonine motif (Zymed), Troponin T (Cell Signaling), total MLC2 (Novus Biologicals, Littleton, CO), P-Ser9 GSK-3P (Sigma-Aldrich), and/or total GSK-3P (Santa Cruz, Dallas, TX). Blots were scanned using an Odyssey Infrared Imager (Li-Cor Biosciences, Lincoln, NE), and analyzed using Odyssey Application Software (v3.0.30, Li-Cor Biosciences). Several of these antibodies had no prior publications supporting their use in canine heart tissue. In these instances, antibodies were optimized and verified by comparing western blots performed in alternate species (mouse). Fortunately, the myofilament proteins share very high amino acid homology across mammalian species. In some cases, blots were stripped with NewBlot Nitro stripping buffer (Li-Cor), and reprobed. Blots were scanned before re-probed to ensure efficient stripping.
Silver Stain
After running, gels were fixed in 40% ethanol, 10% acetic acid for 1 hour, and then washed in H20 overnight. The gel was the sensitized in 0.02% sodium thiosulfate for 1 minute, washed, and then incubated in cold 0.1% silver nitrate, 0.02% formaldehyde for 20 minutes. The gel was then washed and developed in 3% sodium carbonate, 0.05%
formaldehyde for two minutes. The reaction was terminated with 5% acetic acid.
2D-DiGE Gels
Myofilament-enriched HFdys and CRT samples were labeled with cyanine (Cy) 3 or Cy5, and a mixture of the two (as an internal control) labeled with Cy2 (0.4 iL dye/ 50 μg protein). All three samples were then isoelectrically focused on an IEF strip (GE Healthcare Biosciences, Piscataway, NJ). The IEF instrument was run with the following steps: 10 hours active rehydration at 50V, 1 hour rapid ramp to 250V, 1 hour rapid ramp to 500V, 1 hour rapid ramp to 1000V, 4 hours normal ramp to 10,000V, 50,000 V'hours at 10,000V. The strips were alkylated and reduced, and then separated by weight on a large-format (18 cm) 10% Bis-Tris resolving gel in MES buffer. After fixing and washing the gel, it was scanned on a Typhoon scanner (GE Healthcare). Image analysis was performed by REDFIN (Ludesi, Malmo, Sweden).
Mass Spectrometry
Generally, six HFdys and six CRT LV lateral wall myocardial samples were
myo filament-enriched (1 mg protein in 8M urea, 1% SDS) and digested in trypsin (Promega Corporation). Alternatively, myofilament enriched samples from HFdys tissue were split into two equal parts, one incubated in ATP γ-Ρ180 (Cambridge Isotope Laboratories, Inc.) labeled relax buffer, and the second incubated in labeled relax buffer plus 4 μg of GSK-3P (n = 3). Labeled relax buffer was prepared by combining relax buffer with. The labeled relax buffer contained a 1 : 1 ratio of unlabeled and 180 labeled ATP. The samples were incubated at 30 °C for 30 minutes.
All myofilament samples were fractionated by strong-cation exchange to decrease sample complexity and increase sequence coverage. Each fraction was then individually phospho-enriched using titanium oxide affinity chromatography. Phospho-peptides were analyzed on an Agilent 1200 nano-LC system (Agilent Technologies) connected to an LTQ- Orbitrap mass spectrometer (Thermo Scientific), equipped with a nanoelectrospray ion source.
All raw MS/MS data was searched using the Sorcerer 2™-SEQUEST® algorithm (Sage-N Research). Additional analysis was done using Scaffold PTM (vl .1.3, Proteome Software). For studies with GSK-3P treatment, MS data was searched using SEQUEST, using an additional variable PTM: labeled phosphorylation that included an additional 6.01 Da shift due to the 180-labeled phosphate group. Kinase consensus amino acid sequences were predicted using NetPhosK (available on the World Wide Web at
cbs.dtu.dk/services/NetPhosK/) and the Human Protein Reference Database (available on the World Wide Web at hprd.org).
Specifically, myo filament-enriched HFdys and CRT samples (1 mg protein in 8M urea, 1% SDS) were reduced, alkylated, then digested overnight in solution using trypsin (Promega, Madison, WI) (100 μg/1.0 mg protein). The reaction was stopped with 10% TFA, and the samples were desalted with Oasis HLB cartridges (Waters, Milford, MA). Strong cation exchange (SCX) columns were prepared using SCX bulk media (Nest Group, Southborough, MA). The sample was split into two parts: 60% was run through the SCX columns (1/3 collected as flow through, 1/3 eluted with SCX buffer plus 40 mM KC1, and 1/3 eluted with SCX buffer plus 150 mM KC1). These three fractions, along with the 40% not run through the SCX column, were then individually desalted.
To phospho-enrich the samples, 50 μΐ of titanium oxide (Ti02) beads (Glycen Corp., Columbia, MD) was added to each fraction (4 per sample, 6 samples per group) and incubated at room temperature on a shaker (1400 rpms) overnight. The Ti02 beads were then washed, and the sample eluted in 10%> ACN/3%> NH4OH. Samples were then dried down and resuspended in 70% ACN/1 % FA for LC-MS/MS analysis.
Phospho-peptides were analyzed on an Agilent 1200 nano-LC system (Agilent, Santa Clara, CA) connected to an LTQ-Orbitrap mass spectrometer (Thermo Scientific, Waltham, MA), equipped with a nanoelectrospray ion source. The peptides were separated on a C18 RP- HPLC column (75 μτη x 10 cm self-packed with 5 urn, 200 A Magic CI 8; Michrom Bio Resources, Auburn, CA) at a flow rate of 300 nL/min. Each fraction (4 per sample) was run separately to maximize the number of peptides acquired. Each MSI scan was followed by col lision induced dissociation of the 5 most abundant precursor ions. Mono-isotopic precursor selection was enabled and dynamic exclusion was enabled with a repeat count of 2, repeat duration of 30 seconds and exclusion duration of 60 seconds. Only MSI signals exceeding 1000 counts triggered the MS2 scans and +1 and unassigned charge states were not selected for MS2 analysis.
All raw MS/MS data was searched using the Sorcerer 2™-SEQUEST® algorithm (Sage-N Research, Milpitas, CA). Data was searched against a canine database allowing for: carbamidomethyl (fixed modification), oxidation (variable modification), and phosphorylation (variable modification). Tolerances were: 1.00 Da for fragments and 0.037-0.170 Da for parent ions, and allowing for 2 missed cleavages. Analysis was done in Scaffold 3 (v3.4.9, Proteome Software, Portland, OR) with protein and peptide probability thresholds of 95% and 90%), respectively (providing a protein false discovery rate of 0.5% and a peptide FDR of 0.4%)). Additional analysis was done using Scaffold PTM (vl .1.3, Proteome Software). For analysis, spectral counts were summed over all fractions per sample.
Kinase consensus amino acid sequences were predicted using NetPhosK
(cbs.dtu.dk/services/NetPhosK/) and using the Human Protein Reference Database (hprd.org) PhosphoMotif finder. A second set of samples was prepared for the GSK-3P treatment. Myofilament enriched samples (isolated myofibrils) were prepared from HFdys tissue as before. The sample from each dog was split into two equal parts, one incubated in labeled relax buffer, and the second incubated in labeled relax buffer plus 4 μg of GSK-3p. Labeled relax buffer was prepared by combining relax buffer with ATP γ-Ρ180 (Cambridge Isotope Laboratories, Inc., Andover, MA). The labeled relax buffer contained a 1 : 1 ratio of unlabeled and 180 labeled ATP (20 μιηοΐβ of each, per sample). The samples were incubated at 30 °C for 30 minutes. Post treatment, samples were prepared, run on the mass spectrometer, and analyzed in the same manner as above, with one exception: searching also included an additional variable PTM: labeled phosphorylation that includes an additional 6.01 Da shift due to the 180-labeled phosphate group.
Statistical Analysis
Values were compared between the groups using one-way analysis of variance (ANOVA), with post-hoc comparisons made with the Holm-Sidak method using SigmaPlot (vl 1, Systat Software Inc.). MS was quantified based on label free spectra counts. Normalized spectral counts were compared using Student's T-test. GSK-3P phosphorylation relationships were tested using Multivariate Regression Analysis using Systat (Systat Software, Inc.) All tests were two-tailed with a -value < 0.05 considered significant.
Example 2: CRT Globally Restores Myofilament Function
Steady state force-calcium data for skinned myocytes from the left ventricular (LV) lateral endocardium are shown in Figure 2A. Myofilament function was markedly depressed in HFdys, with maximum calcium-activated force (Fmax) declining nearly 50% compared to normal controls (Con: 37.9 ± 3.5 mN»mm~2, n = \5 myocytes from 6 dogs, HFdys: 21.5 ± 3.0 mN»mm~2, n = 16 myocytes from 6 dogs, P < 0.001, Figure 2B). HFdys also exhibited Ca2+- desensitization as quantified by a rise in EC50 (Con: 2.34 ± 0.13 μΜ, HFdys: 3.04 ± 0.17 μΜ, P = 0.003, Figure 2B). CRT remarkably and completely restored both parameters of
myofilament function to normal (Fmax: 40.3 ± 2.60 mN»mm~2, EC50: 2.18 ± 0.13 μΜ, n = 16 myocytes from 7 dogs, P < 0.001 vs. HFdys, P = N.S. vs. Con).
Contractile dyssynchrony with a left-bundle delay results in marked differences in regional stress (high in lateral wall due to early stretch-late contraction, lower in septum/RV with early contraction-late stretch). To determine if this influenced CRT modulation of myofilament function, we studied skinned trabeculae from the right ventricular (RV) free wall (Figures 2C-2D) and single myocytes from the LV septum (Figure 3). Both preparations revealed similarly depressed sarcomere function as found in the LV free wall for HFdys, and marked improvement with CRT. Thus, the precise nature of cyclical regional stress or strain temporal pattern was not a primary determinant of myofilament changes in either condition.
We next tested if CRT alters crossbridge mechano-energetics by simultaneously assessing isolated fiber bundle ATP utilization and force development. The ATP required to achieve a given crossbridge tension (tension cost) and the rate of force re-development upon sudden stress release (crossbridge kinetics, kb) were similarly depressed in HFdys and CRT (Figures 2E-2F). The number of attached crossbridges during tension development, assessed by dynamic stiffness, was unchanged from control values by either HFdys or CRT (Figure 2G). Thus, CRT did not impact the mechano-energetics of the crossbridge.
Example 3: Importance of re-synchronization
The recovery of myofilament function with CRT was not merely due to contraction being synchronous, as sarcomere function was also markedly depressed in HF that was always synchronous (HFsync). As with HFdys, Fmax was much reduced in HFsync (21.08 ± 3.22 mN»mm" 2, P < 0.001 vs. Con and CRT, P = N.S. vs. HFdys, n = 15 myocytes from 5 dogs) (Figure 4). However, whereas HFdys cells displayed a rise in EC50, those from HFsync had a lower value (i.e. sensitized to Ca2+; 1.70 ± 0.11 μΜ, P = 0.007, 0.036, 0.003 vs. Con, CRT and HFdys, respectively).
These data indicated that improved myofibrillar function required pre-existing dyssynchrony that was subsequently reversed. To test this concept further, we modified the HFsync model to include a 2-week period of dyssynchrony induced by RV pacing during weeks 3 and 4 (AVA model) (19). Compared to HFsync, AVA myocardium had a greater Fmax (AVA: 23.4 ± 1.9 mN-mm"2, n = \0 trabeculae from 5 dogs, HFsync: 16.5 ± 1.9, n = 22 trabeculae from 8 dogs, P < 0.05 vs. HFsync, P = N.S. vs. Con) and EC50 returned to normal control values (Figures 5A-5B).
CRT involved direct ventricular myocardial stimulation that might have triggered improved myofibrillar function. To test this, we studied an alternative "CRT" model that used rapid RV free-wall pacing (no LBBB was performed) to generate dyssynchrony and HF, and then subsequently resynchronized the hearts by switching to rapid atrial pacing, avoiding the need for any direct ventricular stimulation. The results for this model (V3A3) were very similar to those with CRT (Figures 5C-5D), indicating direct pacemaker stimulation of the LV was not required. Example 4: Altered Maximum Force is due to Myocyte Hypertrophy
The decline in Fmax (normalized to myocyte cross sectional area, CSA) in both HFdys and HFsync could result from proteolytic cleavage of myofibrillar proteins such as troponin I (27), or isoform switching (e.g. troponin T (28)). However, silver stained ID gels and western blots revealed neither protein cleavage products nor isoform switching in these models
(Figure 6). Normalization of force to CSA is generally performed since one assumes bigger ceils will have more force-generating units. Yet while control, HFdys, and CRT cel ls had similar raw (non-normalized) peak force, CSA differed markedly (Figure 7D). Specifically, HFdys had twice the CSA of controls, but this was reversed by CRT (Con: 251 ± 36, HFdys: 465 ± 64, CRT: 268 ± 44 μπι2, P < 0.012, 0.015 HFdys vs. Con and CRT, respectively) (Figures 7A-7B). To rule out potential selection bias in cells for which physiological data had been measured, we measured CSA from all cells in a given microscopic field (n = 150 myocytes from 3 dogs). This confirmed that HFdys myocytes were enlarged and that CRT reversed this (P < 0.001 , Figure 7C). In trabeculae, Fmax was also depressed (Figure 2C) but CSA was similar among the models (Figure 7E).
Example 5: Ca2+ -sensitization is mediated by myofilament phosphorylation
Figure 8A displays example force-Ca2+ data from RV trabeculae prior to and following incubation with the phosphatase PP1. Dephosphorylation did not change Fmax in any group
(Figure 9A). However, EC50 increased (de-sensitized) in Control (AEC50: 1.09 ± 0.22 μΜ, n = 6) and CRT (AEC5o: 0.70 ± 0.23 μΜ, n = 5) trabeculae, but was unaltered in HFdys (AEC5o: - 0.06 ± 0.14 μΜ, n = 5, P < 0.003, 0.04 vs. Con and CRT, respectively) (Figure 8B). PP1- induced calcium desensitization without a change in Fmax was also observed in V3 A3 and AVA models, indicating phosphorylation was involved with their improved Ca2+-sensitivity as well (Figure 9).
Ca2+ sensitivity is regulated by phosphorylation of the regulatory thin filament proteins troponin I (Tnl), troponin T (TnT), myosin binding protein C (MyBPC), and myosin light chain 2 (MLC2). Table 2 lists the phosphorylation sites involved, their respective influence on Ca2+-sensitivity, and directional changes in each due to heart failure (if reported). Table 2
Figure imgf000079_0001
Table 2 shows myofilament phosphorylation sites that are known to modulate calcium sensitivity. The table provides the direction of a change in calcium sensitivity due to phosphorylation, the direction that phosphorylation is modified by heart failure, and how this in turn impacts calcium sensitivity.
Phosphorylation of Tnl at S22/S23 (PKA) (29) and S43/S45 (PKC) (30, 31) desensitize, whereas at T144 (PKC) (31, 32) or S150 (AMP-kinase, PAK1) (33, 34) increase sensitivity (the latter only reported in vitro). Overall, Tnl phosphorylation declined similarly in HFdys and CRT (Figure 8C), and reduced S22/S23 targeting was directly confirmed by immunoblot
(Figure 10). This supports a Ca2+-sensitization affect (as opposed to desensitization observed in HFdys, e.g. Figure 2A). MyBPC phosphorylated at S273/S282/S302 (35) (by PKA and other kinases (36)) or MLC2 phosphorylation at S15/S19 increase Ca2+-sensitivity (37). These PTMs declined to the same extent in both failure models (Figures 8D-8E) and so could not explain the differences between HFdys and CRT. Lastly, TnT phosphorylation at T206 (38) (PKC) or S278/ T287 (39) (Rho-kinase II) are linked to sensitization, but these sites were not altered in the models (Figure 8F). The lack of differential changes in these sites was further confirmed by mass spectrometry (Table 3) and/or 2-D differential gel electrophoresis (Figure 11).
Table 3
Protein Site Loc Prob Total Spectra P -value
MLC2 SIS 100% 22 N.S. cMyBP-C S279 100% 132 M.S.
cMyBF-C S28S 100% 1800 N.S.
cMyBF-C S290 92% 43 N.S. cTnl S23 100% 312 N.S.
cTnl S24 100% 342 N.S.
cTnl S167 100% 4 N.S.
cTnl S200 100% 7 N.S. ft-Tro omyosi.n T232 93% 736 N.S.
ft-Tropomyosi.n S2S3 100% 3577 N.S.
Table 3 shows a selection of phosphorylation sites identified by mass spectrometry, which were not significantly different between HFdys and CRT. As stated in the online methods, the protein probability threshold was set at 95%; however, MLC2, included in this table, had a 93% protein probability. Loc. Prob., localization probability as calculated by Scaffold PTM (vl .1.3, Proteome Software) using the A-score algorithm.
Example 6: Z-Disk and M-Band Proteins are phosphorylated by CRT; Role of GSKSfi Given that the known phospho-protein targets did not appear to be involved, mass spectrometry was used to broadly interrogate the myofibrillar sub-proteome. A total of 1187 phosphorylation sites on 166 myofilament and myo filament-associated proteins were detected, and CRT significantly altered 33 of these sites. Based on the PPl results, residues exhibiting increased phosphorylation with CRT were assessed, yielding 15 sites on 13 proteins (Table 4 and Figures 12-26). Figure 27 summarizes this analysis.
Table 4
Figure imgf000081_0001
Table 4 shows myofilament phosphorylation sites increased by CRT. The peptide sequence is shown, where the phosphorylated site is indicated in lowercase. Possible kinases were determined using the Human Protein Reference Database and NetphosK. Loc Prob,
Localization probability as determined by the A-Score algorithm using Scaffold PTM;
Abliml, Actin binding LIM protein 1; THRAP3, Thyroid hormone receptor-associated protein 3; Sorbin2, Sorbin and SH3 domain-containing protein 2; LDB3, LIM domain-binding protein 3; LM07, LIM domain only protein 7.
The majority of modified sites resided in proteins expressed in the Z-disk and M-band regions. Though such proteomic analysis can bias towards high-abundance or high molecular weight proteins (e.g. titin or obscurin), our analysis identified only a single site on each, supporting response specificity. In silico analysis of the individual phosphorylation sites (using NetPhosK and Human Protein Reference Database) was used to identify kinase consensus sequences based on the phosphorylation targets (Table 4), and found that glycogen synthase kinase-3P (GSK-3P) and p38 MAPK targeted the most sites. p38 was found to be activated by HFdys and suppressed by CRT whereas GSK-3P was less active in HFdys and activated by CRT.
To determine if the phosphorylated residues increased by CRT were indeed targets of
GSK-3P, we performed a second mass spectrometry experiment. The myofilament subproteome obtained from HFdys dogs was incubated with 180 labeled ATP in the absence or presence of recombinant active GSK-3P for 30 minutes at 30°C and prepared for proteomic analysis. Table 5 lists the phosphorylated amino acid residues targeted by GSK-3P that were also found to increase with CRT. Four of the seven GSK-3P sites revealed by in silico analysis were confirmed, and four additional sites altered by CRT but not predicted by bio- informatics were revealed as GSK-3P targets as well. This supports the notion that GSK-3P converts a subset of the HFdys phospho-proteome into one similar to CRT.
Table 5
Figure imgf000082_0001
Table 5 shows that phosphorylation sites increased by CRT were also in vitro targets of GSK- 3β. Labeled sites incorporated the 180 labeled ATP. Predicted Target indicates whether the phosphorylation site was identified as a possible substrate for GSK-3P using in silico analysis (see Table 4). For each group, n = 3 dogs were pooled. *, low localization probability of the phospho-serine, indicating a nearby serine could also be a possible target.
To test whether GSK-3P myofilament phosphorylation could be responsible for the sensitizing affect, myofilament function was examined in HFdys myocytes before and after exposure to recombinant active GSK-3P for 30 min. Treated cells exhibited a decline in EC50 similar to that observed with CRT (P < 0.001 HFdys vs. pre -treatment, P = N.S. versus CRT) (Figures 27B-27C). By contrast, myocytes from CRT hearts were unchanged by GSK-3P, consistent with existing stimulation of this pathway and underlying sensitization. Control myocytes were also unaffected by GSK-3p. Fmax was unaltered by GSK-3P in any of the models, agreeing with our finding that Fmax is primarily affected by myocyte geometry in these models.
To determine if differential effects on calcium sensitivity was specific to GSK-3P, we exposed HFdys and CRT myocytes to PKA and Akt, two kinases involved in GSK-3P signaling. As expected, PKA modestly increased Fmax while decreasing calcium sensitivity (increased EC50), and it did so similarly in both groups (no interaction between group and treatment) (Figure 28). This result is consistent with reduced Tnl and MyBPC phosphorylation at PKA-targeted sites in both HFdys and CRT (Figures 8C-8D). Although Akt is a recognized upstream kinase modulator of GSK-3P, myocytes incubated with Akt exhibited no change in Fmax or EC50 in either group (Figures 29A-29B), supporting an alternative pathway. While Akt treatment did not detectably increase phosphorylation of any proteins in the myofilament fraction (Figure 29C), it did in whole cell lysate (serine 9 on GSK-3P, Figures 29D-29E). These PKA and Akt data further support the specificity of the CRT myofilament response to GSK-3P phosphorylation of myofilament proteins.
Example 7: Time Course and Offsetting Effects on Ca2+ Sensitivity by Dyssynchrony
The observation that Ca2+-sensitivity declined with HFdys but rose with HFsync suggested differential phosphorylation might mediate dysfunction due to dyssynchrony. To test this further, we examined the time course of changes in both models, performing studies at 3 weeks in myocytes isolated from endocardial biopsies. After three weeks of pacing, both models showed a similar decline in Fmax; however, there was dissociation of Ca2+-sensitivity (Figures 30A-30B), with a fall in EC50 in HFsync (sensitization), but no change in HFdys. There were no further declines in HFsync at week 6, whereas EC50 rose in HFdys. These data support an early sensitization factor in both models that is gradually offset by desensitization solely in HFdys (Figure 30C). This could relate to the decline in Tnl phosphorylation by week 3 that remained similarly reduced at week 6 in both models (Figures 30D-30E). However, the ratio of phospho/total GSK-3P was unaltered at either time in HFsync, whereas there was a gradual rise in the ratio in HFdys (enzyme inactivation) concordant with the gradual increase in EC50. These results support the notion that phospho/total GSK-3P may serve as a biomarker for dyssynchrony with relevance to contractile function. Example 8: Plasma Expression of GSK-3fi Myofilament Phosphorylation Targets
While biomarker analyses for GSK-3P expression, activity, or activation would generally require a cardiac biopsy, it has been described herein that GSK-3P has several myofilament targets that have been determined to be detectable in the blood, such as plasma (Table 6), when the myocardium is damaged. Phosphorylation of these sites is increased by CRT, in concordance with re-activation of GSK-3p. These phosphorylation sites thus represent biomarkers, correlating with myocardial GSK-3P activity and cardiac dyssynchrony. Table 6
Figure imgf000084_0001
Table 6 shows that GSK-3P myofilament phosphorylation targets are detectable in plasma.
The results in Table 6 were generated from proteins identified in plasma samples from the PaxDB website. Specifically, the dataset where generated from data uploaded to pride and the integrated dataset is as follows: 1. Human plasma, Lee, PRIDE,ID:8653-8672', 2 .Human plasma, non-alkylated, Qian,MCP,2005' (weighting 100%), and 3. Human plasma,
Peptideatlas,May,2010.
Example 9: Materials and Methods for Examples 10-13
Chronic Mouse Cardiac Pacemaker
In order to determine the extent that GSK-3P is involved in dyssynchrony mechano- transduction, it is necessary to use genetic modifications to block its inactivation or remove it entirely. Unfortunately, genetic tools in the dog are mostly limited to gene therapy, and even this turns out to be extremely difficult in this species. Thus, a mouse model was developed where the desired GSK-3P genetically engineered lines exist. For example, an inducible GSK-3P knock-out mouse line and a constitutive ly active GSK-3P model are available.
Although a mouse pacemaker system showing regional transcriptome modulation in a sub-acute study is known, the units were expensive to produce, could not be reused, and only lasted for a week. Also, the source (Guidant) ended when Boston Scientific purchased the company. Therefore, an alternative was required to be developed that could be used in chronic studies and larger sample sizes. The developed pacemaker system utilizes a custom designed micro-pacing unit (Figure 31 A), bi-polar implantable lead (Figure 3 IB), a connector at the back of the mouse's neck, a micro-commutator built into the top of the cage (allowing complete rotational freedom), and a power source attached to the outside of each cage. Cardiac dyssynchrony can be induced by single site pacing of the right ventricle (RV). Mice have been successfully paced dyssynchronously for 2 weeks and longer, which has been confirmed by ECG (Figure 31C) and echo (Figure 3 ID). Internal ECG leads can be implanted and passed through the commutator for easy and frequent confirmation of pacing capture. Importantly, RV pacing reduced systolic function (%FS) over the course of the pacing protocol (Figure 3 IE).
Quantitative Mass Spectrometry
Although generally available mass spectrometry (MS) approaches are sensitive and powerful, SWATH improves throughput and sensitivity . SWATH is a new MS approach that allows the quantification of tens of thousands of peptides generated from the tryptic digestion of the proteome and phospho-proteome in every single sample. The platform is stable, robust, fast, reproducible and sensitive. It is particularly useful in situations where post-translational modifications must be tracked over various models and treatments as is proposed below.
Example 10: Determining whether GSK-3fi inactivation is necessary and sufficient for dyssynchrony-induced myofilament calcium desensitization
GSK-3P is inactivated by cardiac dyssynchrony and this depresses calcium sensitivity.
CRT reversed both processes. To confirm GSK-3P is necessary to mediate dyssynchrony and
CRT's effect on calcium sensitivity, genetic models are required. GSK-3P gene-deletion and constitutively active mouse models are used, including pacing the RV to generate
dyssynchrony (and subsequently terminating pacing to mimic CRT) using a pacemaker system described above.
Paced mouse recapitulate the phenotype observed in the dog described in Examples 1- 8. Cardiac dyssynchrony via RV pacing (rate just above sinus at 700 bpm x 10 days) exhibits calcium desensitization compared to a lead implant without pacing (Figure 32A).
Simultaneously, pS9/total GSK-3P increased (de-activation) with RV pacing over no pacing (Figure 32B).
A GSK-3P myocyte targeted knock-out model (combines GSK-3P flox'd mouse with MerCreMer-tamoxifen inducible Cre coupled to aMHC promoter) is obtained. Data are assessed after 4 weeks of tamoxifen treatment to be free from any acute myocardial depression that can occur with this gene-targeting system. As shown in Figure 32C, myocytes from these hearts in which GSK-3P is suppressed show a right shift of the force-Ca2+ dependence, consistent with a desensitization of the myofilament to Ca2+. This supports the hypothesis based on the canine results described above where a loss of function analysis was not feasible.
Briefly, an embodiment of the pacemaker system consists of an implantable lead, the externalized connector, a micro-commutator (electrical swivel), a micro pacing unit, and power source. The bi-polar lead consists of two metal rings (positive, ground), 1.5 mm in diameter, attached (but isolated from each other) using a biocompatible epoxy. Each ring is attached to 5 cm of 36 AWG wire. The two wires terminate in a 4-pin 0.050" pitch male connector. The connector attaches to a female connector at the bottom of the flying lead micro-commutator, which is the smallest and lowest friction commutator available (see Dragonfly Research and Design, Inc.). The micro-commutator is attached to the lid of the mouse cage. The commutator connects to the custom pacing unit. The units are 9.5 mm in diameter, have a depth of 3.2 mm, and are coated in medical grade silicone. The chip is connected to a battery holder with 3 AA batteries (4.5 volts). Both the pacing chip and the batteries attach to the mouse cage lid.
In one embodiment, a mouse is anesthetized with etomidate, intubated, and ventilated. Anesthesia is maintained with inhaled isoflurane. A midline incision exposes the chest wall and abdomen. The right ventricular free wall is exposed by right anterolateral thoracotomy. The pacing lead is placed against the RV free wall and attached using 7-0 prolene suture. The lead is connected to an external stimulator, and ECG used to determine pacing voltage threshold. The threshold typically increases 1.5- to 2-fold over the first week, at which point it is relatively stable. Thus, the lead is repositioned if the threshold is over 2 V (the battery generates 4.5 V). The lead connector is tunneled to the mouse's back, and externalized on the back of the neck so the mouse cannot chew through it. A purse string knot keeps the wound closed around the connector, and a small amount of superglue keeps the connector in place. The chest is closed using 6-0 Prolene and negative pressure in the thorax is restored by removal of air by a chest tube attached to a syringe. The surgery lasts ~30 minutes and is associated with less than 25% mortality. The pacemaker is turned on after a one-week recovery period. During pacing, the animal cages are returned to the animal facility, which reduces stress and avoids weight loss problems during the protocol.
The pacing protocol can be applied to four groups. In one group, GSK-3P knock-out mice (GSK-SP^/Cre) treated with tamoxifen for three weeks, are used. To induce the knock- out, mice will be given tamoxifen (20 mg/kg IP), dissolved in 30% ethanol in sterile PBS at a final concentration of 4 mg/rnL. Previously, GSK-3pfl/fl/Cre mice treated with tamoxifen saw a 77% average reduction in GSK-3P expression (Woulfe, K.C., et al., (2010) Circ Res 106(10): 1635-45); the incomplete knock-out is due in part to its persistence in non-myocytes. In another group, controls for group 1 are mice with inducible Cre lacking the flox'd gene and GSK-3pfl/fl/-, both treated with tamoxifen for the same duration as group 1. For both groups, tamoxifen is administered before the pacemaker implant surgery. In a third group, a constitutively active knock-in model of GSK-3P, with an S9A substitution, GSK-3pS9A, are used. This mouse shows some minor developmental differences from wild-type, but a normal cardiac phenotype. In a fourth group, controls for group 3 are the wild-type littermate controls of the GSK-3pS9A mice (GSK-3pWT).
Intact heart function is assessed in vivo by trans-thoracic echocardiography. Mice are sedated (isoflurane) to facilitate pacing manipulation while images are obtained
(VisualSonics, Vevo 2100). End systolic and end-diastolic dimensions, left- ventricular fractional shortening, and mass are assessed as previously described (Takimoto, E., et al., (2005) Nat Med. 11(2):214-22). The septal-to-posterior wall motion delay (SPWMD), an index of LV dyssynchrony, can be calculated from the M-Mode echo (Pitzalis, M.V., et al., (2002) Journal of the American College of Cardiology 40(9): 1615-22). Dyssynchrony is assessed by speckle tracking in the mouse as well, as an alternative approach that is clinically used.
Skinned myocytes are prepared by homogenizing LV myocardium in isolation solution plus protease and phosphatase inhibitors and Triton X-100. Triton is a detergent that removes the membrane and leaves the myofilament. The myocyte is glued to two minutia pins using silicone. The myocyte is moved to relaxing solution (0 Ca2+), and sarcomere length is set to 2.1 μιη (detected by a high-speed camera). The myocyte is exposed to activating solution (Ca2+=46.8 μΜ), fully activating it. The myocyte is then exposed to sub- max Ca2+ concentrations achieved by mixing Relaxing and Activating solutions . Myocyte width and depth are recorded, to normalize data to cross-sectional area.
GSK-3P activity can also be assessed. Tissue samples are treated to obtain a myofilament-enriched sample as described in Arrell et al. in the presence of protease and phosphatase inhibitors (Arrell, D.K., et al., (2001) Circ Res. 89(6):480-7). In addition to the myofilament enrichment, "everything else" (cytosolic, mitochondria, membrane, etc.) is also kept. Antibodies against total GSK-3P (mouse) and p-S9 (rabbit) can be used simultaneously, using Licor IR secondary antibodies. The membrane is imaged on an Odyssey system (Licor), allowing simultaneous quantitation of phosphor- and total GSK-3P, the ratio of which represents GSK-3P activity.
In one embodiment, the baseline myofilament function and GSK-3P activity is determined in one or more of the four groups. In knock-out mice, this can be done before and after exogenous GSK-3P treatment (as described in Kirk, J.A., et al., (2014) Journal of
Clinical Investigation 124(1): 129-39). This shows if calcium sensitivity can be restored with short-term exposure to GSK-3p. The pacemaker implant is then be done in all four groups, requiring 6-7 mice per group. Additionally, another 6-7 sham controls per group receive an implanted RV lead, but no pacing. This is a total of 48 mice. Seven days post-implant, all four groups are echoed to determine baseline global function. Following echo, the mice are attached to the commutator system, and RV pacing is started (e.g., 700 bpm, 0.25 ms duration, 4.5 V).
While pacing, ECG readings are obtained every other day. Pacing can be confirmed by QRS morphology and heart rate equal to pacing frequency (700). If capture is lost, the mouse is excluded from the study. If capture is intermittent, the commutator is connected to an external stimulator, offering output voltages above 4.5 V. The mice are echoed three times (day 3, 7, and 14). The protocol lasts 14 days, but can be extended if needed. The heart is removed, washed in ice-cold PBS, and dissected to obtain the relevant regions. Each heart is optionally snap frozen in liquid N2 and stored at -80 °C until needed for GSK-3P activity assay, skinned myocytes, or proteomics described further herein. An embodiment of the groups and protocols is shown in Figure 33.
Data analysis includes 2-way ANOVA (mouse group vs. treatment) myofilament function parameters (Fmax, EC50, and ¾ obtained by fitting to Hill Equation using non-linear regression), and myofilament and cytoplasmic GSK-3P activity (p-S9/total GSK-3P). For the echo data, a three-way repeated measures ANOVA is necessary (group, treatment, time). The variance and magnitude of change of these parameters in the canine models described in examples 1-8 are used to identify the number of mice needed. For GSK-3P treatments (pre and post), data are compared via paired t-test.
GSK-3P deactivation in dyssynchrony may mediate myofilament calcium
responsiveness, via decreased phosphorylation of its myofilament targets as described further below. Therefore, the absence of the kinase (knock-out mouse) results in baseline calcium desensitization as confirmed by the data shown in Figure 32C. Constitutively active GSK-3P is protected from desensitization during dyssynchrony, due to the persistent phosphorylation of its myofilament targets. Figure 34 shows a schematic of expected results. Example 11
In parallel or as an alternative, similar experiments are conducted in existing canine models described in examples 1-8. To study GSK-3P in dog, pharmacological inhibitors are used, such as lithium (not very specific), LY2090314, and BIO. The pharmacokinetics of LY2090314 in dog has already been described, and is similar to humans (Zamek- Gliszczynski, M.J., et al, (2013) Drug Metab Dispos 41(4):714-26). To study dyssynchrony separate from HF, an implantable pacemaker in VDD mode, and RV pace at sinus, can be used. Pharmacological inhibitors are useful even in the mouse-pacing model. It establishes specificity, which is important since genetically altered mice can exhibit compensatory signaling cascades that could interfere with our findings. However, this is unlikely in the inducible knock-out model, since GSK-3P can be knocked-down immediately before the experiment. Another useful aspect of pharmacological interventions is that they may be translational, since a small molecule drug could be used in humans to treat heart failure or dyssynchrony.
If echo parameters are not sensitive enough to describe in vivo function (i.e., there are no detectable changes), pressure-volume loops can be conducted in the mice. Capturing PV loops in mice is not a survival surgery, so it can be done only at sacrifice. The procedure provides very sensitive indices of in vivo chamber level cardiac function.
Immune -precipitating GSK-3P, incubating it with tau, and measuring tau p-S199 by
Western blot is also a measure of GSK-3P activity. This correlates with activity determined by p-S9, but may be more sensitive in some cases, and represents a possible alternative. If this is done, GSK-3P is isolated from both myofilament and cytosolic fractions to determine the activity in each pool independently.
Another set of mice is subjected to the same tamoxifen treatment, pacemaker implant, recovery, and two-week pacing protocol as described, although this set would not be sacrificed after two weeks. The pacemaker is turned off at the end of week two, and the mice is returned to synchronous contraction for two additional weeks. Echocardiography and ECG is performed one additional time, two weeks after cessation of pacing, immediately before sacrifice. Tissue is collected, myofilament function assessed, and GSK-3P activity measured as described above (see Figure 33 for a schematic of the protocol). Data analysis is conducted as described above, although since such analysis involves comparison to both baseline and dyssynchronous time points, ANCOVA is used. When the mice are resynchronized (allowed to return to synchrony), the control groups recover, showing calcium re-sensitization as observed in the canine models. The GSK-3pS9A mice are protected from desensitization, so there is no further recovery (Figure 34). In knockout mice, since they cannot re-activate GSK-3P, there is no recovery of calcium sensitivity.
In addition, the pacemaker, as designed, has the capacity for dual chamber pacing, to deliver true bi-ventricular CRT. If resynchronization via pacing cessation doesn't show recovery, an RV and LV lead is implanted, and RV pacing is used for the first two weeks, and Bi-V pacing is used for the second two weeks. Example 12: Determining whether GSK-3fi localizes to the myofilament and is uncoupled from Akt
GSK-3P is inactive when phosphorylated at serine 9. While many kinases are phosphorylate GSK-3P at this residue, Akt is the most common. In dyssynchrony, GSK-3P phosphorylation increases, but Akt activity drops, with the reverse occurring in CRT.
Without being bound by theory, it is believed that there is a pool of GSK-3P bound to the myofilament, which is un-coupled from Akt activity. Indeed, Figure 35 A shows GSK-3P is present in myofilament-enriched samples (using two enrichment protocols, triton and In- Sequence; Kane, L.A., et al, (2007) Methods Mol Biol 357: 87-90). In control dog myocytes, active GSK-3P is present in the myofilament in an alternating pattern with the Z-disk (Figure 35B). When phosphorylated at serine 9 (inactive form), this localization is lost (Figure 35C). Thus, GSK-3P can be determined to be localized in the myofilament using fluorescence confocal, immuno-electron microscopy, and determine its binding partner(s) using, for example, yeast-2 hybrid techniques (Figure 35D).
Activating GSK-3P in dyssynchrony is believed to be beneficial and is believed to preserve myofilament function. However, GSK-3P also increases hypertrophic signaling and its activation would likely have off target effects. However, maintaining activation of a myofilament pool of GSK-3P is a useful therapy. To this end, determining the kinase responsible for de-activating the myofilament pool of GSK-3P is useful. It is believed that two likely candidates are p90rsk and PKCa, since dyssynchrony and CRT (in the dog) altered activity of both in directions commensurate with GSK-3P activity (Figure 36A). Moreover, while Akt has no detectable myofilament targets, and has no effect on myofilament function (Figure 36B), p90rsk and PKCa target myofilament proteins, and PKCa localizes to the myofilament. PKCa and p90rsk are also implicated in mechano-transduction signaling.
Identifying this upstream kinase 1) clarifies the mechanism(s) of mechanical/electrical sensing and signaling in the myocyte, 2) indicates other targets and consequences of cardiac dyssynchrony and resynchronization, and 3) reveals small molecule targets. Thus, important aspects of GSK-3P's regulation in the cardiac myocytes are identified.
In order to determine that there is a myofilament pool of active GSK-3P, localized there through interactions with a myofilament protein, confocal imaging and antibodies (e.g. , those against total GSK-3P, phospho-serine 9 GSK-3P, and a-actinin) are used in, for example, control dog myocytes. Such analyses is also applied to other canine models, dyssynchronous HF, synchronous HF, and CRT. The results will confirm that localization changes in response to various pacing modalities. Confocal imaging is also done in the mouse models described above (e.g., GSK-3P knock-out, GSK-3pS9A, baseline, dyssynchronous pacing, resynchronized, and the like).
Confocal imaging shows the approximate binding of GSK-3P, but more sensitive imaging may be used to specifically locate it in the myofilament. Immuno-electron microscopy is therefore performed on each of the above noted groups to better identify where GSK-3P is binding, and if it specifically translocates to the myofilament in response to dyssynchrony and resynchronization.
In addition, yeast two-hybrid (Y2H) is used to identify the critical proteins to which GSK-3P bind. GSK-3P and GSK-3pS9E (phospho-mimetic S^E, deactivated) are used as bait proteins to probe a human heart library of expressed proteins for interacting protein partners, using, for example, Clontech's Matchmaker Gold Y2H System. Plasmids for these bait proteins are available. Two-Hybrid techniques are similar to those used to identify kinase- myofilament interactions, such as AMPK-Tnl (Oliveira, S.M., et al., (2012) Circ Res
110(9): 1192-201), PKA-TnT (Sumandea, C.A., et al, (2011) J Biol Chem 286(1):530-41), and PKD-MyBPC (Haworth, R.S., et al, (2004) Circ Res,. 95(11): 1091-9.). Differences in protein binding partners obtained from the WT and S9E baits provide insight into the direct mechanisms of binding.
Data is analyzed in a number of ways. For example, some analyses involve quantitative line scans to determine myofilament localization from confocal data. In immuno- EM, many random images are collected, and the number and location of the gold-particle labeled GSK-3P is compared across groups via 1-way ANOVA. The Y2H data reveal a list of the putative interactors with the bait, either active or inactive GSK-3P, or specific amino acid sections thereof.
It is believed that GSK-3P will be localized to the sarcomere and this localization will not change with pacing (control vs. dyssynchrony vs. CRT), but instead be affected by GSK- 3P's phosphorylation status. Thus, the amount of active GSK-3P changes based on pacing, but the active GSK-3P has the same localization. The data described herein indicate that GSK- 3β binds to a thick or thin filament protein, or possibly an M-band protein. The immuno- electron microscopy experiments confirm that, and narrow down exactly where GSK-3P appears. The Y2H indicates the particular proteins to which GSK-3P bind. The S9E GSK-3P bait disrupts this binding.
A substantial amount of GSK-3P is observed in myofilament enriched samples and it is believed that this is myofilament GSK-3P; the confocal staining pattern (Figure 35) could also represent binding to the t-tubular system. Immuno-EM and Y2H confirmation of this results demonstrates that GSK-3P regulates calcium-handling proteins. Phosphorylation levels of calcium handling proteins are then analyzed via Western blots or mass spectrometry techniques described herein. Any lack of possible Y2H interactions between GSK-3P and a myofilament protein could result from: 1) GSK-3P is localized to the myofilament via a secondary binding partner (such as AKAP220), or 2) the myofilament protein GSK-3P interacts with is not currently known or included in the heart protein library. In either case, broader libraries are optionally used. If GSK-3P binds to multiple myofilament proteins, competition experiments are performed in order to determine the more likely binding partner, or to test the ability of GSK-3P to form a complex in the myofilament.
In another embodiment, the kinase responsible phosphorylating and thus inactivating the myofilament pool of GSK-3P is identified. In this process, confirmation of whether Akt is unable to de-activate the myofilament GSK-3P is determined. For example, the activity of PKCa and p90rsk is assessed from the mouse models of dyssynchrony and resynchronization using antibody-based approaches. Next, the kinases are inhibited and/or activated in myocytes from the dog and mice models. Myocytes are isolated, as previously described in both canine (Chakir, K., et al, (2009) Circulation 119(9): 1231-40.) and mouse (Zhang, M., et al, (2012) Circulation 126(8):942-51.) models. Upstream kinases are optionally inhibited using pharmacological inhibitors, such as chelerythrine chloride (general PKC), GO 6976 (PKCa and PKCp specific), LY 333531 hydrochloride (PKCp specific), and BI-D1870 (p90rsk specific [80]). The effect of Akt inhibition is also observed via treatment with Akt Inhibitor XI (Millipore) that does not inhibit any other upstream kinases. Kinase activity is optionally increased by trans fecting with active Akt, PKC, or p90rsk. After inhibition or activation for 24-hours, cells are fixed, stained with total GSK-3P, phosphor-serine 9 GSK-3P, and a- actinin, and observed using a confocal fluorescence microscope. A second pool of cells is myofilament enriched (the cytosolic fraction will also be kept and tested), and changes in GSK-3P phosphorylation are assessed using standard Western blots. Data analysis for some experiments involves GSK-3P localization (via confocal imaging and line scans), and GSK-3P activity in the myo filament-enriched and myo filament-depleted samples. One-way ANOVA is used to compare these data across the multiple groups.
It is believed that PKCa or p90rsk act as a de-activator of myofilament GSK-3P in the dog, and they likely follow the same pattern in the mouse models (increased in dyssynchrony and decreased in resynchronization). Whichever one is primarily responsible increases myo filament-bound GSK-3P phosphorylation, and possibly disrupts the localization pattern. Either inhibition or activation of Akt, on the other hand has no effect on GSK-3P de-activation with dyssynchrony. In mice with S9A, the myocytes are not disrupted by treatment with kinase. Other kinases are also analyzed, such as other PKC isozymes and p70, if neither PKCa nor p90rsk interact with myofilament GSK-3p. In addition, siRNA is used to inhibit kinases if pharmacological inhibition is not specific enough. Example 13: Determining downstream sarcomere targets of GSK-3fi and how they affect myofilament function
Bio-informatics identifies a subset of Z-disk and M-band proteins as GSK-3P targets, which are confirmed in vitro (Table 4). High-value targets include obscurin (S4809), actin- binding LIM protein 1 (abLIMl, S421), and filamin-C (S2228). Obscurin is present in both the Z-disk, where it binds to titin, and the M-band, where it binds to myomesin. abLIMl is also present in the Z-disk, and phosphorylation at S421 is in the region thought to bind to actin. Filamin-C is an actin-binding protein, localized within the Z-disk. Mutations in filamin-C cause DCM, and a splice variant is associated with heart disease. These proteins are schematized in Figure 37, in which the phosphorylation site and known binding domains are highlighted. The phosphorylation sites are genetically modified to contain mutations to mimic constitutive (de) phosphorylation, and transfected into adult rabbit ventricular myocytes.
In order to identify all the myofilament targets of GSK-3P at baseline, with
dyssynchrony, and after resynchronization, and identify the sites conserved across the various species, quantitative mass spectrometry is used since it can identify phosphorylation sites of very low abundance, yet of biological significance. At least two approaches are used to identify the phosphorylation sites of GSK-3P in the myofilament: 1) in vivo phosphorylation sites using the inducible knock-out mouse as a control; and 2) in vitro treatment with the GSK-3P active kinase from dyssynchronous dog and mouse. In each case, tissue is harvested, flash frozen, and enriched for myofilament proteins in the presence of protease and
phosphatase inhibitors. Whether similar targets are identified in the mouse model as in the dog, and/or identify the subset shared by both species, is determined. Using the genetically engineered model, deleting GSK-3P provides a means to definitively test the link between the sarcomere protein phosphorylation changes and the kinase that was not previously possible in the canine model.
A relatively new mass spectrometry (MS) approach known as SWATH is used, in addition or as an alternative to existing mass spectrometry approaches that have been utilized to success (Kirk, J.A., et al., (2014) Journal of Clinical Investigation 124(1): 129-39).
SWATH is based on data-independent acquisition of all observable peptides present in every sample. Based on peptide fragmentation databases that are unique to a particular sample type, thousands of proteins and their modified forms are tracked in every subsequent experimental sample analyzed. Since SWATH provides complete and permanent recording of all peptides in a sample, it is particularly useful when analyzing proteins containing modifiable residues. Thus, it combines the high-throughput of shotgun techniques, and the reproducibility and sensitivity of single/multiple reaction monitoring (SRM/MRM). A 5600 Triple TOF MS is optionally used for the data collection.
For the inducible knock-out mouse, GSK-3pfl/fl/Cre without tamoxifen treatment is compared to those receiving the tamoxifen treatment with Cre but not flox'd GSK-3P since the tamoxifen +MCM model can impact heart function itself. The difference between groups represents GSK-3P targeted phosphorylation. The gradual knock-out and subsequent stabilization period (3-4 weeks) may result in compensatory changes in other kinases. To compensate for this, myofilament samples from GSK-3P knock-out mice are optionally treated with active GSK-3P to determine if this reverses the difference. Data analysis involves quantification of peak area (performed in duplicate) of phospho-peptides between groups. SWATH provides sensitive phospho data without the need for TiO phospho- enrichment, meaning that the phospho form can be normalized to the un-phosphorylated form, providing a sensitive metric.
In addition to expecting previously identified sites to be identified, the genetic mouse models also yields other targets. Similar targets between species enhance their value for pursuing site-mutagenesis studies. This is done with recombinant GSK-3P in rat, showing similar (not all identical) targets to that in dog. Sites found responsible for altering calcium sensitivity are expressed as constitutively phosphorylated in a mouse, and when combined with pacing, they block dyssynchrony-induced calcium desensitization. In another embodiment, the functional significance of specific sites of interest on obscurin, abliml, and filamin-C is determined. For example, primary culture of adult rabbit myocytes is infected with adenoviruses coding for obscurinWT, obscurinS4809E, obscurinS4809A, abLIMl WT, abLIMlS421E, abLIMlS421A, filamin-CWT, filamin-CS2228E, filamin-CS2228A
(produced by the viral vector core). Rabbit is an ideal species, since cardiac myocytes are maintained in cell culture for over a week (unlike mouse), but are easier to work with than dog. The ability to survive in cell culture for a long period is necessary, as some myofilament proteins have long turnover periods, up to a week. Other possible species include guinea pig and cat, which offer similar advantages. The viral expression vectors are optionally tagged, such as with a fluorescent tag (GFP, red-cherry), and tagged vectors optionally serve as negative controls. The myocytes are then prepared for myofilament function experiments as described above. A striated expression pattern is used to identify cells incorporating the mutant protein, and those are used to assess myofilament function. Force-calcium
measurements are obtained before and after exogenous GSK-3P treatment. Western blots using anti-tag antibodies (and obscurin, abLIMl, and filamin-C antibodies), are used to assess percent replacement. Data analysis optionally involves comparing myofilament functional parameters (Fmax, EC50, nH) between the myocytes infected with WT, phospho-mimetic, and phospho-silenced protein, before and after GSK-3P treatment. Thus, two-way ANOVA (group, GSK-3P treatment) is optionally used for statistical analysis.
One or more of the proposed sites affects calcium sensitivity. The phospho-mimetic mutant displays an increase in calcium sensitivity, while the phospho-silenced exhibits a decrease in calcium sensitivity. Moreover, both mutants block the calcium sensitizing effect of GSK-3P treatment, or at least reduce it - depending on the amount of expression of mutant protein. In addition to identifying sites that mediate calcium sensitivity, sites are identified that have other effects on myofilament function. Thus, other possible functional experiments include: stretch-release experiments to measure cross-bridge turnover dynamics (kti); sinusoidal stretch to measure crossbridge stiffness; and in vitro ATPase activity (Kirk, J.A., et ah, (2009) Circ Res 105(12): 1232-9.). In addition to finding the single residue that mediates the
dyssynchrony/CRT-induced calcium sensitivity changes, there are two other possibilities: none of the sites have a functional effect, or more than one does. If these three sites happen to show no functional effect, other sites are explored, such as those listed in Table 4. If multiple sites have a functional effect, neither one completely blocks the effect of GSK-3p. In this case, multiple proteins are transfected at once, to confirm that the multiple sites together can completely reproduce the GSK-3P effect. Listing of Sequences:
Biomarker: Tnl
mRNA Sequence
NCBI Reference Sequence: NM_000363.4
SEQ ID NO: 1
AGTGTCCTCGGGGAGTCTCAAGCAGCCCGGAGGAGACTGACGGTCCCTGGGACCCTGAAGGTCACCCGGG CGGCCCCCTCACTGACCCTCCAAACGCCCCTGTCCTCGCCCTGCCTCCTGCCATTCCCGGCCTGAGTCTC AGCATGGCGGATGGGAGCAGCGATGCGGCTAGGGAACCTCGCCCTGCACCAGCCCCAATCAGACGCCGCT CCTCCAACTACCGCGCTTATGCCACGGAGCCGCACGCCAAGAAAAAATCTAAGATCTCCGCCTCGAGAAA ATTGCAGCTGAAGACTCTGCTGCTGCAGATTGCAAAGCAAGAGCTGGAGCGAGAGGCGGAGGAGCGGCGC GGAGAGAAGGGGCGCGCTCTGAGCACCCGCTGCCAGCCGCTGGAGTTGGCCGGGCTGGGCTTCGCGGAGC TGCAGGACTTGTGCCGACAGCTCCACGCCCGTGTGGACAAGGTGGATGAAGAGAGATACGACATAGAGGC AAAAGTCACCAAGAACATCACGGAGATTGCAGATCTGACTCAGAAGATCTTTGACCTTCGAGGCAAGTTT AAGCGGCCCACCCTGCGGAGAGTGAGGATCTCTGCAGATGCCATGATGCAGGCGCTGCTGGGGGCCCGGG CTAAGGAGTCCCTGGACCTGCGGGCCCACCTCAAGCAGGTGAAGAAGGAGGACACCGAGAAGGAAAACCG GGAGGTGGGAGACTGGCGCAAGAACATCGATGCACTGAGTGGAATGGAGGGCCGCAAGAAAAAGTTTGAG AGCTGAGCCTTCCTGCCTACTGCCCCTGCCCTGAGGAGGGCCCTGAGGAATAAAGCTTCTCTCTGAGCTG AAAAAAAAAAAAAAAAAAAAAAAAAA Amino Acid Sequence
NCBI Reference Sequence: NP_000354.4
SEQ ID NO: 2
MADGSSDAAREPRPAPAPIRRRSSNYRAYATEPHAKKKSKISASRKLQLKTLLLQIAKQELEREAEERRG EKGRALSTRCQPLELAGLGFAELQDLCRQLHARVDKVDEERYDIEAKVTKNITEIADLTQKIFDLRGKFK RPTLRRVRISADAMMQALLGARAKESLDLRAHLKQVKKEDTEKENREVGDWRKNIDALSGMEGRKKKFES
Biomarker: MyBPC
mRNA Sequence
GenBank: BC136389.1
SEQ ID NO: 3
AGGAGGAGGTCCCCCGACATGCCTGAGGCAAAACCAGCGGCCAAAAAGGCCCCCAAAGGCAAAGATGCCC CCAAAGGAGCCCCCAAGGAGGCTCCCCCTAAGGAGGCTCCTGCAGAGGCCCCCAAAGAAGCCCCACCCGA GGACCAGTCCCCGACTGCAGAGGAGCCCACCGGCGTTTTCCTGAAGAAGCCGGACTCCGTCTCAGTGGAG ACTGGGAAGGACGCAGTGGTCGTGGCCAAGGTGAACGGGAAGGAGCTCCCAGACAAACCGACCATCAAGT GGTTCAAGGGGAAGTGGCTGGAGCTGGGCAGCAAGAGTGGCGCCCGCTTCTCCTTCAAGGAGTCCCACAA CTCCGCCAGCAATGTGTACACCGTGGAGCTGCACATTGGGAAGGTGGTACTGGGGGACCGTGGGTATTAC CGCCTCGAGGTCAAAGCCAAGGACACCTGTGACAGCTGTGGCTTCAACATCGATGTGGAGGCACCCCGTC AGGATGCCTCTGGGCAGAGTCTAGAAAGCTTCAAGCGTACGAGTGAAAAGAAGTCGGATACTGCAGGTGA GCTGGATTTCAGTGGCCTGTTGAAGAAGAGGGAGGTGGTGGAGGAGGAGAAGAAGAAGAAAAAGAAAGAT GACGATGACCTAGGCATCCCCCCGGAGATTTGGGAGCTCCTGAAAGGGGCAAAGAAGAGCGAGTACGAGA AAATCGCCTTCCAGTATGGCATCACCGACCTCCGAGGCATGCTGAAGCGGCTGAAAAAGGCTAAGGTCGA GGTCAAGAAGAGTGCAGCATTCACAAAGAAGCTGGATCCAGCCTACCAAGTGGACAGAGGCAACAAGATC AAGTTGATGGTAGAGATCAGCGACCCAGACCTGACCCTCAAGTGGTTCAAGAACGGCCAGGAGATCAAAC CAAGCAGCAAGTACGTGTTTGAGAACGTTGGTAAGAAGCGAATTCTTACCATCAACAAGTGCACGCTGGC GGATGACGCTGCCTATGAAGTAGCTGTCAAGGATGAGAAGTGTTTCACCGAGCTCTTCGTCAAAGAACCT CCAGTCCTAATTGTCACACCTCTTGAGGACCAGCAGGTGTTTGTGGGTGACCGGGTGGAAATGGCAGTGG AGGTGTCAGAAGAGGGTGCCCAGGTGATGTGGATGAAAGATGGTGTGGAACTGACTCGGGAGGATTCCTT CAAGGCCCGGTACCGCTTCAAGAAGGACGGGAAGCGCCACATCCTCATCTTCTCAGACGTGGTCCAGGAG GACAGGGGTCGCTATCAGGTCATAACCAATGGCGGCCAGTGTGAGGCCGAGCTGATTGTGGAAGAGAAAC AGCTGGAGGTCCTGCAGGACATCGCGGATCTGACGGTGAAGGCCTCAGAACAAGCTGTGTTCAAGTGCGA GGTGTCTGATGAGAAAGTGACGGGCAAGTGGTATAAGAATGGGGTCGAGGTGCGGCCCAGCAAGAGGATC ACCATTTCCCATGTAGGCAGGTTCCACAAGCTGGTGATCGATGACGTCCGCCCCGAGGATGAGGGAGACT ACACGTTTGTGCCTGACGGCTACGCCCTGTCGCTCTCGGCCAAGCTCAACTTCCTGGAAATCAAGGTGGA GTACGTTCCCAAGCAAGAGCCACCAAAGATCCACTTGGATTGCTCGGGGAAGACCTCAGAGAATGCGATT GTGGTTGTGGCTGGAAACAAGCTGAGGCTTGACGTGTCCATCACAGGGGAGCCCCCTCCCGTCGCTACCT GGCTGAAGGGAGATGAGGTATTCACGACCACCGAGGGCAGGACCCGCATCGAGAAGCGGGTGGACTGCAG CAGCTTTGTGATTGAGAGTGCGCAGCGGGAAGACGAGGGCCGCTACACCATCAAGGTCACCAACCCCGTC GGCGAGGACGTGGCTTCCATCTTCCTGCAAGTTGTAGATGTCCCAGACCCCCCGGAGGCTGTGCGCATCA CCTCGGTTGGAGAGGATTGGGCCATCCTTGTCTGGGAGCCACCAATGTACGATGGGGGGAAGCCAGTCAC CGGGTACCTCGTAGAGCGGAAGAAGAAGGGCTCTCAGCGCTGGATGAAGCTGAACTTTGAGGTCTTCACA GAGACCACCTATGAGTCCACCAAGATGATCGAGGGCATCCTCTATGAGATGCGTGTCTTCGCCGTCAATG CTATAGGGGTCTCCCAGCCCAGCATGAACACCAAGCCTTTTATGCCTATTGCACCCACGAGTGAACCCCT GCACCTGATAGTGGAGGATGTGACAGACACCACCACCACACTCAAGTGGAGGCCTCCGAACAGGATCGGG GCAGGTGGCATCGATGGGTACCTGGTGGAGTACTGCCTGGAAGGCTCCGAGGAATGGGTCCCTGCCAACA CCGAGCCCGTGGAGCGCTGTGGCTTCACCGTCAAGAATCTCCCGACCGGAGCCAGAATCCTCTTCCGAGT AGTTGGGGTCAACATCGCGGGGCGCAGCGAGCCGGCCACCCTGGCCCAGCCGGTCACCATCAGGGAGATT GCGGAGCCACCCAAGATCCGGCTTCCCCGCCATCTCCGCCAGACCTACATCCGCAAAGTGGGCGAGCAGC TCAACCTTGTCGTCCCCTTCCAGGGAAAGCCCCGGCCCCAGGTGGTGTGGACCAAGGGCGGGGCCCCGCT GGACACCTCCCGCGTGCACGTGCGGACCAGCGACTTCGACACCGTGTTCTTCGTGCGCCAGGCGGCCCGC TCCGACTCCGGGGAGTACGAGCTGAGCGTGCAGATCGAGAACATGAAGGACACCGCCACCATCCGCATCC GCGTTGTGGAAAAGGCTGGGCCCCCCATAAACGTGATGGTGAAGGAGGTGTGGGGCACGAACGCGCTGGT GGAGTGGCAGGCCCCCAAAGATGATGGGAACAGTGAGATCATGGGGTATTTCGTCCAGAAAGCAGACAAA AAAACCATGGAGTGGTTCAACGTCTATGAACGTAACAGGCACACTAGCTGTACTGTGTCCGACCTTATCG TGGGCAATGAATACTATTTCCGAGTTTACACCGAGAACATCTGTGGGCTCAGTGACTCACCTGGTGTCTC CAAGAACACGGCCCGCATCCTCAAGACAGGAATCACCTTCAAACCGTTCGAGTATAAGGAGCATGACTTC CGGATGGCTCCCAAGTTCCTGACACCTCTCATAGACCGCGTGGTCGTGGCTGGGTACTCGGCAGCCCTCA ACTGCGCTGTCAGAGGCCACCCGAAGCCGAAGGTGGTCTGGATGAAGAACAAGATGGAAATCCGTGAAGA TCCCAAGTTCCTGATAACCAATTACCAAGGAGTCCTGACGCTGAACATCCGTCGCCCCTCGCCCTTCGAC GCTGGGACTTACACCTGCCGGGCCGTCAACGAGCTGGGCGAGGCGCTGGCTGAGTGCAAGCTGGAGGTCC GAGTGCCGCAGTGAGACCTGTCCCCTACCTGCCAA Amino Acid Sequence
NCBI Reference Sequence: NP_004524.3
SEQ ID NO: 4
MPEAKPAAKKAPKGKDAPKGAPKEAPPKEAPAEAPKEAPPEDQSPTAEEPTGVFLKKPDSVSVETGKDAV WAKVNGKELPDKPTIKWFKGKWLELGSKSGARFSFKESHNSASNVYTVELHIGKWLGDRGYYRLEVKA KDTCDSCGFNIDVEAPRQDASGQSLESFKRTSEKKSDTAGELDFSGLLKKREWEEEKKKKKKDDDDLGI PPEIWELLKGAKKSEYEKIAFQYGITDLRGMLKRLKKAKVEVKKSAAFTKKLDPAYQVDRGNKIKLMVEI SDPDLTLKWFKNGQEIKPSSKYVFENVGKKRILTINKCTLADDAAYEVAVKDEKCFTELFVKEPPVLIVT PLEDQQVFVGDRVEMAVEVSEEGAQVMWMKDGVELTREDSFKARYRFKKDGKRHILIFSDWQEDRGRYQ VITNGGQCEAELIVEEKQLEVLQDIADLTVKASEQAVFKCEVSDEKVTGKWYKNGVEVRPSKRITISHVG RFHKLVIDDVRPEDEGDYTFVPDGYALSLSAKLNFLEIKVEYVPKQEPPKIHLDCSGKTSENAIVWAGN KLRLDVSITGEPPPVATWLKGDEVFTTTEGRTRIEKRVDCSSFVIESAQREDEGRYTIKVTNPVGEDVAS IFLQWDVPDPPEAVRITSVGEDWAILVWEPPMYDGGKPVTGYLVERKKKGSQRWMKLNFEVFTETTYES TKMIEGILYEMRVFAVNAIGVSQPSMNTKPFMPIAPTSEPLHLIVEDVTDTTTTLKWRPPNRIGAGGIDG YLVEYCLEGSEEWVPANTEPVERCGFTVKNLPTGARILFRWGVNIAGRSEPATLAQPVTIREIAEPPKI RLPRHLRQTYIRKVGEQLNLWPFQGKPRPQWWTKGGAPLDTSRVHVRTSDFDTVFFVRQAARSDSGEY ELSVQIENMKDTATIRIRWEKAGPPINVMVKEVWGTNALVEWQAPKDDGNSEIMGYFVQKADKKTMEWF NVYERNRHTSCTVSDLIVGNEYYFRVYTENICGLSDSPGVSKNTARILKTGITFKPFEYKEHDFRMAPKF LTPLIDRVWAGYSAALNCAVRGHPKPKWWMKNKMEIREDPKFLITNYQGVLTLNIRRPSPFDAGTYTC RAVNELGEALAECKLEVRVPQ
Biomarker: TnT
mRNA Sequence
GenBank: S64668.1
SEQ ID NO: 5
AGAGCAGAGACCATGTCTGACATAGAAGAGGTGGTGGAAGAGTACGAGGAGGAGGAGCAGGAAGAAGCAG CTGTTGAAGAGCAGGAGGAGGCAGCGGAAGAGGATGCTGAAGCAGAGGCTGAGACCGAGGAGACCAGGGC AGAAGAAGATGAAGAAGAAGAGGAAGCAAAGGAGGCTGAAGATGGCCCAATGGAGGAGTCCAAACCAAAG CCCAGGTCGTTCATGCCCAACTTGGTGCCTCCCAAGATCCCCGATGGAGAGAGAGTGGACTTTGATGACA TCCACCGGAAGCGCATGGAGAAGGACCTGAATGAGTTGCAGGCGCTGATTGAGGCTCACTTTGAGAACAG GAAGAAAGAGGAGGAGGAGCTCGTTTCTCTCAAAGACAGGATCGAGAAACGTCGGGCAGAGCGGGCCGAG CAGCAGCGCATCCGGAATGAGCGGGAGAAGGAGCGGCAGAACCGCCTGGCTGAAGAGAGGGCTCGACGAG AGGAGGAGGAGAACAGGAGGAAGGCTGAGGATGAGGCCCGGAAGAAGAAGGCTTTGTCCAACATGATGCA TTTTGGGGGTTACATCCAGAAGCAGGCCCAGACAGAGCGGAAAAGTGGGAAGAGGCAGACTGAGCGGGAA AAGAAGAAGAAGATTCTGGCTGAGAGGAGGAAGGTGCTGGCCATTGACCACCTGAATGAAGATCAGCTGA GGGAGAAGGCCAAGGAGCTGTGGCAGACGATCTATAACTTGGAGGCAGAGAAGTTCGACCTGCAGGAGAA GTTCAAGCAGCAGAAA ATGAGATCAATGTTCTCCGAAACAGGATCAACGATAACCAGAAAGTCTCCAAG ACCCGCGGGAAGGCTAAAGTCACCGGGCGCTGGAAATAGAGCCTGGCCTCCTTCACCAAAGATCTGCTCC TCGCTCGCACCTGCCTCCGCTGCACTCCCCCAGTTCCCGGGCCCTCCTGGGCACCCCAGGCAGCTCCTGT TTGGAAATGGGGAGCTGGCCTAGTGGGAGCCACCACTCCTGCCTGCCCCCACACCCACTCCACACCAGTA ATAAAAAGCCACCACACACTGAAAAAAAAAAAAAAA Amino Acid Sequence
GenBank: AAB27731.1
SEQ ID NO: 6
MSDIEEWEEYEEEEQEEAAVEEQEEAAEEDAEAEAETEETRAEEDEEEEEAKEAEDGPMEESKPKPRSF MPNLVPPKIPDGERVDFDDIHRKRMEKDLNELQALIEAHFENRKKEEEELVSLKDRIEKRRAERAEQQRI RNEREKERQNRLAEERARREEEENRRKAEDEARKKKALSNMMHFGGYIQKQAQTERKSGKRQTEREKKKK ILAERRKVLAIDHLNEDQLREKAKELWQTIYNLEAEKFDLQEKFKQQKYEINVLRNRINDNQKVSKTRGK AKVTGRWK
Biomarker: MLC2
mRNA Sequence
NCBI Reference Sequence: NM_000432.3
SEQ ID NO: 7
GTTCCTGGGCTGCAGAGAGCTGGGCGGAGTGTGGAATTCTTCTCGGGAGGCAGTGCTGGGTCCTTTCCAC CATGGCACCTAAGAAAGCAAAGAAGAGAGCCGGGGGCGCCAACTCCAACGTGTTCTCCATGTTCGAACAG ACCCAAATCCAGGAATTTAAGGAGGCCTTCACTATCATGGACCAGAACAGGGATGGCTTCATTGACAAGA ACGATCTGAGAGACACCTTTGCTGCCCTTGGGCGAGTGAACGTGAAAAATGAAGAAATTGATGAAATGAT CAAGGAGGCTCCGGGTCCAATTAACTTTACTGTGTTCCTCACAATGTTTGGGGAGAAACTTAAGGGAGCG GACCCTGAGGAAACCATTCTCAACGCATTCAAAGTGTTTGACCCTGAAGGCAAAGGGGTGCTGAAGGCTG ATTACGTTCGGGAAATGCTGACCACGCAGGCGGAGAGGTTTTCCAAGGAGGAGGTTGACCAGATGTTCGC CGCCTTCCCCCCTGACGTGACTGGCAACTTGGACTACAAGAACCTGGTGCACATCATCACCCACGGAGAA GAGAAGGACTAGGAGGGGGCTCGCTGCTGCGCCCTGGGCTCGTCTTTGCAGAGTGGTCCCTGCCCTCATC TCTCTCCCCCGAGTACCGCCTCTGTCCCTACCTTGTCTGTTAGCCATGTGGCTGCCCCATTTATCCACCT CCATCTTCTTTGCAGCCTGGGTGGCTATGGGTACTTCGTGGCCGCACATCCTACAGTTGGAAATCCATCC AGAGGCCATGTTCCAATAAACAGGAGGTCGTGTATTTGGTCACGACATTTCTCTGACAAAAAAAAAAAAA AAAAAAAAAAAAAAA
Amino Acid Sequence
NCBI Reference Sequence: NP_000423.2
SEQ ID NO: 8
MAPKKAKKRAGGANSNVFSMFEQTQIQEFKEAFTIMDQNRDGFIDKNDLRDTFAALGRVNVKNEEIDEMI KEAPGPINFTVFLTMFGEKLKGADPEETILNAFKVFDPEGKGVLKADYVREMLTTQAERFSKEEVDQMFA AFPPDVTGNLDYKNLVHI ITHGEEKD
Biomarker: cMyBP-C
mRNA Sequence
NCBI Reference Sequence: NM_000256.3
SEQ ID NO: 9 AGTCCCTCTTTGGGTGACCTGTGCCTGCTTCGTGCCTGGTGTGACGTCTCTCAGGATGCCTGAGCCGGGG AAGAAGCCAGTCTCAGCTTTTAGCAAGAAGCCACGGTCAGTGGAAGTGGCCGCAGGCAGCCCTGCCGTGT TCGAGGCCGAGACAGAGCGGGCAGGAGTGAAGGTGCGCTGGCAGCGCGGAGGCAGTGACATCAGCGCCAG CAACAAGTACGGCCTGGCCACAGAGGGCACACGGCATACGCTGACAGTGCGGGAAGTGGGCCCTGCCGAC CAGGGATCTTACGCAGTCATTGCTGGCTCCTCCAAGGTCAAGTTCGACCTCAAGGTCATAGAGGCAGAGA AGGCAGAGCCCATGCTGGCCCCTGCCCCTGCCCCTGCTGAGGCCACTGGAGCCCCTGGAGAAGCCCCGGC CCCAGCCGCTGAGCTGGGAGAAAGTGCCCCAAGTCCCAAAGGGTCAAGCTCAGCAGCTCTCAATGGTCCT ACCCCTGGAGCCCCCGATGACCCCATTGGCCTCTTCGTGATGCGGCCACAGGATGGCGAGGTGACCGTGG GTGGCAGCATCACCTTCTCAGCCCGCGTGGCCGGCGCCAGCCTCCTGAAGCCGCCTGTGGTCAAGTGGTT CAAGGGCAAATGGGTGGACCTGAGCAGCAAGGTGGGCCAGCACCTGCAGCTGCACGACAGCTACGACCGC GCCAGCAAGGTCTATCTGTTCGAGCTGCACATCACCGATGCCCAGCCTGCCTTCACTGGCAGCTACCGCT GTGAGGTGTCCACCAAGGACAAATTTGACTGCTCCAACTTCAATCTCACTGTCCACGAGGCCATGGGCAC CGGAGACCTGGACCTCCTATCAGCCTTCCGCCGCACGAGCCTGGCTGGAGGTGGTCGGCGGATCAGTGAT AGCCATGAGGACACTGGGATTCTGGACTTCAGCTCACTGCTGAAAAAGAGAGACAGTTTCCGGACCCCGA GGGACTCGAAGCTGGAGGCACCAGCAGAGGAGGACGTGTGGGAGATCCTACGGCAGGCACCCCCATCTGA GTACGAGCGCATCGCCTTCCAGTACGGCGTCACTGACCTGCGCGGCATGCTAAAGAGGCTCAAGGGCATG AGGCGCGATGAGAAGAAGAGCACAGCCTTTCAGAAGAAGCTGGAGCCGGCCTACCAGGTGAGCAAAGGCC ACAAGATCCGGCTGACCGTGGAACTGGCTGACCATGACGCTGAGGTCAAATGGCTCAAGAATGGCCAGGA GATCCAGATGAGCGGCAGCAAGTACATCTTTGAGTCCATCGGTGCCAAGCGTACCCTGACCATCAGCCAG TGCTCATTGGCGGACGACGCAGCCTACCAGTGCGTGGTGGGTGGCGAGAAGTGTAGCACGGAGCTCTTTG TGAAAGAGCCCCCTGTGCTCATCACGCGCCCCTTGGAGGACCAGCTGGTGATGGTGGGGCAGCGGGTGGA GTTTGAGTGTGAAGTATCGGAGGAGGGGGCGCAAGTCAAATGGCTGAAGGACGGGGTGGAGCTGACCCGG GAGGAGACCTTCAAATACCGGTTCAAGAAGGACGGGCAGAGACACCACCTGATCATCAACGAGGCCATGC TGGAGGACGCGGGGCACTATGCACTGTGCACTAGCGGGGGCCAGGCGCTGGCTGAGCTCATTGTGCAGGA AAAGAAGCTGGAGGTGTACCAGAGCATCGCAGACCTGATGGTGGGCGCAAAGGACCAGGCGGTGTTCAAA TGTGAGGTCTCAGATGAGAATGTTCGGGGTGTGTGGCTGAAGAATGGGAAGGAGCTGGTGCCCGACAGCC GCATAAAGGTGTCCCACATCGGGCGGGTCCACAAACTGACCATTGACGACGTCACACCTGCCGACGAGGC TGACTACAGCTTTGTGCCCGAGGGCTTCGCCTGCAACCTGTCAGCCAAGCTCCACTTCATGGAGGTCAAG ATTGACTTCGTACCCAGGCAGGAACCTCCCAAGATCCACCTGGACTGCCCAGGCCGCATACCAGACACCA TTGTGGTTGTAGCTGGAAATAAGCTACGTCTGGACGTCCCTATCTCTGGGGACCCTGCTCCCACTGTGAT CTGGCAGAAGGCTATCACGCAGGGGAATAAGGCCCCAGCCAGGCCAGCCCCAGATGCCCCAGAGGACACA GGTGACAGCGATGAGTGGGTGTTTGACAAGAAGCTGCTGTGTGAGACCGAGGGCCGGGTCCGCGTGGAGA CCACCAAGGACCGCAGCATCTTCACGGTCGAGGGGGCAGAGAAGGAAGATGAGGGCGTCTACACGGTCAC AGTGAAGAACCCTGTGGGCGAGGACCAGGTCAACCTCACAGTCAAGGTCATCGACGTGCCAGACGCACCT GCGGCCCCCAAGATCAGCAACGTGGGAGAGGACTCCTGCACAGTACAGTGGGAGCCGCCTGCCTACGATG GCGGGCAGCCCATCCTGGGCTACATCCTGGAGCGCAAGAAGAAGAAGAGCTACCGGTGGATGCGGCTGAA CTTCGACCTGATTCAGGAGCTGAGTCATGAAGCGCGGCGCATGATCGAGGGCGTGGTGTACGAGATGCGC GTCTACGCGGTCAACGCCATCGGCATGTCCAGGCCCAGCCCTGCCTCCCAGCCCTTCATGCCTATCGGTC CCCCCAGCGAACCCACCCACCTGGCAGTAGAGGACGTCTCTGACACCACGGTCTCCCTCAAGTGGCGGCC CCCAGAGCGCGTGGGAGCAGGAGGCCTGGATGGCTACAGCGTGGAGTACTGCCCAGAGGGCTGCTCAGAG TGGGTGGCTGCCCTGCAGGGGCTGACAGAGCACACATCGATACTGGTGAAGGACCTGCCCACGGGGGCCC GGCTGCTTTTCCGAGTGCGGGCACACAATATGGCAGGGCCTGGAGCCCCTGTTACCACCACGGAGCCGGT GACAGTGCAGGAGATCCTGCAACGGCCACGGCTTCAGCTGCCCAGGCACCTGCGCCAGACCATTCAGAAG AAGGTCGGGGAGCCTGTGAACCTTCTCATCCCTTTCCAGGGCAAGCCCCGGCCTCAGGTGACCTGGACCA AAGAGGGGCAGCCCCTGGCAGGCGAGGAGGTGAGCATCCGCAACAGCCCCACAGACACCATCCTGTTCAT CCGGGCCGCTCGCCGCGTGCATTCAGGCACTTACCAGGTGACGGTGCGCATTGAGAACATGGAGGACAAG GCCACGCTGGTGCTGCAGGTTGTTGACAAGCCAAGTCCTCCCCAGGATCTCCGGGTGACTGACGCCTGGG GTCTTAATGTGGCTCTGGAGTGGAAGCCACCCCAGGATGTCGGCAACACGGAGCTCTGGGGGTACACAGT GCAGAAAGCCGACAAGAAGACCATGGAGTGGTTCACCGTCTTGGAGCATTACCGCCGCACCCACTGCGTG GTGCCAGAGCTCATCATTGGCAATGGCTACTACTTCCGCGTCTTCAGCCAGAATATGGTTGGCTTTAGTG ACAGAGCGGCCACCACCAAGGAGCCCGTCTTTATCCCCAGACCAGGCATCACCTATGAGCCACCCAACTA TAAGGCCCTGGACTTCTCCGAGGCCCCAAGCTTCACCCAGCCCCTGGTGAACCGCTCGGTCATCGCGGGC TACACTGCTATGCTCTGCTGTGCTGTCCGGGGTAGCCCCAAGCCCAAGATTTCCTGGTTCAAGAATGGCC TGGACCTGGGAGAAGACGCCCGCTTCCGCATGTTCAGCAAGCAGGGAGTGTTGACTCTGGAGATTAGAAA GCCCTGCCCCTTTGACGGGGGCATCTATGTCTGCAGGGCCACCAACTTACAGGGCGAGGCACGGTGTGAG TGCCGCCTGGAGGTGCGAGTGCCTCAGTGACCAGGCTGGCTCCTGGGGATGGCCAGGTACAACCGGATGC CAGCCCCGTGCCAGGAGCCTGGAGGGAAGTTGGGGAAACCCCTCCCTACTGTTGGATGTATGTGTGACAA GTGTGTCTCCTGTGCTGCGATGGGGGATCAGCAGGGCAGTTGTCGGGCAGTCCTGAGTGGGTGTTGCACA GACTGGTCCACAGGGCTCCTGAAGGAAGCCCCTGGATCTTTGGGGTAAAAGGAGGGTGGCCTCAAGAAAC AATGTCTGGGGACAGGCCTTTCTGGCCTGCTATGTCTTCCCAATGTTTATTGGGCAATAAAAGATAAGTG CAGTCACAGAGAACTCA
Amino Acid Sequence
NCBI Reference Sequence: NP_000247.2
SEQ ID NO: 10
MPEPGKKPVSAFSKKPRSVEVAAGSPAVFEAETERAGVKVRWQRGGSDISASNKYGLATEGTRHTLTVRE VGPADQGSYAVIAGSSKVKFDLKVIEAEKAEPMLAPAPAPAEATGAPGEAPAPAAELGESAPSPKGSSSA ALNGPTPGAPDDPIGLFVMRPQDGEVTVGGSITFSARVAGASLLKPPWKWFKGKWVDLSSKVGQHLQLH DSYDRASKVYLFELHITDAQPAFTGSYRCEVSTKDKFDCSNFNLTVHEAMGTGDLDLLSAFRRTSLAGGG RRISDSHEDTGILDFSSLLKKRDSFRTPRDSKLEAPAEEDVWEILRQAPPSEYERIAFQYGVTDLRGMLK RLKGMRRDEKKSTAFQKKLEPAYQVSKGHKIRLTVELADHDAEVKWLKNGQEIQMSGSKYIFESIGAKRT LTISQCSLADDAAYQCWGGEKCSTELFVKEPPVLITRPLEDQLVMVGQRVEFECEVSEEGAQVKWLKDG VELTREETFKYRFKKDGQRHHLI INEAMLEDAGHYALC SGGQALAELIVQEKKLEVYQSIADLMVGAKD QAVFKCEVSDENVRGVWLKNGKELVPDSRIKVSHIGRVHKLTIDDVTPADEADYSFVPEGFACNLSAKLH FMEVKIDFVPRQEPPKIHLDCPGRIPDTIVWAGNKLRLDVPISGDPAPTVIWQKAITQGNKAPARPAPD APEDTGDSDEWVFDKKLLCETEGRVRVETTKDRSIFTVEGAEKEDEGVYTVTVKNPVGEDQVNLTVKVID VPDAPAAPKISNVGEDSCTVQWEPPAYDGGQPILGYILERKKKKSYRWMRLNFDLIQELSHEARRMIEGV VYEMRVYAVNAIGMSRPSPASQPFMPIGPPSEPTHLAVEDVSDTTVSLKWRPPERVGAGGLDGYSVEYCP EGCSEWVAALQGLTEHTSILVKDLPTGARLLFRVRAHNMAGPGAPVTTTEPVTVQEILQRPRLQLPRHLR QTIQKKVGEPVNLLIPFQGKPRPQVTWTKEGQPLAGEEVSIRNSPTDTILFIRAARRVHSGTYQVTVRIE NMEDKATLVLQWDKPSPPQDLRVTDAWGLNVALEWKPPQDVGNTELWGYTVQKADKKTMEWFTVLEHYR RTHCWPELI IGNGYYFRVFSQNMVGFSDRAATTKEPVFIPRPGI YEPPNYKALDFSEAPSFTQPLVNR SVIAGYTAMLCCAVRGSPKPKISWFKNGLDLGEDARFRMFSKQGVLTLEIRKPCPFDGGIYVCRATNLQG EARCECRLEVRVPQ
Biomarker: cTnl
mRNA Sequence
NCBI Reference Sequence: NM_000364.3
SEQ ID NO: 11
CTCCATCCTCTGCCTCACCCAGTCCCCGCTGAGACTGAGCAGACGCCTCCAGGATCTGTCGGCAGCTGCT GTTCTGAGGGAGAGCAGAGACCATGTCTGACATAGAAGAGGTGGTGGAAGAGTACGAGGAGGAGGAGCAG GAAGAAGCAGCTGTTGAAGAAGAGGAGGACTGGAGAGAGGACGAAGACGAGCAGGAGGAGGCAGCGGAAG AGGATGCTGAAGCAGAGGCTGAGACCGAGGAGACCAGGGCAGAAGAAGATGAAGAAGAAGAGGAAGCAAA GGAGGCTGAAGATGGCCCAATGGAGGAGTCCAAACCAAAGCCCAGGTCGTTCATGCCCAACTTGGTGCCT CCCAAGATCCCCGATGGAGAGAGAGTGGACTTTGATGACATCCACCGGAAGCGCATGGAGAAGGACCTGA ATGAGTTGCAGGCGCTGATCGAGGCTCACTTTGAGAACAGGAAGAAAGAGGAGGAGGAGCTCGTTTCTCT CAAAGACAGGATCGAGAGACGTCGGGCAGAGCGGGCCGAGCAGCAGCGCATCCGGAATGAGCGGGAGAAG GAGCGGCAGAACCGCCTGGCTGAAGAGAGGGCTCGACGAGAGGAGGAGGAGAACAGGAGGAAGGCTGAGG ATGAGGCCCGGAAGAAGAAGGCTTTGTCCAACATGATGCATTTTGGGGGTTACATCCAGAAGACAGAGCG GAAAAGTGGGAAGAGGCAGACTGAGCGGGAAAAGAAGAAGAAGATTCTGGCTGAGAGGAGGAAGGTGCTG GCCATTGACCACCTGAATGAAGATCAGCTGAGGGAGAAGGCCAAGGAGCTGTGGCAGAGCATCTATAACT TGGAGGCAGAGAAGTTCGACCTGCAGGAGAAGTTCAAGCAGCAGAAATATGAGATCAATGTTCTCCGAAA CAGGATCAACGATAACCAGAAAGTCTCCAAGACCCGCGGGAAGGCTAAAGTCACCGGGCGCTGGAAATAG AGCCTGGCCTCCTTCACCAAAGATCTGCTCCTCGCTCGCACCTGCCTCCGGCCTGCACTCCCCCAGTTCC CGGGCCCTCCTGGGCACCCCAGGCAGCTCCTGTTTGGAAATGGGGAGCTGGCCTAGGTGGGAGCCACCAC TCCTGCCTGCCCCCACACCCACTCCACACCAGTAATAAAAAGCCACCACACACTGACTGGCAAAAAAAAA AAAAA
Amino Acid Sequence
NCBI Reference Sequence: NP_000355.2
SEQ ID NO: 12
MSDIEEWEEYEEEEQEEAAVEEEEDWREDEDEQEEAAEEDAEAEAETEETRAEEDEEEEEAKEAEDGPM EESKPKPRSFMPNLVPPKIPDGERVDFDDIHRKRMEKDLNELQALIEAHFENRKKEEEELVSLKDRIERR RAERAEQQRIRNEREKERQNRLAEERARREEEENRRKAEDEARKKKALSNMMHFGGYIQKTERKSGKRQT EREKKKKILAERRKVLAIDHLNEDQLREKAKELWQSIYNLEAEKFDLQEKFKQQKYEINVLRNRINDNQK VSKTRGKAKVTGRWK
Biomarker: -Tropomyosin
mRNA Sequence
GenBank: AJ001055.1
SEQ ID NO: 13
AATCGGGCTGAGTTTGCGGAGAGGTCAGTAACTAAATTGGAGAAAAGCATTGATGACTTAGAAGAGAAAG TGGCTCATGCCAAAGAAGAAAACCTTAGTATGCATCAGATGCTGGATCAGACTTTACTGGAGTTAAACAA CATGTGAAAACCTCCTTAGCTGCGACCACATTCTTTCATTTTGTTTTGTTTTGTTTTGTTTTGTTTTTAA ACACCTGCTTACCCCTTAAATGCAATTTATTTACTTTTACCACTGTCACAGAAACATCCACAAGATACCA GCTAGGTCAGGGGGTGGGGAAAACACATACAAAANGGNAAGCCCATGTCAGGGCGATCCTGGNTCAAATT GTGCCATTTCCCGGGTTGGATGCTTGCCACACTTGGTGGAGAGTTTAGNAACACAGNGTGCTTAGTGGAA TCCTCACTAAAGCAGGAGAAGTTCCATTCAAAGTGCCAATGATAGAGTCAACAAGGAAGGTTAATGTTGG AAACACAATCAGGTGTGGATTGGTGCTACTTTGAACAAAAGGTCCCCCTGTGGTCTTTTGTTCAACATTG TACAATGTAGAACTCTGTCCAACACTAATTTATTTTGTCTTGAGTTTTACTACAAGATGAGACTATGGAT CCCGCATGCCTGAATTCACTAAAGCCCGAGGGGCTGTAAGCCCCGCTGCTCGTCTGAGACTTCCATTCCT TTCTGATTGGCACACGTGCAGCTCCTGNCAATCTGTAGGATACCAATCAGTGTGGATTTCCACTCTTTTC AGTCCNTCATGTTCNAGATTTAGACACCACANACAACTGGTNAAGGNCGTTTTCTTGAGAGTTTTANCTA TATGTANACATTGTATAATGATATGGAATAAAATGNACATTGTAGGACATTTTCTAAAAAAAAAAA
Amino Acid Sequence
NCBI Reference Sequence: NP_000357.3
SEQ ID NO: 14
MDAIKKKMQMLKLDKENALDRAEQAEADKKAAEDRSKQLEDELVSLQKKLKGTEDELDKYSEALKDAQEK LELAEKKATDAEADVASLNRRIQLVEEELDRAQERLATALQKLEEAEKAADESERGMKVIESRAQKDEEK MEIQEIQLKEAKHIAEDADRKYEEVARKLVI IESDLERAEERAELSEGQVRQLEEQLRIMDQTLKALMAA EDKYSQKEDRYEEEIKVLSDKLKEAETRAEFAERSVTKLEKSIDDLEDELYAQKLKYKAISEELDHALND MTSM
Biomarker: titin
mRNA Sequence
GenBank: X69490.1
SEQ ID NO: 15
CCTAATTACATAGTTGAAAAGCGTGAATCGGGTACAACAGCTTGGCAGCTTGTCAATTCCAGTGTCAAGC GCACTCAAATTAAAGTCACTCATCTCACAAAATACATGGAATATTCTTTCCGTGTCAGTTCAGAGAACAG ATTTGGTGTCAGCAAACCTCTAGAATCAGCACCAATAATTGCTGAACATCCATTTGTCCCACCAAGCGCT CCTACCAGACCTGAGGTCTACCATGTGTCTGCCAATGCCATGTCTATTCGTTGGGAAGAACCCTACCACG ATGGTGGCAGTAAAATCATTGGCTACTGGGTTGAGAAGAAAGAACGTAATACAATTCTTTGGGTGAAAGA AAACAAAGTGCCATGCTTAGAGTGCAACTACAAAGTAACTGGTTTAGTAGAAGGACTGGAATATCAGTTC AGAACTTATGCACTCAATGCTGCAGGTGTTAGCAAGGCCAGCGAAGCTTCAAGACCTATAATGGCTCAAA ATCCAGTTGATGCACCAGGCAGACCAGGGGTGACAGATGTCCAAAGATCAACAGTATCACTGATTTGGTC TGCCCCAGCGTATGATGGAGGCAGCAAGGTTGTGGGCTACATCATAGAGCGTAAGCCAGTCAGTGAGGTA GGAGATGGTCGCTGGCTGAAGTGCAACTACACCATTGTATCTGACAATTTCTTCACCGTGACTGCTCTCA GTGAAGGAGACACTTATGAGTTCCGTGTGTTAGCCAAGAATGCAGCAGGCGTAATTAGCAAAGGGTCTGA ATCTACAGGCCCTGTCACTTGCCGAGATGAATACGCTCCACCCAAAGCCGAACTGGATGCCCGATTACAC GGTGATCTGGTTACCATCAGAGCAGGTTCTGATCTTGTTCTGGATGCTGCAGTTGGTGGCAAACCTGAAC CCAAAATTATCTGGACCAAAGGAGACAAGGAGCTAGATCTCTGTGAAAAAGTCTCTTTGCAGTATACTGG CAAACGAGCAACTGCTGTGATCAAGTTCTGTGACAGAAGTGACAGTGGAAAATACACTTTAACAGTGAAA AATGCCAGCGGGACCAAGGCCGTGTCTGTCATGGTCAAAGTGCTTGATTCCCCTGGCCCATGTGGAAAGC TCACCGTCAGCAGAGTAACACAGGAGAAGTGCACTTTAGCCTGGAGCCTTCCGCAGGAAGACGGAGGAGC AGAAATCACTCACTACATCGTGGAAAGACGCGAGACTAGCAGGCTCAACTGGGTGATTGTTGAAGGCGAA TGCCCAACCCTATCCTATGTCGTTACCAGGCTCATCAAGAACAATGAGTACATATTCCGAGTGAGGGCAG TAAACAAATATGGCCCTGGTGTGCCTGTTGAATCAGAGCCAATTGTAGCCAGAAACTCATTCACTATTCC ATCACCACCCGGCATACCTGAAGAAGTTGGGACTGGCAAAGAGCATATCATCATTCAGTGGACAAAACCT GAATCTGATGGTGGCAATGAAATCAGCAACTACCTAGTAGACAAACGTGAGAAGGAGAGCCTGCGCTGGA CACGTGTCAACAAAGACTATGTGGTGTATGATACCAGGCTGAAGGTGACCAGCCTGATGGAGGGTTGTGA TTACCAGTTCCGGGTGACCGCAGTGAATGCAGCTGGTAACAGTGAGCCCAGCGAACGTTCCAACTTCATC TCATGCAGAGAACCATCATATACCCCTGGACCACCTTCTGCTCCAAGAGTTGTGGATACCACCAAACACA GCATTAGTTTGGCATGGACCAAACCCATGTACGATGGTGGTACTGACATTGTAGGATATGTTCTGGAAAT GCAAGAGAAGGACACTGATCAGTGGTACCGAGTGCATACCAATGCCACAATAAGAAATACTGAATTCACT GTGCCAGACCTTAAAATGGGCCAGAAATATTCCTTCAGAGTTGCTGCCGTGAACGTGAAGGGTATGAGCG AATACAGCGAATCAATTGCTGAAATTGAGCCCGTGGAAAGAATAGAAATACCAGATCTTGAGCTTGCAGA TGATCTAAAGAAGACTGTGACCATCAGGGCTGGGGCCTCCTTGCGCTTGATGGTGTCTGTATCTGGAAGA CCACCTCCTGTCATAACGTGGAGCAAGCAGGGCATTGACCTTGCAAGCCGGGCAATTATTGACACCACTG AGAGCTACTCATTGCTAATAGTGGACAAAGTTAATCGGTACGATGCTGGAAAATACACAATTGAAGCTGA AAACCAATCTGGCAAGAAATCAGCAACAGTCCTTGTTAAAGTCTATGATACTCCTGGTCCCTGTCCTTCA GTGAAAGTTAAGGAAGTATCAAGAGATTCTGTGACTATAACTTGGGAAATTCCCACGATTGATGGTGGAG CTCCAATCAACAATTACATCGTTGAGAAGCGTGAAGCTGCTATGAGAGCATTCAAAACAGTAACTACCAA ATGCAGCAAGACACTTTACAGAATTTCTGGACTTGTAGAAGGAACCATGCACTATTTCAGAGTGCTGCCA GAAAATATTTATGGCATTGGAGAACCTTGTGAAACATCTGATGCAGTACTGGTCTCAGAAGTGCCTTTGG TGCCTGCAAAGCTAGAAGTGGTCGATGTCACCAAATCCACTGTTACCCTTGCCTGGGAAAAACCACTCTA CGATGGTGGTAGCCGACTCACTGGATATGTTCTCGAGGCCTGCAAAGCTGGCACAGAGAGATGGATGAAG GTTGTCACCT TAAAAC C C AC AG C C AG AGC AC AC TGT AC TCCT TAAA GAAGG G AAC AA AC T AT TTAGAATAAGGGCACAAAATGAGAAAGGTGTGTCAGAACCAAGAGAGACTGTCACAGCCGTGACTGTACA AGACCTCAGAGTGTTGCCAACAATCGATCTTTCTACAATGCCTCAGAAGACCATCCATGTCCCAGCTGGC AGACCAGTAGAGCTGGTGATACCTATTGCTGGCCGTCCACCTCCTGCTGCTTCCTGGTTCTTTGCTGGTT C TAAAC T GAG AG AAT C AG AGC G T G T C AC AG T T GAAAC T C AC AC TAAAG T AGC TAAAT T AAC CATCCGTGA AAC C AC TAT C AG AG AT AC T GG AG AAT AC AC AC T T G AAT T G AAG AAT G T T AC C GG AAC T AC T T T AG AAAC C AT T AAAG T T AT C AT T C T T G AC AAGC C T GG T C C AC C AAC AGG AC C T AT T AAG AT T G AT G AAAT T G AT GC T A CATCAATTACCATTTCCTGGGAACCACCTGAATTGGACGGTGGTGCTCCACTGAGTGGTTATGTGGTAGA ACAACGTGACGCTCATCGTCCAGGATGGCTGCCCGTTTCTGAATCAGTGACTAGGTCCACGTTTAAGTTT ACCAGACTCACCGAAGGAAATGAGTATGTGTTCCGTGTGGCTGCAACAAACCGCTTCGGGATTGGCTCTT ACTTGCAGTCTGAGGTCATAGAGTGTCGCAGCAGCATCCGTATTCCTGGACCCCCAGAAACATTACAGAT ATTTGATGTTTCCCGTGATGGCATGACACTTACTTGGTACCCACCAGAGGATGACGGTGGCTCCCAAGTG ACTGGATATATTGTGGAGCGCAAAGAAGTGAGAGCAGATCGATGGGTCCGTGTAAATAAAGTACCTGTGA CAATGACACGGTACCGCTCCACTGGCCTTACTGAAGGCTTAGAATATGAACACCGTGTCACAGCCATTAA TGCAAGAGGGTCTGGGAAACCAAGTCGTCCTTCCAAACCCATCGTTGCCATGGATCCAATTGCTCCTCCA GGAAAGCCACAAAACCCAAGAGTTACTGATACAACAAGGACATCAGTCTCCCTGGCCTGGAGTGTTCCAG AAGATGAAGGAGGATCTAAAGTCACAGGCTACTTGATTGAAATGCAAAAAGTAGATCAACATGAATGGAC C AAG T G T AAC AC C AC T C C AAC C AAG AT T C GAG AG T AT AC T C T AAC AC AC C T AC C T C AGGG T GC AG AAT AC AGGTTCCGCGTCCTAGCTTGTAATGCTGGTGGACCTGGTGAGCCTGCTGAGGTACCAGGAACAGTCAAAG TCACTGAAATGCTTGAATATCCTGATTATGAACTTGATGAAAGATACCAAGAAGGTATCTTTGTAAGGCA AGGTGGCGTCATCAGACTTACCATACCAATCAAAGGGAAACCATTCCCAATATGTAAATGGACCAAGGAA GGCCAGGATATTAGTAAGCGTGCCATGATTGCAACATCTGAAACACACACTGAGCTTGTGATCAAAGAAG CAGACAGGGGTGATTCTGGCACTTATGACCTGGTTCTGGAAAATAAATGTGGCAAGAAGGCTGTCTACAT CAAGGTCAGGGTGATAGGAAGTCCCAACAGTCCAGAAGGGCCACTGGAATATGATGACATCCAAGTCCGC TCTGTGAGGGTCAGCTGGAGACCTCCTGCTGATGATGGTGGTGCTGACATCTTAGGCTACATCCTCGAGA GACGAGAAGTGCCTAAAGCCGCCTGGTATACCATTGATTCCAGAGTCCGAGGTACATCTCTGGTGGTAAA AGGCCTCAAAGAGAATGTAGAATACCATTTCCGTGTTTCAGCAGAAAACCAGTTTGGCATAAGCAAACCC T T G AAAT C T G AGG AAC C AG T C AC AC C AAAAAC AC C AT T G AAT C C T C C AG AAC C T C C AAGC AAT C C T C C AG AAGTACTCGATGTAACCAAGAGTTCTGTTAGCTTGTCCTGGTCCCGGCCCAAAGATGATGGTGGTTCTAG AGTCACAGGCTACTACATCGAACGCAAAGAGACATCCACTGACAAGGTGGTCAGACACAACAAGACTCAG ATCACCACCACAATGTACACTGTCACAGGGCTTGTTCCCGATGCTGAGTATCAGTTCCGCATCATCGCAC AGAATGATGTTGGCCTGAGTGAGACCAGCCCTGCTTCTGAACCAGTTGTTTGCAAAGATCCATTTGATAA AC C AAGC C AAC C AGG AG AAC T T GAG AT TCTTTCAATATC C AAAG AT AG T G T C AC T C T AC AG T GGG AG AAA CCTGAATGTGATGGTGGTAAAGAAATTCTTGGATACTGGGTTGAATATAGACAGTCTGGAGACAGTGCCT GGAAGAAGAGCAATAAGGAACGTATTAAGGACAAGCAATTCACAATAGGAGGTTTGCTGGAAGCTACTGA GTATGAATTCAGGGTTTTTGCTGAGAATGAGACTGGGCTGAGCAGACCTCGCAGAACTGCTATGTCTATA AAGACTAAACTCACATCTGGAGAGGCCCCAGGAATACGCAAAGAAATGAAGGATGTTACCACAAAATTGG GTGAAGCTGCTCAACTCTCATGCCAGATTGTTGGAAGGCCTCTTCCTGACATTAAATGGTACAGATTTGG TAAAGAGCTCATACAAAGCCGGAAATACAAAATGTCTTCAGATGGACGCACACACACTCTTACAGTAATG ACAGAGGAACAGGAAGATGAAGGTGTTTATACCTGCATAGCCACCAATGAGGTTGGAGAAGTAGAAACCA GTAGTAAGCTTCTCCTGCAAGCAACACCGCAGTTCCATCCTGGTTACCCACTGAAAGAGAAATATTATGG AGCTGTGGGTTCCACACTTCGGCTTCATGTTATGTACATTGGTCGTCCAGTACCTGCCATGACTTGGTTC CATGGTCAGAAACTTTTGCAAAACTCAGAAAACATTACTATTGAAAACACTGAGCACTATACTCATCTTG TCATGAAGAATGTCCAACGTAAGACTCATGCTGGGAAATACAAAGTCCAGCTCAGCAATGTTTTTGGAAC AGTTGATGCCATCCTTGATG T GG AAAT AC AAG AT AAAC C AG AC AAAC C T AC AGG AC CAATTGTGATC G AA GCTCTATTGAAGAACTCCGCAGTGATCAGCTGGAAACCACCCGCAGATGACGGAGGCTCCTGGATCACCA ACTATGTGGTGGAAAAATGTGAGGCCAAGGAGGGGGCTGAATGGCAATTGGTGTCTTCAGCCATCTCAGT GACAACCTGTAGAATTGTGAACCTCACAGAAAATGCTGGCTATTACTTCCGGGTTTCAGCTCAGAACACT TTCGGCATCAGTGACCCTCTAGAAGTGTCCTCAGTTGTGATCATTAAGAGTCCATTTGAAAAGCCAGGTG CTCCTGGCAAACCAACTATTACTGCTGTCACAAAAGATTCTTGTGTTGTGGCCTGGAAGCCACCTGCCAG TGATGGAGGTGCAAAGATTAGAAATTACTACCTTGAGAAGCGTGAGAAGAAGCAGAATAAATGGATTTCT G T G AC AAC AG AAG AAAT T C GAG AAAC TGTCTTTT C AG T G AAAAAC C T T AT T G AAGG T C T T G AAT AC GAG T TTCGTGTGAAATGTGAAAATCTAGGTGGGGAAAGTGAATGGAGTGAAATATCAGAACCCATCACTCCCAA ATCTGATGTCCCAATTCAGGCACCACACTT T AAAG AGG AAC T GAG AAAT C T AAAT G T C AG AT AT C AG AGC AATGCTACCTTGGTCTGCAAAGTGACTGGTCATCCAAAACCTATCGTCAAATGGTACAGACAAGGCAAAG AAATCATTGCAGATGGATTAAAATATAGGATTCAAGAATTTAAGGGTGGCTACCACCAGCTCATCATTGC AAGTGTCACAGATGATGATGCCACAGTTTACCAAGTCAGAGCTACCAACCAAGGGGGATCTGTGTCTGGC ACTGCCTCCTTGGAAGTGGAAGTTCCAGCTAAGATACACTTACCTAAAACTCTTGAAGGCATGGGAGCAG TTCATGCTCTCCGAGGTGAAGTGGTCAGCATCAAGATTCCTTTCAGTGGCAAACCAGATCCTGTGATCAC CTGGCAGAAAGGACAAGATCTCATTGACAATAATGGCCACTACCAAGTTATTGTCACAAGATCCTTCACA TCACTTGTTTTCCCCAATGGGGTAGAGAGAAAAGATGCTGGTTTCTATGTGGTCTGTGCTAAAAACAGAT TTGGAATTGATCAGAAGACAGTTGAACTGGATGTGGCTGATGTTCCTGACCCACCCAGAGGAGTCAAAGT TAGTGATGCCTCACGAGATTCTGTCAACTTAACATGGACTGAGCCAGCCTCTGATGGTGGCAGCAAAATC ACCAACTACATTGTTGAAAAATGTGCAACTACTGCAGAAAGATGGCTCCGTGTAGGACAGGCCCGAGAAA CACGTTATACCGTGATCAACTTATTTGGAAAAACAAGTTACCAGTTCCGGGTAATAGCTGAAAATAAATT TGGTCTGAGCAAGCCTTCAGAGCCTTCAGAACCAACCATAACCAAAGAAGATAAGACCAGAGCTATGAAC TATGATGAAGAGGTAGATGAAACCAGGGAAGTCTCCATGACTAAAGCATCTCACTCTTCAACCAAGGAAC TCTATGAGAAATATATGATTGCTGAAGATCTTGGGCGTGGTGAGTTTGGAATTGTCCATCGTTGTGTTGA AACATCCTCAAAGAAGACATACATGGCCAAATTTGTTAAAGTCAAAGGGACTGATCAGGTTTTGGTAAAG AAGGAAATTTCCATTCTGAATATTGCTAGGCATAGAAACATCTTACACCTCCATGAATCATTTGAAAGCA TGGAAGAATTAGTTATGATCTTTGAGTTTATATCAGGACTTGACATATTTGAGCGCATTAACACAAGTGC TTTTGAACTTAATGAAAGAGAAATTGTAAGTTATGTTCACCAGGTCTGTGAAGCACTTCAGTTTTTACAC AGTCATAATATTGGACACTTTGACATTAGACCAGAAAATATCATTTACCAAACCAGAAGAAGCTCTACCA TTAAAATCATAGAATTTGGTCAAGCCCGTCAGCTGAAACCAGGGGACAACTTCAGGCTTCTATTCACTGC CCCAGAATACTATGCACCTGAAGTCCACCAGCATGATGTTGTCAGCACAGCCACAGACATGTGGTCACTT GGAACACTGGTATATGTGCTATTGAGTGGTATCAACCCATTCCTGGCTGAAACTAACCAACAGATCATTG AGAATATCATGAATGCTGAATATACTTTCGATGAGGAAGCATTCAAAGAGATTAGCATTGAAGCCATGGA TTTTGTTGACCGGTTGTTAGTGAAAGAGAGGAAATCTCGCATGACAGCATCGGAGGCTCTCCAGCACCCA T GG T T G AAGC AG AAG AT AG AAAG AG T C AG T AC TAAAG T T AT C AG AAC AT T AAAAC AC CGGCGTTATTACC ACACCCTGATCAAGAAAGACCTCAACATGGTTGTGTCAGCAGCCCGGATCTCCTGTGGTGGTGCAATTCG ATCTCAGAAGGGAGTGAGTGTTGCTAAAGTTAAAGTGGCATCCATTGAAATTGGCCCAGTTTCTGGGCAG ATAATGCATGCAGTTGGTGAAGAAGGAGGACATGTCAAATATGTATGCAAAATTGAAAATTATGATCAGT C T AC C C AAG T G AC T T GG T AC TTTGGTGTCC G AC AGC T GG AG AAC AG T G AG AAAT AC G AAAT C AC C T AC G A AGATGGAGTGGCCATCCTCTATGTCAAAGACATTACCAAATTAGATGATGGTACCTACAGATGCAAAGTA G T C AAT G AC T AT GG T G AAG AC AG T T C T T AT GC AG AGC TAT T T G T TAAAGGT G T GAG AG AAG T C T AT G AC T ATTACTGCCGTAGAACCATGAAGAAAATTAAGCGCAGAACAGACACAATGAGACTCCTGGAAAGGCCACC AGAATTTACCCTGCCTCTCTATAATAAGACAGCTTATGTAGGTGAAAATGTCCGGTTTGGAGTAACTATA ACTGTCCACCCAGAGCCTCATGTAACATGGTATAAATCAGGTCAGAAAATCAAACCAGGTGACAATGACA AG AAG T AC AC AT T T GAG T C AG AC AAGGG T CTTTACCAAT T AAC AAT C AAC AG T G T C AC T AC AG AT GAT G A CGCTGAATATACTGTTGTGGCAAGGAACAAATATGGTGAAGACAGCTGTAAAGCAAAGCTGACAGTAACC CTACACCCACCTCCAACAGATAGTACCTTAAGACCCATGTTCAAAAGGTTACTGGCAAATGCAGAATGCC AAGAAGGCCAAAGTGTCTGCTTTGAGATCAGAGTGTCTGGCATCCCCCCACCAACATTAAAATGGGAGAA AGATGGTCAGCCACTGTCCCTCGGGCCTAACATTGAAATTATCCATGAAGGCTTGGATTATTATGCTCTG CACATCAGGGACACTTTGCCTGAAGACACGGGTTATTATAGAGTCACAGCCACTAACACAGCTGGGTCCA CCAGCTGCCAGGCTCACCTACAAGTGGAACGCCTGAGGTACAAGAAACAGGAATTCAAGAGTAAGGAGGA GCATGAGCGACACGTACAAAAACAAATTGACAAAACCCTCAGAATGGCTGAAATTCTTTCTGGAACTGAA AGTGTACCACTGACACAGGTAGCTAAAGAGGCTCTGAGAGAAGCTGCTGTCCTTTATAAACCGGCTGTAA GCACCAAGACTGTAAAAGGGGAATTCAGACTTGAGATAGAAGAAAAGAAGGAGGAGAGAAAACTCCGGAT GCCTTATGATGTACCAGAGCCACGCAAGTATAAGCAGACTACCATAGAAGAAGACCAACGCATCAAGCAG TTCGTGCCCATGTCTGACATGAAGTGGTATAAAAAGATACGTGATCAGTATGAAATGCCTGGGAAACTTG ACAGAGTTGTACAGAAACGACCCAAGCGCATCCGCCTTTCAAGATGGGAACAGTTCTATGTGATGCCTCT T C C AC GC AT T AC AG AT C AAT AC AG AC C T AAAT GGC G T AT T C C T AAAC T G T C C C AAG AT G AT C T T G AG AT A GTGAGACCAGCCCGCCGGCGTACACCTTCTCCTGATTATGACTTTTACTACCGACCTAGAAGACGTTCTC TTGGGGACATCTCTGATGAAGAATTACTCCTCCCCATTGATGACTACTTAGCAATGAAAAGAACAGAGGA AGAGAGGCTGCGTCTTGAAGAAGAGCTTGAGTTAGGTTTTTCAGCTTCACCCCCAAGTCGAAGCCCTCCA CACTTTGAGCTTTCTAGCCTACGTTACTCTTCACCACAAGCTCATGTCAAGGTGGAGGAAACAAGAAAAA ACTTCAGGTATTCAACCTATCACATCCCAACGAAGGCTGAAGCTAGTACAAGTTATGCAGAACTGAGGGA ACGGCATGCCCAGGCTGCGTACAGACAGCCAAAGCAACGGCAAAGAATCATGGCTGAGAGGGAGGATGAA GAGTTGCTTCGCCCAGTTACGACCACCCAGCATCTCTCAGAATACAAAAGCGAACTTGACTTCATGTCAA AGGAGGAAAAGTCTAGAAAGAAATCAAGGCGACAAAGAGAAGTGACAGAAATAACAGAAATTGAGGAAGA ATACGAAATCTCAAAACATGCTCAAAGAGAATCATCCTCATCTGCGTCTAGACTACTGAGACGACGGCGC TCCCTGTCTCCAACTTATATTGAGTTAATGAGGCCAGTGTCTGAGCTGATCCGGTCACGTCCACAACCGG C T G AGG AAT AC G AAG AT G AC AC AG AAAG AAGG T C AC C T AC T C C AG AG AG AAC TCGCCCACGATCCCCCAG CCCTGTGTCTAGTGAGAGATCACTCTCGAGATTTGAGAGGTCTGCAAGATTTGATATCTTTTCCAGGTAT GAGTCCATGAAAGCTGCTTTAAAAACTCAGAAGACATCAGAAAGGAAGTATGAAGTTTTGAGTCAGCAGC CTTTCACACTGGACCATGCCCCTCGAATCACACTGAGAATGCGCTCGCACAGGGTACCATGTGGCCAAAA TACACGTTTTATTTTAAATGTTCAGTCTAAGCCAACTGCCGAAGTTAAATGGTACCACAATGGTGTGGAA C T C C AAG AAAGC AG T AAG AT T CAT T AC AC C AAC AC GAG T GG AG TTCTCACCC T GG AAAT T C T GG AC T G T C ATACTGATGACAGTGGAACCTACCGTGCTGTGTGCACCAACTACAAGGGCGAACGTTCTGACTATGCAAC GTTGGACGTGACAGGAGGGGATTATACCACCTATGCTTCCCAACGCAGAGATGAAGAGGTCCCCAGATCT GTTTTCCCTGAGCTGACAAGAACAGAGGCGTATGCTGTTCCATCATTTAAGAAAACATCTGAGATGGAAG CTTCGTCTTCTGTCAGGGAAGTGAAATCACAGATGACGGAGACAAGGGAAAGTCTCTCCTCATATGAACA CTCTGCATCTGCAGAAATGAAAAGTGCTGCATTAGAAGAAAAGTCACTGGAAGAAAAATCCACAACCAGA AAGATCAAGACGACTTTGGCGGCAAGAATTCTAACAAAGCCACGGTCCATGACCGTCTACGAGGGCGAGT CTGCAAGGTTTTCTTGTGACACCGATGGTGAGCCGGTACCAACTGTGACCTGGCTGCGTAAAGGACGAGT GC T AAG T AC T T C T GC C C GC C AC C AAG T G AC C AC C AC AAAG T AC AAAT C AAC C T T T G AG AT C T C T T C AG T C CAGGCTTCCGATGAGGGCAATTACAGCGTGGTGGTAGAAAACAGTGAAGGGAAACAAGAAGCAGAGTTCA CTCTGACTATTCAAAAGGCCAGGGTAACTGAAAAGGCTGTGACATCACCACCAAGAGTCAAATCCCCAGA GCCTCGGGTGAAATCCCCAGAAGCAGTTAAGTCTCCAAAACGAGTGAAATCTCCAGAACCTTCTCACCCG AAAGC C G T AT C AC C C AC AG AG AC AAAAC C AAC AC C AAT AG AG AAAG TTCAGCACCTCCCAGTCTCTGCCC CACCAAAGATTACTCAGTTCCTGAAAGCAGAAGCTTCTAAAGAGATTGCAAAACTGACCTGTGTGGTTGA AAGCAGTGTATTAAGGGCAAAAGAGGTCACCTGGTATAAAGATGGCAAGAAACTGAAGGAAAATGGGCAT T T C C AG T T T C AT T AT T C AGC AG AT GG T AC C T AT G AGC T C AAAAT C AAT AAC C T C AC T G AAT C T G AT C AAG GAGAATATGTTTGTGAGATTTCTGGTGAAGGTGGAACGTCTAAAACCAACTTACAATTTATGGGGCAAGC C T T T AAG AG T AT C C AT GAG AAGG TAT C AAAAAT AT C AG AAAC T AAG AAAT C AG AT C AG AAAAC C AC T GAG T C AAC AG T AAC C AG AAAAAC T G AAC CAAAAGCT C C T G AAC CAATTTCCT C AAAAC CAGTAATTGTTACTG GGTTGCAGGATACAACTGTTTCTTCAGACAGTGTTGCTAAATTTGCAGTTAAGGCTACTGGAGAACCCCG GCCAACTGCCATCTGGACAAAAGATGGAAAGGCCATTACACAAGGAGGTAAATATAAACTCTCTGAAGAC AAGGGAGGGTTCTTCTTAGAAATTCATAAGACTGATACTTCTGACAGTGGACTTTATACTTGTACAGTAA AAAATTCAGCTGGATCTGTGTCCTCTAGCTGCAAATTAACAATAAAAGCTATAAAAGATACTGAGGCACA GAAAGTCTCTACACAAAAGACTTCTGAAATTACACCTCAGAAGAAAGCTGTTGTCCAAGAGGAAATTTCC CAAAAAGCCCTAAGGTCTGAAGAAATTAAGATGTCAGAGGCAAAATCTCAAGAAAAGTTAGCCCTCAAAG AGGAAGCTTCAAAGGTTCTGATTTCTGAAGAAGTCAAGAAATCAGCAGCAACCTCCCTGGAAAAATCCAT TGTCCATGAGGAAATCACTAAAACATCACAGGCATCAGAAGAAGTCAGAACTCATGCTGAGATTAAAGCA TTTTCTACTCAGATGAGCATAAACGAAGGTCAAAGACTGGTTTTAAAAGCCAACATTGCTGGTGCCACTG ATGTGAAATGGGTACTGAATGGCGTAGAGCTTACCAACTCTGAGGAGTACCGATATGGTGTCTCAGGCAG CGATCAGACCCTAACCATCAAGCAAGCCAGTCACAGAGATGAAGGAATCCTCACCTGCATAAGCAAAACC AAGGAAGGAATCGTCAAGTGTCAGTATGATTTGACACTGAGCAAAGAACTCTCAGATGCTCCAGCCTTCA TCTCACAGCCTAGATCTCAAAATATTAATGAAGGACAAAATGTTCTCTTTACTCGTGAAATCAGTGGCGA GCCATCCCCTGAAATCGAATGGTTTAAAAACAACCTGCCAATTTCTATTTCTTCAAATGTCAGCATAAGC CGCTCCAGAAATGTATACTCCCTTGAAATCCGAAATGCATCAGTCAGCGACAGTGGAAAGTACACAATTA AGGCCAAAAATTTCCGTGGCCAGTGTTCAGCTACAGCTTCCTTAATGGTCCTTCCTCTAGTTGAAGAACC TTCCAGAGAGGTAGTATTGAGAACAAGTGGTGACACAAGCTTGCAAGGAAGCTTCTCGTCTCAGTCAGTC CAAATGTCTGCCTCCAAGCAGGAGGCCTCCTTCAGCAGTTTCAGCAGCAGCAGTGCTAGCAGCATGACTG AGATGAAATTTGCAAGCATGTCTGCCCAAAGCATGTCCTCCATGCAAGAGTCCTTTGTAGAAATGAGTTC CAGCAGCTTTATGGGAATATCTAATATGACACAACTGGAAAGCTCAACTAGTAAAATGCTTAAAGCAGGC ATAAGAGGAATTCCGCCTAAAATTGAAGCTCTTCCATCTGATATCAGCATTGATGAAGGCAAAGTTCTAA CAGTAGCCTGTGCTTTCACGGGTGAGCCTACCCCAGAAGTAACATGGTCCTGTGGTGGAAGAAAAATCCA CAGTCAAGAACAGGGGAGGTTCCACATTGAAAACACAGATGACCTGACAACCCTGATCATCATGGACGTA CAGAAACAAGATGGTGGACTTTATACCCTGAGTTTAGGGAATGAATTTGGATCTGACTCTGCCACTGTGA ATATACATATTCGATCCATTTAAGAGGGCCTGTGCCTTATACTCTACACTCATTCTTAACTTTTCGCAAA CGTTTCACACGGACTAATCTTTCTGAACTGTAAATATTTAAAGAAAAAAAGTAGTTTTGTATCAACCTAA ATGAGTCAAAGTTCAAAAATATTCATTTCAATCTTTTCATAATTGTTGACCTAAGAATATAATACATTTG CTAGTGACATGTACATACTGTATATAGCCGGATTAACGGTTATAAAGTTTTGTACCATTTATTTTATGAC ATTTTACAATGTAAGTTTTGAAACTAACTGTTGGTAGGAGAAAGTTTCTTATGGAACGAATACCCTGCTC AACATTTAATCAATCTTTGTGCCTCAACATACTGTTGATGTCTAAGTATGCCTCAGTGGGTTGAGAAAAT CCCCATTGAAGATGTCCTGTCCACCTAAAAGAGAATGATGCTGTGCATATCACTTGATATGTGCACCAAT ACCTACTGAATCAGAAATGTAAGGCATTGGTGATGTTTGCATTTACCCTCCTGTAAGCAACACTTTAACG TCTTACATTTTCTCTGATGATGTCACACAAAATTATCATGACAAATATTACCAGAGCAAAGTGTAACGGC CAACACTTTGTTCGCTCATTTTACGCTGTCTCTGACATAAGGAGTGCCTGAATAGCTTGGAAAAGTAACA TCTCCTGGCCATCCCTTCATTTAACCAAGCTATTCAAGTATTCCTATGCCAGAGCAGTGCCAACTCTTGG AGGTCCCAGAGTGCAGCCAATGCCTTTGTGTGGTAGTTCTAAATTTTAATTGCACCTGAAAAACCTGGGC ACCTAAGCAATGAGCCACAGCAAAAAGTAAAGAACAACAACAAAATAAAGCTGTTGTTAAATTTTAAACA ATATTACTAATTGCCCAAAATGTCAATTTGATGTAGTTCTTTTCATGCAAGTATAAATTCAATTGTTAGT TATAATTGTTGGACCTCCTTGAGATAGTAACAACAAAATAAAGCAAGCTATCTGCACCTCAAAAAAAAAA AAAAA
Amino Acid Sequence
GenBank: CAA62188.1
SEQ ID NO: 16
MTTQAPTFTQPLQSVWLEGSTATFEAHISGFPVPEVSWFRDGQVISTSTLPGVQISFSDGRAKLTIPAV TKANSGRYSLKATNGSGQATSTAELLVKAETAPPNFVQRLQSMTVRQGSQVRLQVRVTGIPNPWKFYRD GAEIQSSLDFQISQEGDLYSLLIAEAYPEDSGTYSVNATNSVGRATSTAELLVQGEEEVPAKKTKTIVST AQISESRQTRIEKKIEAHFDARSIATVEMVIDGAAGQQLPHKTPPRIPPKPKSRSPTPPSIAAKAQLARQ QSPSPIRHSPSPVRHVRAPTPSPVRSVSPAARISTSPIRSVRSPLLMRKTQASTVATGPEVPPPWKQEGY VASSSEAEMRETTLTTSTQIRTEERWEGRYGVQEQVTISGAAGAAASVSASASYAAEAVATGAKEVKQDA DKSAAVATWAAVDMARVREPVI SAVEQTAQRTTTTAVHIQPAQEQVRKEAEKTAVTKVWAADKAKEQE LKSRTKEI ITTKQEQMHVTHEQIRKETEKTFVPKWISAAKAKEQETRISEEI KKQKQVTQEAIMKETR KTWPKVIVATPKVKEQDLVSRGREGITTKREQVQITQEKMRKEAEKTALSTIAVATAKAKEQETILRTR ETMATRQEQIQVTHGKVDVGKKAEAVATWAAVDQARVREPREPGHLEESYAQQTTLEYGYKERISAAKV AEPPQRPASEPHWPKAVKPRVIQAPSETHIKTTDQKGMHISSQIKKTTDLTTERLVHVDKRPRTASPHF TVSKISVPKTEHGYEASIAGSAIATLQKELSATSSAQKITKSVKAPTVKPSETRVRAEPTPLPQFPFADT PDTYKSEAGVEVKKEVGVSITGTTVREERFEVLHGREAKVTETARVPAPVEIPVTPPTLVSGLKNVTVIE GESVTLECHISGYPSPTVTWYREDYQIESSIDFQITFQSGIARLMIREAFAEDSGRFTCSAVNEAGTVST SCYLAVQVSEEFEKETTAVTEKFTTEEKRFVESRDWMTDTSLTEEQAGPGEPAAPYFITKPWQKLVEG GSWFGCQVGGNPKPHVYWKKSGVPLTTGYRYKVSYNKQTGECKLVISMTFADDAGEYTIWRNKHGETS ASASLLEEADYELLMKSQQEMLYQTQVTAFVQEPEVGETAPGFVYSEYEKEYEKEQALIRKKMAKDTVW RTYVEDQEFHISSFEERLIKEIEYRI IKTTLEELLEEDGEEKMAVDISESEAVESGFDLRIKNYRILEGM GVTFHCKMSGYPLPKIAWYKDGKRIKHGERYQMDFLQDGRASLRIPWLPEDEGIYTAFASNIKGNAICS GKLYVEPAAPLGAPTYIPTLEPVSRIRSLSPRSVSRSPIRMSPARMSPARMSPARMSPARMSPGRRLEET DESQLERLYKPVFVLKPVSFKCLEGANCRFDLKWGRPMPETFWFHDGQQIVNDYTHKWIKEDGTQSLI IVPATPSDSGEWTWAQNRAGRSSISVILTVEAVEHQVKPMFVEKLKNVNIKEGSRLEMKVRATGNPNPD IVWLKNSDI IVPHKYPKIRIEGTKGEAALKIDSTVSQDSAWYTATAINKAGRDTTRCKVNVEVEFAEPEP ERKLI IPRGTYRAKEIAAPELEPLHLRYGQEQWEEGDLYDKEKQQKPFFKKKL SLRLKRFGPAHFECRL TPISDPTMWEWLHDGKPLEAANRLRMINEFGYCSLDYGVAYSRDSGI ITCRATNKYGTDHTSATLIVKD EKSLVEESQLPEGRKGLQRIEELERMAHEGALTGVTTDQKEKQKPDIVLYPEPVRVLEGETARFRCRVTG YPQPKVNWYLNGQLIRKSKRFRVRYDGIHYLDIVDCKSYDTGEVKVTAENPEGVIEHKVKLEIQQREDFR SVLRRAPEPRPEFHVHEPGKLQFEVQKVDRPVDTTETKEWKLKRAERI HEKVPEESEELRSKFKRRTE EGYYEAITAVELKSRKKDESYEELLRKTKDELLHWTKELTEEEKKALAEEGKITIPTFKPDKIELSPSME APKIFERIQSQTVGQGSDAHFRVRWGKPDPECEWYKNGVKIERSDRIYWYWPEDNVCELVIRDVTAEDS ASIMVKAINIAGETSSHAFLLVQAKQLITFTQELQDWAKEKDTMATFECETSEPFVKVKWYKDGMEVHE GDKYRMHSDRKVHFLSILTIDTSDAEDYSCVLVEDENVKTTAKLIVEGAWEFVKELQDIEVPESYSGEL ECIVSPENIEGKWYHNDVELKSNGKYTITSRRGRQNLTVKDVTKEDQGEYSFVIDGKKTTCKLKMKPRPI AILQGLSDQKVCEGDIVQLEVKVSLESVEGVWMKDGQEVQPSDRVHIVIDKQSHMLLIEDMTKEDAGNYS FTIPALGLSTSGRVSVYSVDVITPLKDVNVIEGTKAVLECKVSVPDVTSVKWYLNDEQIKPDDRVQAIVK GTKQRLVINRTHASDEGPYKLIVGRVETNCNLSVEKIKI IRGLRDLTCTETQNWFEVELSHSGIDVLWN FKDKEIKPSSKYKIEAHGKIYKLTVLNMMKDDEGKYTFYAGENMTSGKLTVAGGAISKPLTDQTVAESQE AVFECEVANPDSKGEWLRDGKHLPLTN IRSESDGHKRRLI IAATKLDDIGEYTYKVA SK SAKLKVEA VKIKKTLKNLTVTETQDAVFTVELTHPNVKGVQWIKNGWLESNEKYAISVKGTIYSLRIKNCAIVDESV YGFRLGRLGASARLHVETVKI IKKPKDVTALENATVAFEVSVSHDTVPVKWFHKSVEIKPSDKHRLVSER KVHKLMLQNISPSDAGEYTAWGQLECKAKLFVETLHITKTMKNIEVPETKTASFECEVSHFNVPSMWLK NGVEIEMSEKFKIWQGKLHQLI IMN S EDSAEYTFVCGNDQVSATLTV PIMI SMLKDINAEEKD I TFEVTVNYEGISYKWLKNGVEIKSTDKCQMRTKKLTHSLNIRNVHFGDAADYTFVAGKATSTATLYVEAR HIEFRKHIKDIKVLEKKRAMFECEVSEPDITVQWMKDDQELQITDRIKIQKEKYVHRLLIPSTRMSDAGK YTWAGGNVSTAKLFVEGRDVRIRSIKKEVQVIEKQRAWEFEVNEDDVDAHWYKDGIEINFQVQERHKY WERRIHRMFISETRQSDAGEYTFVAGRNRSSVTLYVNAPEPPQVLQELQPVTVQSGKPARFCAMISGRP QPKISWYKEEQLLSTGFKCKFLHDGQEYTLLLIEAFPEDAAVYTCEAKNDYGVATTSASLSVEVPEWSP DQEMPVYPPAI ITPLQDTV SEGQPARFQCRVSGTDLKVSWYSKDKKIKPSRFFRMTQFEDTYQLEIAEA YPEDEGTYTFVANNAVGQVSSTANLSLEAPESILHERIEQEIEMEMKEFSSSFLSAEEEGLHSAELQLSK INETLELLSESPVYPTKFDSEKEGTGPIFIKEVSNADISMGDVATLSVTVIGIPKPKIQWFFNGVLLTPS ADYKFVFDGDDHSLI ILFTKLEDEGEYTCMASNDYGK ICSAYLKINSKGEGHKDTETESAVAKSLEKLG GPCPPHFLKELKPIRCAQGLPAIFEYTWGEPAPTVTWFKENKQLC SVYY I IHNPNGSGTFIVNDPQR EDSGLYICKAENMLGESTCAAELLVLLEDTDMTDTPCKAKSTPEAPEDFPQTPLKGPAVEALDSEQEIAT FVKDTILKAALITEENQQLSYEHIAKANELSSQLPLGAQELQSILEQDKLTPESTREFLCINGSIHFQPL KEPSPNLQLQIVQSQKTFSKEGILMPEEPETQAVLSDTEKIFPSAMSIEQINSLTVEPLKTLLAEPEGNY PQSSIEPPMHSYLTSVAEEVLSLKEKTVSDTNREQRVTLQKQEAQSALILSQSLAEGHVESLQSPDVMIS QVNYEPLVPSEHSCTEGGKILIESANPLENAGQDSAVRIEEGKSLRFPLALEEKQVLLKEEHSDNWMPP DQI IESKREPVAIKKVQEVQGRDLLSKESLLSGI PEEQRLNLKIQICRALQAAVASEQPGLFSEWLR IE KVEVEAVNITQEPRHIMCMYLVTSAKSVTEEVTI I IEDVDPQMANLKMELRDALCAI IYEEIDILTAEGP RIQQGAKTSLQEEMDSFSGSQKVEPITEPEVESKYLISTEEVSYFNVQSRVKYLDATPVTKGVASAWSD EKQDESLKPSEEKEESSSESGTEEVATVKIQEAEGGLIKEDGPMIHTPLVDTVSEEGDIVHLTTSITNAK EVNWYFENKLVPSDEKFKCLQDQNTYTLVIDKVNTEDHQGEYVCEALNDSGKTATSAKLTWKRAAPVIK RKIEPLEVALGHLAKFTCEIQSAPNVRFQWFKAGREIYESDKCSIRSSKYISSLEILRTQWDCGEYTCK ASNEYGSVSCTATLTVTVPGGEKKVRKLLPERKPEPKEEWLKSVLRKRPEEEEPKVEPKKLEKVKKPAV PEPPPPKPVEEVEVPTVTKRERKIPEPTKVPEIKPAIPLPAPEPKPKPEAEVKTIKPPPVEPEPTPIAAP VTVPWGKKAEAKAPKEEAAKPKGPIKGVPKKTPSPIEAERRKLRPGSGGEKPPDEAPFTYQLKAVPLKF VKEIKDI ILTESEFVGSSAIFECLVSPS AI TWMKDGS IRESPKHRFIADGKDRKLHI IDVQLSDAGE YTCVLRLGNKEKTSTAKLWEELPVRFVKTLEEEVTWKGQPLYLSCELNKERDWWRKDGKIWEKPGR IVPGVIGLMRALTINDADDTDAGTYTVTVENANNLECSSCVKWEVIRDWLVKPIRDQHVKPKGTAIFAC DIAKDTPNIKWFKGYDEIPAEPNDKTEILRDGNHLYLKIKNAMPEDIAEYAVEIEGKRYPAKLTLGEREV ELLKPIEDVTIYEKESASFDAEISEADIPGQWKLKGELLRPSPTCEIKAEGGKRFLTLHKVKLDQAGEVL YQALNAI TAILTVKEIELDFAVPLKDVTVPERRQARFECVLTREANVIWSKGPDI IKSSDKFDI IADGK KHILVINDSQFDDEGVYTAEVEGKKTSARLFVTGIRLKFMSPLEDQTVKEGETATFVCELSHEKMHWWF KNDAKLHTSRTVLISSEGKTHKLEMKEVTLDDISQIKAQVKELSSTAQLKVLEADPYFTVKLHDKTAVEK DEITLKCEVSKDVPVKWFKDGEEIVPSPKYSIKADGLRRILKIKKADLKDKGEYVCDCGTDKTKANVTVE ARLIEVEKPLYGVEVFVGETAHFEIELSEPDVHGQWKLKGQPLTASPDCEI IEDGKKHILILHNCQLGMT GEVSFQAANAKSAANLKVKELPLIFITPLSDVKVFEKDEAKFECEVSREPKTFRWLKGTQEITGDDRFEL IKDGTKHSMVIKSAAFEDEAKYMFEAEDKH SGKLI IEGIRLKFL PLKDVTAKEKESAVFTVELSHD I RVKWFKNDQRLHTTRSVSMQDEGKTHSITFKDLSIDDTSQIRVEAMGMSSEAKLTVLEGDPYFTGKLQDY TGVEKDEVILQCEISKADAPVKWFKDGKEIKPSKNAVIKTDGKKRMLILKKALKSDIGQYTCDCGTDKTS GKLDIEDREIKLVRPLHSVEVMETETARFETEISEDDIHANWKLKGEALLQTPDCEIKEEGKIHSLVLHN CRLDQTGGVDFQAANVKSSAHLRVKPRVIGLLRPLKDVTVTAGETATFDCELSYEDIPVEWYLKGKKLEP SDKWPRSEGKVHTLTLRDVKLEDAGEVQLTAKDFKTHANLFVKEPPVEFTKPLEDQTVEEGATAVLECE VSRENAKVKWFKNGTEILKSKKYEIVADGRVRKLVIHDCTPEDIKTYTCDAKDFKTSCNLNWPPHVEFL RPLTDLQVREKEMARFECELSRENAKVKWFKDGAEIKKGKKYDI ISKGAVRILVINKCLLDDEAEYSCEV RTAR SGMLTVLEEEAVFTKNLA IEVSETD IKLVCEVSKPGAEVIWYKGDEEI IETGRYEILTEGRKR ILVIQNAHLEDAGNYNCRLPSSRTDGKVKVHELAAEFISKPQNLEILEGEKAEFVCSISKESFPVQWKRD DKTLESGDKYDVIADGKKRVLWKDATLQDMGTYWMVGAARAAAHLTVIEKLRIWPLKDTRVKEQQEV VFNCEVNTEGAKAKWFRNEEAIFDSSKYI ILQKDLVYTLRIRDAHLDDQANYNVSLTNHRGENVKSAANL IVEEEDLRIVEPLKDIETMEKKSVTFWCKVNRLNVTLKWTKNGEEVPFDNRVSYRVDKYKHMLTIKDCGF PDEGEYIVTAGQDKSVAELLI IEAPTEFVEHLEDQTVTEFDDAVFSCQLSREKANVKWYRNGREIKEGKK YKFEKDGSIHRLI IKDCRLDDECEYACGVEDRKSRARLFVEEIPVEI IRPPQDILEAPGADWFLAELNK DKVEVQWLRNNMVWQGDKHQMMSEGKIHRLQICDIKPRDQGEYRFIAKDKEARAKLELAAAPKIKTADQ DLWDVGKPLTMWPYDAYPKAEAEWFKENEPLSTKTIDTTAEQTSFRILEAKKGDKGRYKIVLQNKHGK AEGFINLKVIDVPGPVRNLEVTETFDGEVSLAWEEPLTDGGSKI IGYWERRDIKRKTWVLATDRAESCE FTVTGLQKGGVEYLFRVSARNRVGTGEPVETDNPVEARSKYDVPGPPLNVTITDVNRFGVSLTWEPPEYD GGAEI NYVIELRDK SIRWDTAMTVRAEDLSATVTDWEGQEYSFRVRAQNRIGVGKPSAA PFVKVAD PIERPSPPVNL SSDQTQSSVQLKWEPPLKDGGSPILGYI IERCEEGKDNWIRCNMKLVPELTYKVTGLE KGNKYLYRVSAENKAGVSDPSEILGPLTADDAFVEPTMDLSAFKDGLEVIVPNPITILVPSTGYPRPTAT WCFGDKVLETGDRVKMKTLSAYAELVISPSERSDKGIYTLKLENRVKTISGEIDVNVIARPSAPKELKFG DITKDSVHLTWEPPDDDGGSPLTGYWEKREVSRKTWTKVMDFVTDLEFTVPDLVQGKEYLFKVCARNKC GPGEPAYVDEPVNMSTPATVPDPPENVKWRDRTANSIFLTWDPPKNDGGSRIKGYIVERCPRGSDKWVAC GEPVAETKMEVTGLEEGKWYAYRVKTLNRQGASKPSRPTEEIQAVDTQEAPEIFLDVKLLAGLTVKAGTK IELPATVTGKPEPKI WTKADMILKQDKRI IENVPKKSTV IVDSKRSDTGTYI IEAVNVCGRATAWE VNVLDKPGPPAAFDITDVTNESCLLTWNPPRDDGGSKITNYWERRATDSEVWHKLSSTVKDTNFKATKL IPNKEYIFRVAAENMYGAGEPVQASPITAKYQFDPPGPPTRLEPSDITKDAVTLTWCEPDDDGGSPITGY WVERLDPDTDKWVRCNKMPVKDTTYRVKGLTNKKKYRFRVLAENLAGPGKPSKSTEPILIKDPIDPPWPP GKPTVKDVGKTSVRLNWTKPEHDGGAKIESYVIEMLKTGTDEWVRVAEGVPTTQHLLPGLMEGQEYSFRV RAVNKAGESEPSEPSDPVLCREKLYPPSPPRWLEVI I KNTADLKWTVPEKDGGSPI NYIVEKRDVRR KGWQTVDTTVKDTKCTVTPLTEGSLYVFRVAAENAIGQSDYTEIEDSVLAKDTFTTPGPPYALAWDVTK RHVDLKWEPPKNDGGRPIQRYVIEKKERLGTRWVKAGKTAGPDCNFRVTDVIEGTEVQFQVRAENEAGVG HPSEPTEILSIEDPTSPPSPPLDLHVTDAGRKHIAIAWKPPEKNGGSPI IGYHVEMCPVGTEKWMRVNSR PIKDLKFKVEEGWPDKEYVLRVRAVNAIGVSEPSEISENWAKDPDCKP IDLETHDI IVIEGEKLSIP VPFRAVPVPTVSWHKDGKEVKASDRLTMKNDHISAHLEVPKSVRADAGIY I LENKLGSATASINVKVI GLPGPCKDIKASDITKSSCKLTWEPPEFDGGTPILHYVLERREAGRRTYIPVMSGENKLSWTVKDLIPNG EYFFRVKAVNKVGGGEYIELKNPVIAQDPKQPPDPPVDVEVHNPTAEAM I WKPPLYDGGSKIMGYI IE KIAKGEERWKRCNEHLVPILTYTAKGLEEGKEYQFRVRAENAAGISEPSRATPPTKAVDPIDAPKVILRT SLEVKRGDEIALDASISGSPYPTITWIKDENVIVPEEIKKRAAPLVRRRKGEVQEEEPFVLPLTQRLSID NSKKGESQLRVRDSLRPDHGLYMIKVENDHGIAKAPCTVSVLDTPGPPINFVFEDIRKTSVLCKWEPPLD DGGSEI INYTLEKKDKTKPDSEWIW S LRHCKYSVTKLIEGKEYLFRVRAENRFGPGPPCVSKPLVAK DPFGPPDAPDKPIVEDVTSNSMLVKWNEPKDNGSPILGYWLEKREVNSTHWSRVNKSLLNALKANVDGLL EGLTYVFRVCAENAAGPGKFSPPSDPKTAHDPISPPGPPIPRVTDTSSTTIELEWEPPAFNGGGEIVGYF VDKQLVGTNKWSRCTEKMIKVRQYTVKEIREGADYKLRVSAVNAAGEGPPGETQPVTVAEPQEPPAVELD VSVKGGIQIMAGKTLRIPAWTGRPVPTKVWTKEEGELDKDRWIDNVGTKSELI IKDALRKDHGRYVI ATNSCGSKFAAARVEVFDVPGPVLDLKPWTNRKMCLLNWSDPEDDGGSEI G I IERKDAKMHTWRQPI ETERSKCDITGLLEGQEYKFRVIAKNKFGCGPPVEIGPILAVDPLGPPTSPERLTYTERQRSTITLDWKE PRSNGGSPIQGYI IEKRRHDKPDFERVNKRLCPT SFLVENLDEHQMYEFRVKAVNEIGESEPSLPLNW IQDDEVPPTIKLRLSVRGDTIKVKAGEPVHIPADVTGLPMPKIEWSKNETVIEKPTDALQITKEEVSRSE AKTELSIPKAVREDKGTYTVTASNRLGSVFRNVHVEVYDRPSPPRNLAVTDIKAESCYLTWDAPLDNGGS EITHYVIDKRDASRKKAEWEEVTNTAVEKRYGIWKLIPNGQYEFRVRAVNKYGISDECKSDKWIQDPYR LPGPPGKPKVLARTKGSMLVSWTPPLDNGGSPITGYWLEKREEGSPYWSRVSRAPITKVGLKGVEFNVPR LLEGVKYQFRAMAINAAGIGPPSEPSDPEVAGDPIFPPGPPSCPEVKDKTKSSISLGWKPPAKDGGSPIK GYIVEMQEEGTTDWKRVNEPDKLITTCECWPNLKELRKYRFRVKAVNEAGESEPSDTTGEIPATDIQEE PEVFIDIGAQDCLVCKAGSQIRIPAVIKGRPTPKSSWEFDGKAKKAMKDGVHDIPEDAQLETAENSSVI I IPECKRSHTGKYSITAKNKAGQKTANCRVKVMDVPGPPKDLKVSDITRGSCRLSWKMPDDDGGDRIKGYV IEKRTIDGKAWTKVNPDCGSTTFWPDLLSEQQYFFRVRAENRFGIGPPVETIQRTTARDPIYPPDPPIK LKIGLITKNTVHLSWKPPKNDGGSPVTHYIVECLAWDPTGTKKEAWRQCNKRDVEELQFTVEDLVEGGEY EFRVKAVNAAGVSKPSATVGPCDCQRPDMPPSIDLKEFMEVEEGTNVNIVAKIKGVPFPTLTWFKAPPKK PDNKEPVLYDTHVNKLWDDTCTLVIPQSRRSDTGLYTITAVNNLGTASKEMRLNVLGRPGPPVGPIKFE SVSADQMTLSWFPPKDDGGSKITNYVIEKREANRKTWVHVSSEPKECTYTIPKLLEGHEYVFRIMAQNKY GIGEPLDSEPETARNLFSVPGAPDKPTVSSVTRNSMTVNWEEPEYDGGSPVTGYWLEMKDTTSKRWKRVN RDPIKAMTLGVSYKVTGLIEGSDYQFRVYAINAAGVGPASLPSDPATARDPIAPPGPPFPKVTDWTKSSA DLEWSPPLKDGGSKVTGYIVEYKEEGKEEWEKGKDKEVRGTKLWTGLKEGAFYKFRVSAVNIAGIGEPG EVTDVIEMKDRLVSPDLQLDASVRDRIWHAGGVIRI IAYVSGKPPPTVTWNMNERTLPQEA IETTAIS SSMVIKNCQRSHQGVYSLLAKNEAGERKK I IVDVLDVPGPVG PFLAHNLTNESCKLTWFSPEDDGGSP ITNYVIEKRESDRRAWTPVTYTVTRQNATVQGLIQGKAYFFRIAAENSIGMGPFVETSEALVIREPITVP ERPEDLEVKEVTKNTVTLTWNPPKYDGGSEI INYVLESRLIGTEKFHKVTNDNLLSRKYTVKGLKEGDTY EYRVSAVNIVGQGKPSFCTKPITCKDELAPPTLHLDFRDKLTIRVGEAFALTGRYSGKPKPKVSWFKDEA DVLEDDRTHIKTTPATLALEKIKAKRSDSGKYCVWENSTGSRKGFCQVNWDHPGPPVGPVSFDEVTKD YMVISWKPPLDDGGSKI NYI IEKKEVGKDVWMPV SASAKTTCKVSKLLEGKDYIFRIHAENLYGISDP LVSDSMKAKDRFRVPDAPDQPIVTEVTKDSALVTWNKPHDGGKPITNYILEKRETMSKRWARVTKDPIHP YTKFRVPDLLEGCQYEFRVSAENEIGIGDPSPPSKPVFAKDPIAKPSPPVNPEAIDTTCNSVDLTWQPPR HDGGSKILGYIVEYQKVGDEEWRRANHTPESCPETKYKVTGLRDGQTYKFRVLAVNAAGESDPAHVPEPV LVKDRLEPPELILDANMAREQHIKVGDTLRLSAI IKGVPFPKVTWKKEDRDAPTKARIDV PVGSKLEIR NAAHEDGGIYSLTVENPAGSKTVSVKVLVLDKPGPPRDLEVSEIRKDSCYLTWKEPLDDGGSVITNYWE RRDVASAQWSPLSATSKKKSHFAKHLNEGNQYLFRVAAENQYGRGPFVETPKPIKALDPLHPPGPPKDLH HVDVDKTEVSLVWNKPDRDGGSPITGYLVEYQEEGTQDWIKFKTVTNLECWTGLQQGKTYRFRVKAENI VGLGLPDT IPIECQEKLVPPSVELDVKLIEGLWKAGTTVRFPAI IRGVPVPTAKWTTDGSEIKTDEHY TVETDNFSSVLTIKNCLRRDTGEYQITVSNAAGSKTVAVHLTVLDVPGPPTGPINILDVTPEHMTISWQP PKDDGGSPVINYIVEKQDTRKDTWGWSSGSSKTKLKIPHLQKGCEYVFRVRAENKIGVGPPLDSTPTVA KHKFSPPSPPGKPWTDITENAATVSWTLPKSDGGSPITGYYMERREVTGKWVRVNKTPIADLKFRVTGL YEGNTYEFRVFAENLAGLSKPSPSSDPIKACRPIKPPGPPINPKLKDKSRETADLVWTKPLSDGGSPILG YWECQKPGTAQWNRINKDELIRQCAFRVPGLIEGNEYRFRIKAANIVGEGEPRELAESVIAKDILHPPE VELDVTCRDVITVRVGQTIRILARVKGRPEPDITWTKEGKVLVREKRVDLIQDLPRVELQIKEAVRADHG KYI ISAKNSSGHAQGSAIVNVLDRPGPCQNLKVTNVTKENC ISWENPLDNGGSEI NFIVEYRKPNQKG
- I l l - WSIVASDVTKRLIKANLLANNEYYFRVCAENKVGVGPTIETKTPILAINPIDRPGEPENLHIADKGKTFV YLKWRRPDYDGGSPNLSYHVERRLKGSDDWERVHKGSIKETHYMVDRCVENQIYEFRVQTKNEGGESDWV KTEEVWKEDLQKPVLDLKLSGVLTVKAGDTIRLEAGVRGKPFPEVAWTKDKDATDLTRSPRVKIDTRAD SSKFSLTKAKRSDGGKYWTATNTAGSFVAYATVNVLDKPGPVRNLKIVDVSSDRCTVCWDPPEDDGGCE IQNYILEKCETKRMVWSTYSATVLTPGTTVTRLIEGNEYIFRVRAENKIGTGPPTESKPVIAKTKYDKPG RPDPPEVTKVSKEEMTWWNPPEYDGGKSITGYFLEKKEKHSTRWVPVNKSAIPERRMKVQNLLPDHEYQ FRVKAENEIGIGEPSLPSRPWAKDPIEPPGPPTNFRWDTTKHSI LGWGKPVYDGGAPI IGYWEMRP KIADASPDEGWKRCNAAAQLVRKEFTVTSLDENQEYEFRVCAQNQVGIGRPAELKEAIKPKEILEPPEID LDASMRKLVIVRAGCPIRLFAIVRGRPAPKVTWRKVGIDNWRKGQVDLVDTMAFLVIPNSTRDDSGKYS LTLVNPAGEKAVFVNVRVLDTPGPVSDLKVSDVTKTSCHVSWAPPENDGGSQVTHYIVEKREADRKTWST VTPEVKKTSFHVTNLVPGNEYYFRVTAVNEYGPGVPTDVPKPVLASDPLSEPDPPRKLEATEMTKNSATL AWLPPLRDGGAKIDGYI ISYREEEQPADRWTEYSWKDLSLWTGLKEGKKYKFRVAARNAVGVSLPREA EGVYEAKEQLLPPKILMPEQITIKAGKKLRIEAHVYGKPHPTCKWKKGEDEWTSSHLAVHKADSSSILI IKDVTRKDSGYYSLTAENSSGTDTQKIKVWMDAPGPPQPPFDISDIDADACSLSWHIPLEDGGSNITNY IVEKCDVSRGDWVTALASVTKTSCRVGKLIPGQEYIFRVRAENRFGISEPLTSPKMVAQFPFGVPSEPKN ARVTKVNKDCIFVAWDRPDSDGGSPI IGYLIERKERNSLLWVKANDTLVRSTEYPCAGLVEGLEYSFRIY ALNKAGSSPPSKPTEYVTARMPVDPPGKPEVIDVTKS VSLIWARPKHDGGSKI IGYFVEACKLPGDKWV RCNTAPHQIPQEEYTATGLEEKAQYQFRAIARTAV ISPPSEPSDPV ILAENVPPRIDLSVAMKSLLTV KAGTNVCLDATVFGKPMPTVSWKKDGTLLKPAEGIKMAMQRNLCTLELFSVNRKDSGDYTITAENSSGSK SATIKLKVLDKPGPPASVKINKMYSDRAMLSWEPPLEDGGSEITNYIVDKRETSRPNWAQVSATVPITSC SVEKLIEGHEYQFRICAENKYGVGDPVFTEPAIAKNPYDPPGRCDPPVISNITKDHMTVSWKPPADDGGS PI GYLLEKRETQAVNWTKVNRKPI IERTLKATGLQEGTEYEFRVTAINKAGPGKPSDASKAAYARDPQY PPAPPAFPKVYDTTRSSVSLSWGKPAYDGGSPI IGYLVEVKRADSDNWVRCNLPQNLQKTRFEVTGLMED TQYQFRVYAVNKIGYSDPSDVPDKHYPKDILIPPEGEHDADLRKTLILRAGVTMRLYVPVKGRPPPKI W SKPNVNLRDRIGLDIKSTDFDTFLRCENVNKYDAGKYILTLENSCGKKEYTIWKVLDTPGPPINVTVKE ISKDSAYVTWEPPI IDGGSPI INYWQKRDAERKSWS VTTECSK SFRVPNLEEGKSYFFRVFAENEYG IGDPGETRDAVKASQ PGPWDLKVRSVSKSSCSIGWKKPHSDGGSRI IGYWDFLTEENKWQRVMKSLS LQYSAKDLTEGKEYTFRVSAENENGEGTPSEITWARDDWAPDLDLKGLPDLCYLAKENSNFRLKIPIK GKPAPSVSWKKGEDPLATDTRVSVESSAVNTTLIVYDCQKSDAGKYTITLKNVAGTKEGTISIKWGKPG IPTGPIKFDEVTAEAMTLKWAPPKDDGGSEITNYILEKRDSVNNKWVTCASAVQKTTFRVTRLHEGMEYT FRVSAENKYGVGEGLKSEPIVARHPFDVPDAPPPPNIVDVRHDSVSLTWTDPKKTGGSPITGYHLEFKER NSLLWKRANK PIRMRDFKVTGLTEGLEYEFRVMAINLAGVGKPSLPSEPWALDPIDPPGKPEVI I R NSVTLIWTEPKYDGGHKLTGYIVEKRDLPSKSWMKANHVNVPECAFTVTDLVEGGKYEFRIRAKNTAGAI SAPSES E I ICKDEYEAP IVLDP IKDGL IKAGD IVLNAISILGKPLPKSSWSKAGKDIRPSDI Q ITSTPTSSMLTIKYATRKDAGEYTITATNPFGTKVEHVKVTVLDVPGPPGPVEISNVSAEKATLTWTPPL EDGGSPIKSYILEKRETSRLLWTWSEDIQSCRHVATKLIQGNEYIFRVSAVNHYGKGEPVQSEPVKMVD RFGPPGPPEKPEVSNVTKNTATVSWKRPVDDGGSEITGYHVERREKKSLRWVRAIKTPVSDLRCKVTGLQ EGSTYEFRVSAENRAGIGPPSEASDSVLMKDAAYPPGPPSNPHVTDTTKKSASLAWGKPHYDGGLEITGY WEHQKVGDEAWIKDTTGTALRITQFWPDLQTKEKYNFRISAINDAGVGEPAVIPDVEIVEREMAPDFE LDAELRRTLWRAGLSIRIFVPIKGRPAPEVTWTKD INLKNRA IENTESFTLLI IPECNRYDTGKFVM TIENPAGKKSGFVNVRVLDTPGPVLNLRPTDITKDSVTLHWDLPLIDGGSRITNYIVEKREATRKSYSTA TTKCHKCTYKVTGLSEGCEYFFRVMAENEYGIGEPTETTEPVKASEAPSPPDSLNIMDITKSTVSLAWPK PKHDGGSKITGYVIEAQRKGSDQWTHITTVKGLECWRNLTEGEEYTFQVMAVNSAGRSAPRESRPVIVK EQTMLPELDLRGIYQKLVIAKAGDNIKVEIPVLGRPKPTVTWKKGDQILKQTQRVNFETTATSTILNINE CVRSDSGPYPLTARNIVGEVGDVITIQVHDIPGPPTGPIKFDEVSSDFVTFSWDPPENDGGVPISNYWE MRQTDSTTWVELATTVIRTTYKATRLTTGLEYQFRVKAQNRYGVGPGITSAWIVANYPFKVPGPPGTPQV TAVTKDSMTISWHEPLSDGGSPILGYHVERKERNGILWQTVSKALVPGNIFKSSGLTDGIAYEFRVIAEN MAGKSKPSKPSEPMLALDPIDPPGKPVPLNITRHTVTLKWAKPEYTGGFKITSYIVEKRDLPNGRWLKAN FSNILENEFTVSGLTEDAAYEFRVIAKNAAGAISPPSEPSDAITCRDDVEAPKIKVDVKFKDTVILKAGE AFRLEADVSGRPPPTMEWSKDGKELEGTAKLEIKIADFSTNLVNKDSTRRDSGAYTLTATNPGGFAKHIF NVKVLDRPGPPEGPLAVTEVTSEKCVLSWFPPLDDGGAKIDHYIVQKRETSRLAWTNVASEVQVTKLKVT KLLKGNEYIFRVMAVNKYGVGEPLESEPVLAVNPYGPPDPPKNPEVT I KDSMWCWGHPDSDGGSEI I NYIVERRDKAGQRWIKCNKKTLTDLRYKVSGLTEGHEYEFRIMAENAAGISAPSPTSPFYKACDTVFKPG PPGNPRVLDTSRSSISIAWNKPIYDGGSEITGYMVEIALPEEDEWQIVTPPAGLKATSYTITGLTENQEY KIRIYAMNSEGLGEPALVPGTPKAEDRMLPPEIELDADLRKWTIRACCTLRLFVPIKGRPDPEVKWARD HGESLDKASIESASSYTLLIVGNVNRFDSGKYILTVENSSGSKSAFVNVRVLDTPGPPQDLKVKEVTKTS VTLTWDPPLLDGGSKIKNYIVEKRESTRKAYSTVATNCHKTSWKVDQLQEGCSYYFRVLAENEYGIGLPA ETAESVKASERPLPPGKITLMDVTRNSVSLSWEKPEHDGGSRILGYIVEMQTKGSDKWATCATVKVTEAT ITGLIQGEEYSFRVSAQNEKGISDPRQLSVPVIAKDLVIPPAFKLLFNTFTVLAGEDLKVDVPFIGRPTP AVTWHKDNVPLKQTTRVNAESTENNSLLTIKDACREDVGHYWKLTNSAGEAIETLNVIVLDKPGPPTGP VKMDEVTADSITLSWGPPKYDGGSSINNYIVEKRDTSTTTWQIVSATVARTTIKACRLKTGCEYQFRIAA ENRYGKSTYLNSEPTVAQYPFKVPGPPGTPWTLSSRDSMEVQWNEPISDGGSRVIGYHLERKERNSILW VKLNK PIPQTKFKTTGLEEGVEYEFRVSAE IVGIGKPSKVSECYVARDPCDPPGRPEAI IVTRNSVTL QWKKPTYDGGSKI GYIVEKKELPEGRWMKASFT I IDTHFEVTGLVEDHRYEFRVIARNAAGVFSEPSE STGAITARDEVDPPRISMDPKYKDTIWHAGESFKVDADIYGKPIPTIQWIKGDQELSNTARLEIKSTDF ATSLSVKDAVRVDSGNYILKAKNVAGERSVTVNVKVLDRPGPPEGPWISGVTAEKCTLAWKPPLQDGGS DIINYIVERRETSRLVWTWDANVQTLSCKVTKLLEGNEYTFRIMAVNKYGVGEPLESEPWAKNPFWP DAPKAPEVTTVTKDSMIWWERPASDGGSEILGYVLEKRDKEGIRWTRCHKRLIGELRLRVTGLIENHDY EFRVSAENAAGLSEPSPPSAYQKACDPIYKPGPPNNPKVIDITRSSVFLSWSKPIYDGGCEIQGYIVEKC DVNVGEWTMC PPTGINKT IEVEKLLEKHEYNFRICAINKAGVGEHADVPGPI IVEEKLEAPDIDLDLE LRKI I IRAGGSLRLFVPIKGRP PEVKWGKVDGEIRDAAI IDV SSF SLVLDNVNRYDSGKYTLTLEN SSGTKSAFVTVRVLDTPSPPVNLKVTEITKDSVSITWEPPLLDGGSKIKNYIVEKREATRKSYAAWTNC HKNSWKIDQLQEGCSYYFRVTAENEYGIGLPAQTADPIKVAEVPQPPGKITVDDVTRNSVSLSWTKPEHD GGSKI IQYIVEMQAKHSEKWSECARVKSLQAVI NLTQGEEYLFRWAVNEKGRSDPRSLAVPIVAKDLV IEPDVKPAFSSYSVQVGQDLKMEVPISGRPKPTITWTKDGLPLKQTTRINVTDSLDLTTLSIKETHKDDG GQYGITVANWGQKTASIEIVTLDKPDPPKGPVKFDDVSAESITLSWNPPLYTGGCQITNYIVQKRDTTT TVWDWSATVARTTLKVTKLKTGTEYQFRIFAENRYGQSFALESDPIVAQYPYKEPGPPGTPFATAISKD SMVIQWHEPVNNGGSPVIGYHLERKERNSILWTKVNK I IHDTQFKAQNLEEGIEYEFRVYAE IVGVGK ASKNSECYVARDPCDPPGTPEPIMVKRNEITLQWTKPVYDGGSMITGYIVEKRDLPDGRWMKASFTNVIE TQFTVSGLTEDQRYEFRVIAKNAAGAISKPSDSTGPITAKDEVELPRISMDPKFRDTIWNAGETFRLEA DVHGKPLPTIEWLRGDKEIEESARCEIKNTDFKALLIVKDAIRIDGGQYILRASNVAGSKSFPVNVKVLD RPGPPEGPVQVTGVTSEKCSLTWSPPLQDGGSDISHYWEKRETSRLAWTWASEWTNSLKVTKLLEGN EYVFRIMAVNKYGVGEPLESAPVLMKNPFVLPGPPKSLEVT IAKDSMTVCWNRPDSDGGSEI IGYIVEK RDRSGIRWIKCNKRRITDLRLRVTGLTEDHEYEFRVSAENAAGVGEPSPATVYYKACDPVFKPGPPTNAH IVDTTKNSITLAWGKPIYDGGSEILGYWEICKADEEEWQIVTPQTGLRVTRFEISKLTEHQEYKIRVCA LNKVGLGEATSVPGTVKPEDKLEAPELDLDSELRKGIWRAGGSARIHIPFKGRPMPEITWSREEGEFTD KVQIEKGVNYTQLSIDNCDRNDAGKYILKLENSSGSKSAFVTVKVLDTPGPPQNLAVKEVRKDSAFLVWE PPI IDGGAKVKNYVIDKRES RKAYANVSSKCSK SFKVENLTEGAIYYFRVMAENEFGVGVPVETVDAV KAAEPPSPPGKVTLTDVSQTSASLMWEKPEHDGGSRVLGYWEMQPKGTEKWSIVAESKVCNAWTGLSS GQEYQFRVKAYNEKGKSDPRVLGVPVIAKDLTIQPSLKLPFNTYSIQAGEDLKIEIPVIGRPRPNISWVK DGEPLKQTTRVNVEETATSTVLHIKEGNKDDFGKYTVTATNSAGTATENLSVIVLEKPGPPVGPVRFDEV SADFWISWEPPAYTGGCQISNYIVEKRDTTTTTWHMVSATVARTTIKITKLKTGTEYQFRIFAENRYGK SAPLDSKAVIVQYPFKEPGPPG PFV SISKDQMLVQWHEPVNDGGTKI IGYHLEQKEKNSILWVKLNKT PIQDTKFKTTGLDEGLEYEFKVSAENIVGIGKPSKVSECFVARDPCDPPGRPEAIVITRNNVTLKWKKPA YDGGSKITGYIVEKKDLPDGRWMKASFTNVLETEFTVSGLVEDQRYEFRVIARNAAGNFSEPSDSSGAIT ARDEIDAPNASLDPKYKDVIWHAGETFVLEADIRGKPIPDWWSKDGKELEETAARMEIKSTIQKTTLV VKDCIRTDGGQYILKLSNVGGTKSIPITVKVLDRPGSPEGPLKVTGVTAEKCYLAWNPPLQDGGANISHY I IEKRE SRLSWTQVS EVQALNYKVTKLLPGNEYIFRVMAVNKYGIGEPLESGPVTACNPYKPPGPPS PEVSAITKDSMWTWARPVDDGGTEIEGYILEKRDKEGVRWTKCNKKTLTDLRLRVTGLTEGHSYEFRVA AENAAGVGEPSEPSVFYRACDALYPPGPPSNPKVTDTSRSSVSLAWSKPIYDGGAPVKGYWEVKEAAAD EWTTCTPPTGLQGKQFTVTKLKENTEYNFRICAINSEGVGEPATLPGSWAQERIEPPEIELDADLRKW VLRASATLRLFVTIKGRPEPEVKWEKAEGILTDRAQIEVTSSFTMLVIDNVTRFDSGRYNLTLENNSGSK TAFVNVRVLDSPSAPVNLTIREVKKDSVTLSWEPPLIDGGAKITNYIVEKRETTRKAYATITNNCTKTTF RIENLQEGCSYYFRVLASNEYGIGLPAETTEPVKVSEPPLPPGRVTLVDVTRNTATIKWEKPESDGGSKI TGYWEMQTKGSEKWSTCTQVKTLEATISGLTAGEEYVFRVAAVNEKGRSDPRQLGVPVIARDIEIKPSV ELPFHTFNVKAREQLKIDVPFKGRPQATVNWRKDGQTLKETTRVNVSSSKTVTSLSIKEASKEDVGTYEL CVSNSAGSITVPITIIVLDRPGPPGPIRIDEVSCDSITISWNPPEYDGGCQISNYIVEKKETTSTTWHIV SQAVARTSIKIVRLTTGSEYQFRVCAENRYGKSSYSESSAWAEYPFSPPGPPGTPKWHATKSTMLVTW QVPVNDGGSRVIGYHLEYKERSSILWSKANKILIADTQVKVSGLDEGLMYEYRVYAENIAGIGKCSKSCE PVPARDPCDPPGQPEVTNITRKSVSLKWSKPHYDGGAKITGYIVERRELPDGRWLKCNYTNIQETYFEVT ELTEDQRYEFRVFARNAADSVSEPSES GPI IVKDDVEPPRVMMDVKFRDVIWKAGEVLKINADIAGRP LPVISWAKDGIEIEERARTEI ISTDNHTLLTVKDCIRRDTGQYVLTLKNVAGTRSVAVNCKVLDKPGPPA GPLEINGLTAEKCSLSWGRPQEDGGADIDYYHRKKRETSHLAWTICEGELQMTSCKVTKLLKGNEYIFRV TGVNKYGVGEPLESVAIKALDPFTVPSPPTSLEI SVTKESMTLCWSRPESDGGSEISGYI IERREKNSL RWVRVNKKPVYDLRVKSTGLREGCEYEYRVYAENAAGLSLPSETSPLIRAEDPVFLPSPPSKPKIVDSGK TTITIAWVKPLFDGGAPITGYTVEYKKSDDTDWKTSIQSLRGTEYTISGLTTGAEYVFRVKSVNKVGASD PSDSSDPQIAKEREEEPLFDIDSEMRKTLIVKAGASFTMTVPFRGRPVPNVLWSKPDTDLRTRAYVDTTD SRTSLTIENANRNDSGKYTLTIQNVLSAASLTLWKVLDTPGPPTNITVQDVTKESAVLSWDVPENDGGA PVKNYHIEKREASKKAWVSVTNNCNRLSYKVTNLQEGAIYYFRVSGENEFGVGIPAETKEGVKITEKPSP PEKLGVTSISKDSVSLTWLKPEHDGGSRIVHYWEALEKGQKNWVKCAVAKSTHHWSGLRENSEYFFRV FAENQAGLSDPRELLLPVLIKEQLEPPEIDMKNFPSHTVYVRAGSNLKVDIPISGKPLPKVTLSRDGVPL KATMRFNTEITAENLTINLKESVTADAGRYEITAANSSGTTKAFINIWLDRPGPPTGPWISDITEESV TLKWEPPKYDGGSQVTNYILLKRETSTAVWTEVSATVARTMMKVMKLTTGEEYQFRIKAENRFGISDHID SACVTVKLPYTTPGPPSTPWVTNVTRESITVGWHEPVSNGGSAWGYHLEMKDRNSILWQKANKLVIRTT HFKVTTISAGLIYEFRVYAENAAGVGKPSHPSEPVLAIDACEPPRNVRITDISKNSVSLSWQQPAFDGGS KITGYIVERRDLPDGRWTKASFTNVTETQFTISGLTQNSQYEFRVFARNAVGSISNPSEWGPITCIDSY GGPVIDLPLEYTEWKYRAGTSVKLRAGISGKPAPTIEWYKDDKELQTNALVCVENTTDLASILIKDADR LNSGCYELKLRNAMASASATIRVQILDKPGPPGGPIEFKTVTAEKITLLWRPPADDGGAKITHYIVEKRE SRWWSMVSEHLEECI ITTTKI IKGNEYIFRVRAVNKYGIGEPLESDSWAKNAFV PGPPGIPEVTKI TKNSMTWWSRPIADGGSDISGYFLEKRDKKSLGWFKVLKETIRDTRQKVTGLTENSDYQYRVCAVNAAG QGPFSEPSEFYKAADPIDPPGPPAKIRIADSTKSSITLGWSKPVYDGGSAVTGYWEIRQGEEEEWTTVS TKGEVRTTEYWSNLKPGVNYYFRVSAVNCAGQGEPIEMNEPVQAKDILEAPEIDLDVALRTSVIAKAGE DVQVLIPFKGRPPPTVTWRKDEKNLGSDARYSIENTDSSSLLTIPQVTRNDTGKYILTIENGVGEPKSST VSVKVLDTPAACQKLQVKHVSRGTVTLLWDPPLIDGGSPI INYVIEKRDATKRTWSWSHKCSSTSFKLI DLSEKTPFFFRVLAENEIGIGEPCETTEPVKAAEVPAPIRDLSMKDSTKTSVILSWTKPDFDGGSVITEY WERKGKGEQTWSHAGISKTCEIEVSQLKEQSVLEFRVFAKNEKGLSDPV IGPI VKELI ITPEVDLSD IPGAQVTVRIGHNVHLELPYKGKPKPSISWLKDGLPLKESEFVRFSKTENKITLSIKNAKKEHGGKYTVI LDNAVCRIAVPITVITLGPPSKPKGPIRFDEIKADSVILSWDVPEDNGGGEITCYSIEKRETSQTNWKMV CSSVARTTFKVPNLVKDAEYQFRVRAENRYGVSQPLVSSI IVAKHQFRIPGPPGKPVIYNVTSDGMSLTW DAPVYDGGSEVTGFHVEKKERNSILWQKVNTSPISGREYRATGLVEGLDYQFRVYAENSAGLSSPSDPSK FTLAVSPVDPPGTPDYIDVTRETITLKWNPPLRDGGSKIVGYSIEKRQGNERWVRCNFTDVSECQYTVTG LSPGDRYEFRI IARNAVG I SPPSQSSGI IMTRDENVPPIVEFGPEYFDGLI IKSGESLRIKALVQGRPV PRVTWFKDGVEIEKRMNMEITNVLGSTSLFVRDATRDHRGVYTVEAKNASGSAKAEIKVKVQDTPGKWG PIRFT I GEKMTLWWDAPLNDGCAPI HYI IEKRE SRLAWALIEDKCEAQSYTAIKLINGNEYQFRVS AVNKFGVGRPLDSDPWAQIQYTVPDAPGIPEPSNITGNSITLTWARPESDGGSEIQQYILERREKKSTR WVKVISKRPISETRFKVTGLTEGNEYEFHVMAENAAGVGPASGISRLIKCREPVNPPGPPTWKVTDTSK TTVSLEWSKPVFDGGMEI IGYI IEMCKTDLGDWHKVNAEACVKTRYTVTDLQAGEEYKFRVSAINGAGKG DSCEVTG IKAVDRLTAPELDIDANFKQTHWRAGASIRLFIAYQGRP PTAVWSKPDSNLSLRADIHTT DSFSTLTVENCNRNDAGKYTLTVENNSGSKSITFTVKVLDTPGPPGPITFKDVTRGSATLMWDAPLLDGG ARIHHYWEKREASRRSWQVISEKCTRQIFKVNDLAEGVPYYFRVSAVNEYGVGEPYEMPEPIVATEQPA PPRRLDWDTSKSSAVLAWLKPDHDGGSRITGYLLEMRQKGSDLWVEAGHTKQLTFTVERLVEKTEYEFR VKAKNDAGYSEPREAFSSVI IKEPQIEPTADLTGI NQLI CKAGSPF IDVPISGRPAPKVTWKLEEMR LKETDRVSITTTKDRTTLTVKDSMRGDSGRYFLTLENTAGVKTFSVTVWIGRPGPVTGPIEVSSVSAES CVLSWGEPKDGGGTEITNYIVEKRESGTTAWQLVNSSVKRTQIKVTHLTKYMEYSFRVSSENRFGVSKPL ESAPI IAEHPFVPPSAPTRPEVYHVSA AMSIRWEEPYHDGGSKI IGYWVEKKERN ILWVKENKVPCLE CNYKVTGLVEGLEYQFRTYALNAAGVSKASEASRPIMAQNPVDAPGRPEVTDVTRSTVSLIWSAPAYDGG SKWGYI IERKPVSEVGDGRWLKCNY IVSDNFFTVTALSEGDTYEFRVLAKNAAGVISKGSES GPVTC RDEYAPPKAELDARLHGDLV IRAGSDLVLDAAVGGKPEPKI IWTKGDKELDLCEKVSLQYTGKRATAVI KFCDRSDSGKYTLTVKNASGTKAVSVMVKVLDSPGPCGKLTVSRVTQEKCTLAWSLPQEDGGAEITHYIV ERRETSRLNWVIVEGECPTLSYWTRLIKNNEYIFRVRAVNKYGPGVPVESEPIVARNSFTIPSPPGIPE EVGTGKEHI I IQWTKPESDGGNEISNYLVDKREKESLRWTRVNKDYWYDTRLKVTSLMEGCDYQFRVTA VNAAGNSEPSERSNFISCREPSYTPGPPSAPRWDTTKHSISLAWTKPMYDGGTDIVGYVLEMQEKDTDQ WYRVHTNATIRNTEFTVPDLKMGQKYSFRVAAVNVKGMSEYSESIAEIEPVERIEIPDLELADDLKKTVT IRAGASLRLMVSVSGRPPPVI WSKQGIDLASRAI IDTTESYSLLIVDKVNRYDAGKY IEAENQSGKKS ATVLVKVYDTPGPCPSVKVKEVSRDSVTITWEIPTIDGGAPINNYIVEKREAAMRAFKTVTTKCSKTLYR ISGLVEGTMHYFRVLPENIYGIGEPCETSDAVLVSEVPLVPAKLEWDVTKSTVTLAWEKPLYDGGSRLT GYVLEACKAGTERWMKWTLKPTVLEHTVTSLNEGEQYLFRIRAQNEKGVSEPRETVTAVTVQDLRVLPT IDLSTMPQKTIHVPAGRPVELVIPIAGRPPPAASWFFAGSKLRESERVTVETHTKVAKLTIRETTIRDTG EYTLELKNVTGTTSETIKVI ILDKPGPPTGPIKIDEIDATSITISWEPPELDGGAPLSGYWEQRDAHRP GWLPVSESVTRSTFKFTRLTEGNEYVFRVAATNRFGIGSYLQSEVIECRSSIRIPGPPETLQIFDVSRDG MTLTWYPPEDDGGSQVTGYIVERKEVRADRWVRVNKVPVTMTRYRSTGLTEGLEYEHRVTAINARGSGKP SRPSKPIVAMDPIAPPGKPQNPRVTDTTRTSVSLAWSVPEDEGGSKVTGYLIEMQKVDQHEWTKCNTTPT KIREYTLTHLPQGAEYRFRVLACNAGGPGEPAEVPGTVKVTEMLEYPDYELDERYQEGIFVRQGGVIRLT IPIKGKPFPICKWTKEGQDISKRAMIATSETHTELVIKEADRGDSGTYDLVLENKCGKKAVYIKVRVIGS PNSPEGPLEYDDIQVRSVRVSWRPPADDGGADILGYILERREVPKAAWYTIDSRVRGTSLWKGLKENVE YHFRVSAENQFGISKPLKSEEPVTPKTPLNPPEPPSNPPEVLDVTKSSVSLSWSRPKDDGGSRVTGYYIE RKE S DKWRHNKTQI TTMYTVTGLVPDAEYQFRI IAQNDVGLSE SPASEPWCKDPFDKPSQPGEL EILSISKDSVTLQWEKPECDGGKEILGYWVEYRQSGDSAWKKSNKERIKDKQFTIGGLLEATEYEFRVFA ENETGLSRPRRTAMSIKTKLTSGEAPGIRKEMKDVTTKLGEAAQLSCQIVGRPLPDIKWYRFGKELIQSR KYKMSSDGRTHTLTVMTEEQEDEGVYTCIATNEVGEVETSSKLLLQATPQFHPGYPLKEKYYGAVGSTLR LHVMYIGRPVPAMTWFHGQKLLQNSENITIENTEHYTHLVMKNVQRKTHAGKYKVQLSNVFGTVDAILDV EIQDKPDKPTGPIVIEALLKNSAVISWKPPADDGGSWITNYWEKCEAKEGAEWQLVSSAISVTTCRIVN LTENAGYYFRVSAQNTFGISDPLEVSSWI IKSPFEKPGAPGKP I AVTKDSCWAWKPPASDGGAKIR NYYLEKREKKQNKWISVTTEEIRETVFSVKNLIEGLEYEFRVKCENLGGESEWSEISEPITPKSDVPIQA PHFKEELRNLNVRYQSNATLVCKVTGHPKPIVKWYRQGKEI IADGLKYRIQEFKGGYHQLI IASVTDDDA TVYQVRATNQGGSVSGTASLEVEVPAKIHLPKTLEGMGAVHALRGEWSIKIPFSGKPDPVITWQKGQDL IDNNGHYQVIVTRSFTSLVFPNGVERKDAGFYWCAKNRFGIDQKTVELDVADVPDPPRGVKVSDASRDS VNLTWTEPASDGGSKITNYIVEKCATTAERWLRVGQARETRYTVINLFGKTSYQFRVIAENKFGLSKPSE PSEPTITKEDKTRAMNYDEEVDETREVSMTKASHSSTKELYEKYMIAEDLGRGEFGIVHRCVETSSKKTY MAKFVKVKGTDQVLVKKEISILNIARHRNILHLHESFESMEELVMIFEFISGLDIFERINTSAFELNERE IVSYVHQVCEALQFLHSHNIGHFDIRPENI IYQTRRSS IKI IEFGQARQLKPGDNFRLLFTAPEYYAPE VHQHDWSTATDMWSLGTLVYVLLSGINPFLAETNQQI IENIMNAEYTFDEEAFKEISIEAMDFVDRLLV KERKSRMTASEALQHPWLKQKIERVSTKVIRTLKHRRYYHTLIKKDLNMWSAARISCGGAIRSQKGVSV AKVKVASIEIGPVSGQIMHAVGEEGGHVKYVCKIENYDQSTQVTWYFGVRQLENSEKYEITYEDGVAILY VKDITKLDDGTYRCKWNDYGEDSSYAELFVKGVREVYDYYCRRTMKKIKRRTDTMRLLERPPEFTLPLY NKTAYVGENVRFGVTITVHPEPHVTWYKSGQKIKPGDNDKKYTFESDKGLYQLTINSVTTDDDAEYTWA RNKYGEDSCKAKLTVTLHPPPTDSTLRPMFKRLLANAECQEGQSVCFEIRVSGIPPPTLKWEKDGQPLSL GPNIEI IHEGLDYYALHIRDTLPEDTGYYRVTATNTAGSTSCQAHLQVERLRYKKQEFKSKEEHERHVQK QIDKTLRMAEILSGTESVPLTQVAKEALREAAVLYKPAVSTKTVKGEFRLEIEEKKEERKLRMPYDVPEP RKYKQTTIEEDQRIKQFVPMSDMKWYKKIRDQYEMPGKLDRWQKRPKRIRLSRWEQFYVMPLPRITDQY RPKWRIPKLSQDDLEIVRPARRRTPSPDYDFYYRPRRRSLGDISDEELLLPIDDYLAMKRTEEERLRLEE ELELGFSASPPSRSPPHFELSSLRYSSPQAHVKVEETRKNFRYSTYHIPTKAEASTSYAELRERHAQAAY RQPKQRQRIMAEREDEELLRPVTTTQHLSEYKSELDFMSKEEKSRKKSRRQREVTEITEIEEEYEISKHA QRESSSSASRLLRRRRSLSPTYIELMRPVSELIRSRPQPAEEYEDDTERRSPTPERTRPRSPSPVSSERS LSRFERSARFDIFSRYESMKAALKTQKTSERKYEVLSQQPFTLDHAPRITLRMRSHRVPCGQNTRFILNV QSKPTAEVKWYHNGVELQESSKIHYTNTSGVLTLEILDCHTDDSGTYRAVCTNYKGEASDYATLDVTGGD YTTYASQRRDEEVPRSVFPELTRTEAYAVPSFKKTSEMEASSSVREVKSQMTETRESLSSYEHSASAEMK SAALEEKSLEEKS TRKIKTTLAARILTKPRSMTVYEGESARFSCDTDGEPVPTVTWLRKGQVLS SARH QVTTTKYKSTFEISSVQASDEGNYSVWENSEGKQEAEFTLTIQKARVTEKAVTSPPRVKSPEPRVKSPE AVKSPKRVKSPEPSHPKAVSPTETKP PREKVQHLPVSAPPKI QFLKAEASKEIAKLTCWESSVLRAK EVTWYKDGKKLKENGHFQFHYSADGTYELKINNLTESDQGEYVCEISGEGGTSKTNLQFMGQAFKSIHEK VSKISETKKSDQKTTESTVTRKTEPKAPEPISSKPVIVTGLQDTTVSSDSVAKFAVKATGEPRPTAIWTK DGKAITQGGKYKLSEDKGGFFLEIHKTDTSDSGLYTCTVKNSAGSVSSSCKLTIKAIKDTEAQKVSTQKT SEITPQKKAWQEEISQKALRSEEIKMSEAKSQEKLALKEEASKVLISEEVKKSAATSLEKSIVHEEITK TSQASEEVRTHAEIKAFSTQMSINEGQRLVLKANIAGATDVKWVLNGVELTNSEEYRYGVSGSDQTLTIK QASHRDEGILTCISKTKEGIVKCQYDLTLSKELSDAPAFISQPRSQNINEGQNVLFTCEISGEPSPEIEW FKNNLPISISSNVSISRSRNVYSLEIRNASVSDSGKYTIKAKNFRGQCSATASLMVLPLVEEPSREWLR TSGDTSLQGSFSSQSVQMSASKQEASFSSFSSSSASSMTEMKFASMSAQSMSSMQESFVEMSSSSFMGIS NMTQLESSTSKMLKAGIRGIPPKIEALPSDISIDEGKVLTVACAFTGEPTPEVTWSCGGRKIHSQEQGRF HIENTDDLTTLI IMDVQKQDGGLYTLSLGNEFGSDSATVNIHIRSI
Biomarker: obscurin
mRNA Sequence
GenBank: AJ002535.1
SEQ ID NO: 17
TGCCTACCAGCAGCCCACACTCCGGCCGCTGCCCAGAGCCCCCATAGAGAGAGGTCCCCGCCGCCACCGT CATGGATCAGCCACAGTTCAGCGGGGCGCCCCGCTTTCTCACCCGGCCCAAGGCCTTCGTGGTGTCGGTG GGCAAGGACGCCACCCTCAGCTGCCAGATCGTGGGTAATCCCACGCCACAGGTGAGCTGGGAGAAGGACC AGCAGCCGGTGACGGCCGGCGCGCGCTTCCGTCTGGCCCAGGACGGCGACCTCTACCGCCTCACTATCCT GGACCTGGCGCTGGGCGACAGTGGGCAATACGTGTGCCGCGCGCGCAATGCCATAGGCGAGGCCTTCGCT GCCGTGGGCCTGCAGGTGGACGCGGAGGCCGCGTGCGCCGAGCAGGCGCCGCACTTCCTGCTGCGGCCCA CGTCCATCCGCGTGCGCGAGGGCTCAGAGGCCACCTTCCGCTGCCGCGTGGGTGGCTCCCCGAGGCCGGC AGTGAGCTGGTCCAAGGACGGGCGGCGCCTGGGTGAGCCCGACGGCCCCCGCGTGCGCGTGGAGGAGCTC GGCGAGGCAAGTGCGCTGCGCATTCGGGCGGCGCGGCCGCGCGACGGCGGCACTTACGAGGTCCGCGCCG AGAACCCGCTGGGCGCTGCCAGCGCCGCCGCGGCGCTAGTGGTGGACTCGGACGCCGCGGACACGGCCAG CCGGCCCGGGACCTCCACGGCCGCGCTCCTGGCGCACCTGCAGCGGCGGCGCGAGGCTATGCGCGCCGAG GGCGCCCCCGCCTCACCGCCCAGCACCGGCACGCGCACCTGCACGGTGACTGAAGGCAAGCACGCGCGCC TCAGCTGCTACGTGACCGGCGAGCCCAAGCCCGAGACGGTGTGGAAGAAGGACGGCCAGCTGGTGACCGA GGGCCGGCGCCACGTGGTGTACGAGGACGCGCAGGAGAACTTCGTGCTCAAGATCCTCTTCTGCAAGCAG TCGGACCGCGGCCTCTACACCTGCACGGCGTCCAACCTCGTGGGCCAGACCTACAGCTCTGTGCTGGTCG TAGTGCGCGAGCCCGCGGTTCCCTTCAAAAAGCGGCTGCAAGATCTGGAGGTGCGGGAGAAGGAGTCGGC TACGTTCCTATGTGAGGTGCCCCAGCCGTCCACTGAGGCCGCGTGGTTCAAGGAGGAGACGCGGTTGTGG GCGAGCGCCAAGTACGGCATCGAGGAGGAGGGCACCGAGCGCCGCCTGACCGTGCGCAATGTCTCGGCCG ACGACGACGCGGTGTACATCTGCGAGACGCCAGAGGGCAGCCGCACGGTGGCGGAGCTCGCAGTCCAAGG AAACCTCCTCCGAAAGCTCCCTCGGAAGACGGCGGTGCGCGTGGGCGACACGGCTATGTTTTGCGTGGAG CTGGCGGTCCCGGTGGGCCCCGTCCACTGGCTGCGGAACCAGGAGGAAGTGGTGGCGGGGGGCCGCGTGG CCATCTCCGCGGAGGGCACGCGCCACACACTGACCATCTCCCAGTGCTGCCTGGAGGATGTGGGCCAGGT GGCCTTTATGGCTGGCGACTGCCAGACGTCCACCCGGTTCTGCGTGTCGGCCCCCAGGAAGCCTCCCCTG CAACCCCCTGTGGATCCTGTGGTAAAGGCCAGGATGGAGAGTTCCGTGATTCTCAGCTGGTCCCCACCAC CCCATGGGGAACGCCCTGTCACTATCGACGGCTACCTGGTAGAGAAGAAGAAGCTTGGCACCTACACCTG GATCAGGTGCCACGAGGCTGAATGGGTGGCTACACCTGAGCTGACCGTGGCTGATGTGGCGGAGGAGGGG AACTTCCAGTTCCGAGTGTCCGCTCTCAACAGCTTTGGTCAGAGTCCCTACCTCGAGTTCCCGGGGACTG TCCACCTGGCCCCCAAGCTGGCCGTGAGGACACCGCTGAAGGCGGTGCAGGCGGTAGAGGGTGGCGAGGT CACTTTCTCCGTGGACCTCACGGTGGCCTCAGCGGGTGAGTGGTTCCTGGATGGGCAGGCCCTGAAGGCC AGCAGTGTGTATGAGATCCACTGTGATCGCACCCGGCACACGCTCACCATCCGGGAGGTGCCCGCCAGCC TGCACGGGGCGCAGCTGAAGTTCGTGGCCAACGGCATTGAGAGCAGCATCCGGATGGAGGTCCGGGCGGC CCCAGGGCTGACTGCCAACAAGCCGCCAGCCGCAGCTGCCCGGGAGGTGCTGGCTCGGCTGCACGAGGAG GCGCAGCTGCTGGCTGAGCTGTCAGATCAGGCTGCGGCTGTGACGTGGCTGAAGGATGGTCGCACACTGT CCCCAGGCCCCAAGTATGAGGTGCAGGCATCGGCCGGGCGGCGGGTGCTCCTTGTGCGAGATGTGGCCCG GGACGATGCAGGCCTCTACGAGTGCGTCAGCCGCGGGGGCCGCATCGCCTACCAGCTCTCCGTGCAAGGC CTCGCGCGCTTTCTGCACAAGGACATGGCGGGCAGCTGTGTGGATGCCGTGGCTGGGGGCCCGGCGCAGT TTGAGTGTGAGACCTCCGAAGCCCACGTCCACGTGCACTGGTACAAGGATGGCATGGAGCTGGGCCACTC CGGTGAGCGCTTCTTGCAGGAGGATGTGGGGACGCGGCACCGGCTGGTGGCAGCCACAGTCACCAGGCAG GATGAAGGCACCTACTCCTGCCGCGTGGGCGAGGACTCTGTGGACTTCCGGCTCCGCGTCTCTGAGCCCA AGGTGGTGTTTGCTAAGGAGCAGCTGGCACGCAGGAAGCTGCAGGCAGAGGCAGGAGCCAGTGCCACACT GAGCTGCGAGGTGGCCCAGGCCCAGACGGAGGTGACGTGGTACAAGGATGGGAAGAAGCTGAGCTCCAGC TCGAAAGTGTGCATGGAGGCCACAGGCTGCACGCGCAGGCTGGTTGTGCAGCAGGCAGGCCAGGCGGATG CCGGGGAGTATAGCTGCGAGGCTGGGGGCCAGCGGCTCTCCTTCCATCTGGATGTCAAAGAGCCCAAGGT GGTGTTTGCCAAGGACCAGGTGGCACACAGTGAGGTGCAGGCTGAGGCAGGGGCCAATGCCACGCTGAGC TGCGAGGTGGCCCAGGCCCAGGCGGAGGTGATGTGGTACAAAGATGGGAAGAAGCTGAGCTCCAGCTTGA AAGTGCATGTAGAGGCCAAAGGCTGCAGACGGAGGCTGGTGGTGCAGCAGGCAGGCAAGACGGATGCCGG GGACTACAGCTGCGAGGCCAGGGGCCAGAGGGTCTCCTTCCGCCTGCACATCACAGAGCCCAAGATGATG TTTGCAAAGGAGCAGTCAGTGCATAATGAGGTGCAGGCTGAGGCGGGGGCCAGTGCCATGCTGAGCTGTG AGGTGGCCCAGGCCCAGACGGAGGTGACGTGGTACAAGGATGGGAAGAAGCTGAGCTCCAGCTCAAAAGT GGGCATGGAGGTCAAAGGGTGCACACGGAGGCTGGTGCTGCCACAGGCGGGCAAAGCAGATGCTGGGGAG TACAGCTGTGAGGCTGGGGGCCAGAGAGTCTCCTTCCACCTGCACATCACAGAGCCCAAGGGGGTGTTTG CGAAGGAGCAGTCAGTGCATAATGAGGTGCAGGCTGAGGCGGGGACCACTGCCATGCTGAGCTGTGAGGT GGCCCAGCCCCAGACGGAGGTGACGTGGTACAAGGACGGGAAGAAGCTGAGCTCCAGCTCAAAAGTACGC ATGGAGGTCAAGGGCTGCACACGAAGGCTGGTAGTGCAGCAGGTGGGCAAAGCAGATGCTGGGGAGTACA GCTGCGAGGCTGGGGGCCAGAGAGTCTCCTTTCAACTGCACATCACAGAGCCCAAGGCAGTGTTTGCCAA GGAGCAGTTGGTGCATAATGAGGTGCGGACTGAGGCAGGGGCCAGTGCCACACTGAGCTGTGAGGTGGCC CAGGCCCAGACAGAGGTGACGTGGTACAAGGATGGGAAGAAGCTGAGCTCCAGTTCGAAAGTGCGCATAG AGGCTGCGGGCTGCATGCGGCAGCTGGTGGTGCAGCAGGCAGGCCAGGCAGATGCTGGGGAGTACACCTG TGAGGCTGGGGGCCAGCGGCTCTCCTTCCACCTGGATGTTTCAGAGCCCAAGGCGGTGTTTGCAAAGGAG CAGCTGGCACACAGGAAGGTGCAGGCCGAGGCGGGGGCCATTGCCACGCTGAGCTGCGAGGTGGCCCAGG CCCAGACAGAGGTGACGTGGTACAAGGACGGGAAGAAGCTGAGCTCCAGCTCGAAAGTTCGAATGGAGGC TGTGGGCTGCACACGGAGGCTGGTGGTGCAGCAGGCATGCCAGGCGGACACCGGGGAGTATAGCTGCGAG GCCGGGGGCCAGCGGCTCTCCTTCAGCCTGGACGTGGCAGAGCCCAAGGTGGTGTTTGCCAAGGAGCAGC CAGTGCACAGGGAGGTGCAGGCCCAGGCGGGGGCCAGCACCACACTCAGCTGCGAGGTGGCTCAGGCCCA GACGGAGGTGATGTGGTACAAGGACGGGAAGAAGCTGAGCTTCAGCTCGAAAGTGCGCATGGAGGCTGTG GGCTGCACACGGAGGCTGGTGGTGCAGCAGGCGGGCCAGGCGGACGCCGGGGAGTACAGCTGCGAGGCGG GGAGCCAGCGGCTCTCCTTCCACCTGCACGTGGCAGAGCCCAAGGCGGTGTTTGCCAAGGAGCAGCCAGC GAGCAGGGAGGTGCAGGCTGAGGCGGGGACCAGTGCCACGCTGAGCTGCGAGGTGGCCCAGGCCCAGACA GAGGTGACGTGGTACAAGGACGGGAAGAAACTGAGCTCCAGCTCGAAAGTGCGAATGGAGGCCGTGGGCT GCACACGGAGGCTGGTGGTGCAGGAGGCAGGCCAGGCGGACGCCGGGGAGTACAGCTGCAAGGCCGGGGA TCAGCGGCTGTCCTTCCACCTGCACGTGGCAGAGCCCAAGGTGGTGTTTGCCAAGGAGCAGCCAGCACAC AGGGAGGTGCAGGCTGAGGCGGGGGCCAGTGCCACGCTGAGCTGCGAGGTGGCCCAGGCCCAGACAGAGG TGACGTGGTACAAGGATGGGAAGAAGCTGAGTTCCAGCTCGAAAGTGCGCGTGGAGGCCGTGGGCTGCAC ACGGAGGCTGGTGGTGCAGCAGGCGGGCCAGGCGGATGCTGGGGAGTACAGCTGTGAGGCGGGGGGCCAA CGGCTGTCCTTCCGCCTGCACGTGGCAGAGCTGGAGCCCCAAATTTCAGAGAGACCCTGCCGCAGGGAGC CTCTGGTGGTCAAGGAGCATGAAGACATCATCCTGACCGCCACACTGGCCACACCCTCTGCGGCCACGGT GACCTGGCTCAAGGATGGTGTGGAGATTCGCCGCAGCAAGCGGCATGAGACAGCCAGCCAGGGGGACACC CACACCCTGACCGTGCATGGCGCCCAGGTTCTGGACAGCGCCATCTACAGCTGCCGTGTGGGCGCAGAGG GGCAGGACTTCCCAGTGCAGGTGGAAGAGGTGGCCGCCAAGTTCTGCCGGCTGCTGGAGCCTGTGTGCGG CGAGCTGGGTGGCACGGTGACACTGGCCTGCGAGCTAAGCCCAGCGTGTGCAGAGGTGGTGTGGCGCTGC GGCAACACGCAGCCTCGGGTGGGCAAGCGCTTCCAGATGGTGGCCGAGGGGCCCGTGCGCTCACTCACTG TGTTGGGGCTGCGCGCAGAGGACGCAGGGGAGTACGTGTGTGAGAGCCGTGATGACCACACCAGTGCGCA GCTCACCGTCAGTGTGCCCCGAGTGGTGAAGTTTATGTCTGGGCTGAGCACCGTGGTCGCAGAGGAGGGC GGCGAGGCCACCTTCCAGTGCGTGGTGTCCCCCAGTGATGTGGCAGTCGTGTGGTTCCGGGACGGTGCCC TGCTTCAGCCCAGCGAGAAGTTTGCCATATCACAGAGTGGCGCCAGCCACAGCCTGACCATCTCAGACCT GGTGCTGGAGGACGCGGGCCAGATCACCGTGGAGGCTGAGGGCGCCTCATCCTCTGCTGCCCTGAGGGTC CGAGAGGCGCCTGTGCTGTTCAAAAAGAAGCTGGAGCCGCAGACGGTGGAGGAGCGGAGCTCGGTGACCC TGGAGGTGGAGCTGACGCGGCCGTGGCCGGAGCTGAGGTGGACACGGAACGCGACGGCCCTGGCGCCGGG AAAGAACGTGGAGATCCACGCCGAGGGCGCCCGCCACCGCCTGGTTCTGCACAACGTAGGTTTTGCCGAC CGTGGCTTCTTTGGCTGCGAGACGCCGGATGACAAGACACAGGCCAAACTCACCGTGGAGATGCGCCAGG TACGGCTCGTACGGGGCCTGCAGGCAGTGGAGGCACGGGAGCAGGGCACGGCTACCATGGAGGTGCAGCT GTCGCATGCGGACGTGGATGGCAGCTGGACTCGTGACGGTCTGCGGTTCCAGCAGGGGCCCACGTGCCAC CTGGCTGTGCGGGGCCCCATGCACACCCTCACACTCTCGGGGCTGCGGCCAGAGGATAGTGGCCTTATGG TCTTCAAGGCCGAAGGAGTGCACACGTCGGCGCGGCTCGTGGTCACCGAGCTTCCCGTGAGCTTCAGCCG CCCGCTGCAGGACGTGGTGACCACTGAGAAGGAGAAGGTTACCCTGGAGTGCGAGCTGTCGCGTCCTAAT GTGGATGTGCGCTGGCTGAAGGACGGTGTGGAGCTGCGGGCAGGCAAGACGATGGCCATCGCAGCCCAGG GCGCCTGCAGGAGCCTCACCATTTACCGGTGCGAGTTCGCGGATCAGGGAGTGTATGTGTGTGATGCCCA TGATGCCCAGAGCTCTGCCTCCGTGAAGGTACAAGGAAGGACATACACTCTCATCTACCGGAGAGTCCTG GCGGAAGATGCAGGAGAGATCCAATTTGTAGCCGAAAATGCAGAATCGCGAGCCCAGCTCCGAGTGAAGG AGCTGCCAGTGACCCTCGTGCGCCCGCTGCGGGACAAGATTGCCATGGAGAAGCACCGCGGTGTGCTGGA GTGTCAGGTGTCCCGGGCCAGCGCCCAGGTGCGGTGGTTCAAGGGCAGTCAGGAGCTGCAGCCCGGGCCC AAGTACGAGCTGGTCAGTGATGGCCTCTACCGCAAGCTGATCATCAGTGATGTCCACGCAGAGGACGAGG ACACCTACACCTGTGACGCCGGTGATGTCAAGACCAGTGCACAGTTCTTCGTGGAAGAGCAATCCATCAC CATTGTGCGGGGTCTGCAGGACGTGACAGTGATGGAGCCCGCTCCTGCCTGGTTTGAGTGTGAGACCTCC ATCCCCTCAGTGCGGCCACCTAAGTGGCTCCTGGGGAAGACGGTGTTGCAGGCTGGGGGGAACGTGGGCC TGGAGCAGGAGGGCACGGTGCACCGGCTGATGCTGCGGCGGACCTGCTCCACCATGACCGGGCCCGTGCA CTTCACCGTTGGCAAGTCGCGCTCCTCTGCCCGCCTGGTGGTCTCAGACATCCCCGTAGTCCTCACACGG CCGTTGGAGCCCAAGACAGGGCGTGAGCTGCAGTCAGTGGTCCTGTCCTGCGACTTCCGGCCAGCCCCCA AGGCTGTGCAGTGGTACAAGGATGACACGCCCCTGTCTCCCTCTGAGAAGTTTAAGATGAGCCTGGAGGG TCAGATGGCTGAGCTGCGCATCCTCCGGCTCATGCCTGCTGATGCTGGTGTCTACCGGTGCCAGGCGGGC AGTGCCCACAGCAGCACTGAGGTCACTGTGGAAGCGCGGGAGGTGACAGTGACAGGGCCGCTACAGGATG CAGAGGCCACGGAGGAGGGCTGGGCCAGCTTCTCCTGTGAGCTGTCCCACGAGGATGAGGAGGTCGAGTG GTCGCTCAACGGGATGCCCCTGTACAACGACAGCTTCCATGAGATCTCACACAAGGGCCGGCGCCACACG CTGGTACTGAAGAGCATCCAGCGGGCTGATGCGGGCATAGTACGCGCCTCCTCCCTGAAGGTGTCGACCT CTGCCCGCCTGGAGGTCCGAGTGAAGCCGGTGGTGTTCCTGAAGGCGCTGGATGACCTGTCCGCAGAGGA GCGCGGCACCCTGGCCCTGCAGTGTGAAGTCTCTGACCCCGAGGCCCATGTGGTGTGGCGCAAAGATGGC GTGCAGCTGGGCCCCAGTGACAAGTATGACTTCCTGCACACGGCGGGCACGCGGGGGCTCGTGGTGCATG ACGTGAGCCCTGAAGACGCCGGCCTGTACACCTGCCACGTGGGCTCCGAGGAGACCCGGGCCCGGGTCCG CGTGCACGATCTGCACGTGGGCATCACCAAGAGGCTGAAGACAATGGAGGTGCTGGAAGGGGAAAGCTGC AGCTTTGAGTGCGTCCTGTCCCACGAGAGTGCCAGCGACCCGGCCATGTGGACAGTCGGTGGGAAGACAG TGGGCAGCTCCAGCCGCTTCCAGGCCACACGTCAGGGCCGAAAATACATCCTGGTGGTCCGGGAGGCTGC ACCAAGTGATGCCGGGGAGGTGGTCTTCTCTGTGCGGGGCCTCACCTCCAAGGCCTCACTCATTGTCAGA GAGAGGCCGGCCGCCATCATCAAGCCCCTGGAAGACCAGTGGGTGGCGCCAGGGGAGGACGTGGAGCTGC GCTGTGAGCTGTCACGGGCGGGAACGCCCGTGCACTGGCTGAAGGACAGGAAGGCCATCCGCAAGAGCCA GAAGTATGATGTGGTCTGCGAGGGCACGATGGCCATGCTGGTCATCCGCGGGGCCTCGCTCAAGGACGCG GGCGAGTACACGTGTGAGGTGGAGGCTTCCAAGAGCACAGCCAGCCTCCATGTGGAAGAAAAAGCAAACT GCTTCACAGAGGAGCTGACCAATCTGCAGGTGGAGGAGAAAGGCACAGCTGTGTTCACGTGCAAGACGGA GCACCCCGCGGCCACAGTGACCTGGCGCAAGGGCCTCTTGGAGCTACGGGCCTCAGGGAAGCACCAGCCC AGCCAGGAGGGCCTGACCCTGCGGCTCACCATCAGTGCCCTGGAGAAGGCAGACAGCGACACCTATACCT GCGACATTGGCCAGGCCCAGTCCCGGGCCCAGCTCCTAGTGCAAGGCCGGAGAGTGCACATCATCGAGGA CCTGGAGGATGTGGATGTGCAGGAGGGCTCCTCGGCCACCTTCCGTTGCCGGATCTCCCCGGCCAACTAC GAGCCTGTGCACTGGTTCCTGGACAAGACACCCCTGCATGCCAACGAGCTCAATGAGATCGATGCCCAGC CCGGGGGCTACCACGTGCTGACCCTGCGGCAGCTGGCGCTCAAGGACTCGGGCACCATCTACTTTGAGGC GGGTGACCAGCGGGCCTCGGCCGCCCTGCGGGTCACTGAGAAGCCAAGCGTCTTCTCCCGGGAGCTCACA GATGCCACCATCACAGAGGGTGAGGACTTGACCCTGGTGTGCGAGACCAGCACCTGCGACATTCCTATGT GCTGGACCAAGGATGGGAAGACCCTGCGGGGGTCTGCCCGGTGCCAGCTGAGCCATGAGGGCCACCGGGC CCAGCTGCTCATCACTGGGGCCACCCTGCAGGACAGTGGACGCTACAAGTGTGAGGCTGGGGGCGCCTGC AGCAGCTCCATTGTCAGGGTGCATGCGCGGCCAGTGCGGTTCCAGGAGGCCCTGAAGGACCTGGAGGTGC TGGAGGGTGGTGCTGCCACACTGCGCTGTGTGCTGTCATCTGTGGCTGCGCCCGTGAAGTGGTGCTATGG AAACAACGTCCTGAGGCCAGGTGACAAATACAGCCTACGCCAGGAGGGTGCCATGCTGGAGCTGGTGGTC CGGAACCTCCGGCCGCAGGACAGCGGGCGGTACTCATGCTCCTTCGGGGACCAGACTACTTCTGCCACCC TCACAGTGACTGCCCTGCCTGCCCAGTTCATCGGGAAACTGAGAAACAAGGAGGCCACAGAAGGGGCCAC GGCCACGCTGCGGTGTGAGCTGAGCAAGACAGCCCCTGTGGAGTGGAGAAAGGGGTCCGAGACCCTCAGA GATGGGGACAGATACTGTCTGAGGCAGGACGGGGCCATGTGTGAGCTGCAGATCCGTGGCCTGGCCATGG TGGATGCCGCGGAGTACTCGTGTGTGTGTGGAGAGGAGAGGACCTCAGCCTCACTCACCATCAGGCCCAT GCCTGCCCACTTCATAGGAAGACTGAGACACCAAGAGAGCATAGAAGGGGCCACAGCCACGCTGCGGTGT GAGCTGAGCAAGGCGGCCCCCGTGGAGTGGAGGAAGGGGCGTGAGAGCCTCAGAGATGGGGACAGACATA GCCTGAGGCAGGACGGGGCTGTGTGCGAGCTGCAGATCTGTGGCCTGGCTGTGGCAGATGCTGGGGAGTA CTCCTGTGTGTGTGGGGAGGAGAGGACCTCTGCCACTCTCACCGTGAAGGCCCTGCCAGCCAAGTTCACA GAGGGTCTGAGGAATGAAGAGGCCGTGGAAGGGGCCACAGCCATGTTGTGGTGTGAACTGAGCAAGGTGG CCCCTGTGGAGTGGAGGAAGGGGCCCGAGAACCTCAGAGATGGGGACAGATACATCCTGAGGCAGGAGGG GACCAGGTGTGAGCTGCAGATCTGTGGCCTGGCCATGGCGGACGCCGGGGAGTACTTGTGTGTGTGCGGG CAGGAGAGGACCTCAGCCACGCTCACCATCAGGGCTCTGCCTGCCAGGTTCATAGAAGATGTGAAAAACC AGGAGGCCAGAGAAGGGGCCACGGCTGTGCTGCAGTGTGAGCTGAACAGTGCAGCCCCTGTGGAGTGGAG AAAGGGGTCTGAGACCCTCAGAGATGGGGACAGATACAGCCTGAGGCAGGACGGGACTAAATGTGAGCTG CAGATTCGTGGCCTGGCCATGGCAGACACTGGGGAGTACTCGTGCGTGTGCGGGCAGGAGAGGACCTCGG CTATGCTCACCGTCAGGGCTCTACCCATCAAGTTCACAGAGGGTCTGAGGAACGAAGAGGCCACAGAAGG GGCAACAGCCGTGCTGCGGTGTGAGCTGAGCAAGATGGCCCCCGTGGAGTGGTGGAAGGGGCATGAGACC CTCAGAGATGGAGACAGACACAGCCTGAGGCAGGACGGGGCCAGGTGTGAGCTGCAGATCCGCGGCCTCG TGGCAGAGGACGCTGGGGAGTACCTGTGCATGTGCGGGAAGGAGAGGACCTCAGCCATGCTCACCGTCAG GGCCATGCCTTCCAAGTTCATAGAGGGTCTGAGGAATGAAGAGGCCACAGAAGGGGACACGGCCACGCTG TGGTGTGAGCTGAGCAAGGCGGCACCGGTGGAGTGGAGGAAGGGGCATGAGACCCTCAGAGATGGGGACA GACACAGCCTGAGGCAGGACGGGTCCAGGTGTGAGCTGCAGATCCGTGGCCTGGCTGTGGTGGATGCCGG GGAGTACTCGTGTGTGTGCGGGCAGGAGAGGACCTCAGCCACACTCACTGTCAGGGCCCTGCCTGCCAGA TTCATAGAAGATGTGAAAAACCAGGAGGCCAGAGAAGGGGCCACGGCCGTGCTGCAATGTGAGCTGAGCA AGGCGGCCCCCGTGGAGTGGAGGAAGGGGTCTGAGACCCTCAGAGGTGGGGACAGATACAGCCTGAGGCA GGATGGGACCAGATGTGAGCTGCAGATTCATGGCCTGTCTGTGGCAGACACTGGGGAGTACTCGTGTGTG TGCGGGCAGGAGAGGACCTCGGCCACACTCACCGTCAGGGCCCCACAGCCAGTGTTCCGGGAGCCGCTGC AGAGTCTGCAGGCGGAGGAGGGCTCCACGGCCACCCTGCAGTGTGAGCTGTCTGAGCCCACTGCTACAGT GGTCTGGAGCAAGGGTGGCCTGCAGCTGCAGGCCAATGGGCGCCGGGAGCCACGGCTTCAGGGCTGCACC GCGGAGCTGGTGTTACAGGACCTACAACGTGAAGACACTGGCGAATACACTTGCACCTGTGGCTCCCAGG CCACCAGTGCCACCCTCACTGTCACAGCTGCGCCTGTGCGGTTCCTCCGAGAGCTGCAGCACCAGGAGGT GGATGAGGGAGGCACCGCACACTTATGCTGCGAGCTGAGCCGGGCGGGTGCGAGCGTGGAGTGGCGCAAG GGCTCCCTACAGCTCTTCCCTTGTGCCAAGTACCAGATGGTGCAGGATGGTGCAGCTGCAGAGCTGCTGG TACGCGGAGTGGAGCAGGAGGATGCGGGTGACTACACGTGTGACACGGGCCACACGCAGAGCATGGCCAG CCTCTCTGTCCGTGTCCCCAGGCCCAAGTTCAAGACCCGGCTTCAGAGTCTGGAGCAGGAGACAGGTGAC ATAGCCCGGCTGTGCTGTCAGCTGAGTGATGCAGAGTCGGGGGCCGTGGTGCAATGGCTCAAGGAGGGCG TGGAGCTGCATGCGGGCCCCAAGTACGAGATGCGGAGCCAGGGGGCCACGCGGGAGCTGCTGATCCACCA ACTGGAGGCCAAGGACACGGGCGAGTATGCCTGTGTGACAGGCGGCCAGAAAACCGCTGCCTCCCTCAGG GTCACAGAGCCTGAGGTGACCATTGTACGGGGGCTGGTTGATGCGGAGGTGACGGCCGATGAGGATGTTG AGTTCAGCTGTGAGGTGTCCAGGGCTGGAGCCACAGGCGTGCAGTGGTGCCTACAGGGCCTGCCACTGCA AAGCAATGAGGTGACAGAGGTGGCTGTGCGGGATGGCCGCATCCACACCCTGCGGCTGAAGGGCGTGACG CCCGAGGACGCTGGCACTGTCTCCTTCCATTTGGGAAACCATGCTTCCTCTGCCCAGCTCACCGTCAGAG CTCCTGAGGTGACCATCCTGGAGCCCCTGCAGGACGTGCAGCTCAGTGAGGGCCAGGATGCCAGCTTCCA GTGCCGGCTATCCAGAGCTTCAGGCCAGGAGGCCCGCTGGGCTTTAGGAGGGGTGCCCCTGCAGGCCAAC GAGATGAATGACATCACTGTGGAGCAGGGCACACTCCACCTGCTCACCCTGCACAAGGTGACCCTTGAGG ATGCTGGAACTGTCAGTTTCCACGTGGGCACGTGTAGCTCTGAGGCCCAGCTGAAAGTCACAGCCAAGAA CACGGTGGTGCGGGGGCTGGAGAATGTGGAGGCGCTGGAGGGCGGCGAGGCGCTGTTCGAGTGCCAGCTG TCCCAGCCCGAGGTGGCCGCCCACACCTGGCTGCTGGACGACGAACCCGTGCGCACCTCGGAGAACGCCG AGGTGGTCTTCTTCGAGAACGGCCTGCGCCACCTGCTGCTGCTCAAAAACTTGCGGCCACAAGACAGCTG CCGGGTGACCTTCCTGGCTGGGGATATGGTGACGTCCGCATTCCTCACGGTCCGAGGCTGGCGCCTGGAG ATCCTGGAGCCTCTGAAAAACGCGGCGGTCCGGGCCGGCGCACAGGCACGCTTCACCTGCACGCTCAGCG AGGCGGTGCCCGTGGGAGAGGCGTCCTGGTACATCAATGGCGCGGCAGTGCAGCCGGATGACAGCGACTG GACTGTCACCGCCGACGGCAGTCACCAAGCCCTACTGCTGCGCAGCGCCCAGCCCCACCACGCCGGGGAG GTCACCTTCGCTTGCCGCGACGCCGTGGCCTCTGCACGGCTCACCGTGCTGGGCCTCCCTGATCCCCCAG AGGATGCTGAGGTGGTGGCTCACAGCAGCCACACTGTGACACTGTCTTGGGCAGCTCCCATGAGTGATGG AGGCGGTGGTCTCTGTGGCTACCGCGTGGAGGTGAAGGAGGGGGCCACAGGCCAGTGGCGGCTGTGCCAC GAGCTGGTGCCTGGACCCGAGTGTGTGGTGGATGGCCTGGCCCCCGGGGAGACCTACCGCTTCCGTGTGG CAGCTGTGGGCCCTGTGGGTGCTGGGGAACCGGTTCACCTGCCCCAGACAGTGCGGCTTGCAGAGCCACC GAAGCCTGTGCCTCCCCAGCCCTCAGCCCCTGAGAGCCGGCAGGTGGCAGCTGGTGAAGATGTCTCTCTG GAGCTTGAGGTGGTGGCTGAGGCTGGTGAGGTCATCTGGCACAAGGGAATGGAGCGCATCCAGCCCGGTG GGCGGTTCGAGGTGGTCTCCCAGGGTCGGCAACAGATGCTGGTGATCAAGGGCTTCACGGCAGAAGACCA GGGCGAGTACCACTGTGGCCTGGCTCAGGGCTCCATCTGCCCTGCGGCTGCCACCTTCCAGGTGGCACTG AGCCCAGCCTCTGTGGATGAGGCCCCTCAGCCCAGCTTGCCCCCCGAGGCAGCCCAGGAGGGTGACCTGC ACCTACTGTGGGAGGCCCTGGCTCGGAAACGTCGCATGAGCCGTGAGCCCACGCTGGACTCCATTAGCGA GCTGCCAGAGGAGGACGGCCGCTCGCAGCGCCTGCCACAGGAGGCAGAGGAGGTGGCACCTGATCTCTCT GAAGGCTACTCCACGGCCGATGAGCTGGCCCGCACTGGAGATGCTGACCTCTCACACACCAGCTCTGATG ATGAGTCCCGGGCAGGCACCCCTTCCCTGGTCACCTACCTCAAGAAGGCTGGGAGGCCAGGCACCTCACC ACTGGCCAGCAAGGTTGGGGCCCCAGCAGCCCCCTCTGTGAAGCCACAGCAGCAGCAGGAGCCACTGGCT GCTGTGCGCCCACCACTGGGAGACCTGAGCACCAAAGACCTGGGTGATCCCTCAATGGACAAGGCAGCTG TGAAGATCCAGGCTGCCTTTAAGGGCTACAAGGTCCGGAAGGAGATGAAGCAGCAGGAAGGGCCCATGTT CTCCCACACATTTGGGGACACCGAGGCACAGGTGGGGGATGCCCTGCGGCTGGAGTGTGTCGTGGCCAGC AAGGCAGATGTGCGAGCCCGCTGGCTGAAGGATGGTGTGGAGCTGACCGATGGGCGGCACCATCACATCG ACCAGCTTGGGGATGGCACCTGCTCTCTGCTGATCGCTGGCCTGGACCGTGCTGATGCTGGCTGCTACAC CTGTCAGGTGAGCAACAAGTTTGGCCAGGTGACCCACAGTGCCTGTGTGGTGGTCAGTGGGTCAGAGAGT GAAGCCGAGAGCTCCTCTGGGGGTGAGCTGGACGATGCCTTCCGCCGGGCTGCCCGTCGGCTGCACCGGC TCTTCCGCACCAAAAGTCCGGCTGAAGTTTCAGATGAGGAGCTCTTCCTGAGTGCAGACGAGGGCCCTGC AGAGCCAGAGGAGCCCGCGGACTGGCAGACATACCGCGAAGATGAGCATTTCATCTGCATCCGTTTTGAG GCGCTCACTGAGGCCCGCCAGGCGGTAACTCGCTTCCAGGAGATGTTTGCCACACTGGGCATTGGGGTGG AGATCAAGCTGGTGGAACAGGGGCCTCGGAGGGTAGAGATGTGCATCAGCAAAGAGACTCCTGCCCCTGT GGTGCCTCCAGAGCCATTGCCCAGCCTACTGACTTCTGACGCTGCCCCAGTGTTCCTGACTGAGTTGCAG AACCAAGAAGTGCAGGATGGGTATCCTGTGAGCTTTGACTGCGTGGTGACAGGTCAGCCCATGCCCAGTG TGCGCTGGTTCAAGGATGGGAAGTTGTTGGAGGAGGATGATCACTACATGATTAATGAAGACCAACAGGG TGGCCATCAGCTCATCATCACAGCCGTGGTGCCAGCAGACATGGGCGTCTACCGCTGCCTGGCCGAGAAC AGCATGGGTGTCTCCTCCACCAAGGCTGAGCTCCGTGTGGACTTGACAAGCACAGACTATGACACTGCAG CAGATGCCACGGAGTCCTCATCCTACTTCAGTGCCCAAGGCTACCTGTCCAGCCGGGAGCAGGAGGGAAC AGAGTCCACCACTGATGAGGGCCAGCTGCCCCAGGTGGTGGAGGAGCTGAGAGACCTCCAGGTGGCCCCT GGCACACGCCTGGCCAAGTTCCAGCTCAAGGTGAAAGGCTACCCTGCTCCCAGATTATACTGGTTCAAAG ATGGCCAGCCCCTGACCGCATCTGCCCACATCCGCATGACTGACAAGAAGATCCTGCACACCCTGGAGAT CATCTCCGTCACCCGGGAGGACTCTGGCCAGTATGCAGCCTATATCAGCAATGCCATGGGTGCTGCCTAC TCGTCTGCCCGGCTGCTGGTTCGAGGCCCTGATGAGCCAGAAGAGAAGCCTGCATCAGATGTGCATGAGC AGCTGGTGCCGCCCCGAATGCTGGAGAGGTTCACCCCCAAGAAAGTGAAGAAAGGCTCCAGCATCACCTT CTCTGTGAAGGTAGAAGGACGCCCGGTGCCCACCGTGCACTGGCTCAGGGAGGAGGCTGAGAGAGGCGTG CTGTGGATTGGCCCTGACACACCGGGCTACACCGTGGCCAGCTCTGCGCAGCAGCACAGCCTGGTCCTGC TGGACGTGGGCCGGCAGCACCAGGGCACCTACACATGCATTGCCAGCAACGCTGCCGGCCAGGCCCTCTG CTCCGCCAGCCTGCACGTCTCGGGCCTGCCTAAGGTGGAGGAGCAGGAGAAAGTGAAGGAAGCGCTGATT TCCACTTTCCTGCAGGGGACCACACAAGCCATCTCAGCACAGGGGTTCCAAACTGCGAGTTTTGCTGACC TTGGTGGGCAGAGGAAAGAAGAGCCTCTGGCTGCCAAGGAGGCCCTCGGCCACCTGTCCCTCGCTGAGGT GGGCACAGAGGAGTTCCTGCAGAAACTGACCTCCCAGATCACTGAGATGGTATCGGCCAAGATCACGCAG GCCAAGCTGCAGGTGCCCGGAGGTGACAGTGATGAGGACTCCAAGACACCATCTGCATCCCCCCGCCATG GCCGATCACGGCCATCCTCCAGCATCCAGGAGTCTTCCTCAGAGTCAGAGGACGGCGATGCCCGAGGCGA GATCTTTGACATCTACGTGGTCACCGCTGACTACCTGCCCCTAGGGGCTGAGCAGGATGCCATCACGCTG CGGGAAGGCCAGTATGTGGAGGTCCTGGATGCAGCCCACCCACTGCGCTGGCTTGTCCGCACCAAGCCCA CCAAGTCCAGCCCCTCACGGCAGGGCTGGGTGTCACCAGCCTACCTGGACAGGAGGCTCAAGCTGTCACC TGAGTGGGGGGCCGCTGAGGCCCCTGAGTTCCCTGGGGAGGCTGTGTCTGAAGACGAATACAAGGCAAGG CTGAGCTCTGTGATCCAGGAGCTGCTGAGTTCTGAGCAGGCCTTCGTGGAGGAGCTGCAGTTCCTGCAGA GCCACCACCTGCAGCACCTGGAGCGCTGCCCCCACGTGCCCATAGCTGTGGCCGGCCAGAAGGCAGTCAT CTTCCGCAATGTGCGGGACATCGGCCGCTTCCACAGCAGCTTCCTGCAGGAGTTGCAGCAGTGCGACACG GACGACGACGTGGCCATGTGCTTCATCAAGAACCAGGCGGCCTTTGAGCAGTACCTGGAGTTCCTGGTGG GGCGTGTGCAGGCTGAGTCGGTGGTCGTCAGCACGGCCATCCAGGAGTTCTACAAGAAATACGCGGAGGA GGCCCTGTTGGCAGGGGACCCCTCTCAGCCCCCGCCACCACCTCTGCAGCACTACCTGGAGCAGCCAGTG GAGCGGGTGCAGCGCTACCAGGCCTTGCTGAAGGAGCTGATCCGCAACAAGGCGCGGAACAGACAGAACT GCGCGCTGCTGGAGCAGGCCTATGCCGTGGTGTCTGCCCTGCCACAGCGCGCTGAGAACAAGCTGCACGT GTCCCTCATGGAGAACTACCCAGGCACCCTGCAGGCCCTGGGCGAGCCCATCCGCCAGGGCCACTTCATC GTGTGGGAGGGTGCACCGGGGGCCCGCATGCCCTGGAAGGGCCACAACCGTCACGTGTTCCTCTTCCGCA ACCACCTGGTAATCTGCAAGCCCCGGCGAGACTCCCGCACCGATACCGTCAGCTACGTGTTCCGGAACAT GATGAAGCTGAGCAGCATCGACCTGAACGACCAGGTGGAGGGGGATGACCGCGCCTTCGAGGTGTGGCAG GAGCGGGAGGACTCGGTGCGCAAGTACCTGCTGCAGGCACGGACAGCCATTATCAAGAGCTCGTGGGTGA AGGAGATCTGTGGCATCCAGCAGCGTCTGGCCCTGCCTGTGTGGCGGCCCCCGGACTTTGAAGAGGAGCT GGCCGACTGCACAGCCGAGCTGGGTGAGACAGTCAAGCTGGCCTGCCGCGTGACGGGCACACCCAAGCCT GTCATCAGCTGGTACAAAGATGGGAAAGCAGTGCAGGTGGACCCCCACCACATCCTCATTGAAGACCCTG ATGGCTCGTGTGCACTCATCCTGGACAGCCTGACCGGTGTGGACTCTGGCCAGTACATGTGCTTCGCGGC CAGCGCCGCTGGCAACTGCAGTACCCTGGGCAAGATCCTGGTGCAAGTCCCACCACGGTTCGTGAACAAG GTCCTGGCCTCACCCTTTGTGGAGGGAGAGGACGCCCAGTTCACCTGCACCATCGAAGGCGCCCCGTACC CGCAGATCAGGTGGTACAAGGACGGGGCCCTGCTGACCACTGGCAACAAGTTCCAGACACTGAGTGAGCC TCGCAGCGGCCTGCTAGTGCTGGTGATCCGGGCGGCCAGCAAGGAGGACCTGGGGCTCTACGAGTGTGAG CTGGTGAACCGGCTGGGCTCCGCGCGGGCTAGTGCGGAGCTGCGCATTCAGAGCCCCATGCTGCAGGCCC AGGAGCAGTGTCACAGGGAGCAGCTCGTGGCTGCAGTGGAAGTCACTGAGCAAGAGACTAAAGTCCCCAA GAAAACCGTCATCATAGAAGAGACCATCACCACTGTGGTGAAGAGCCCACGTGGCCAACGACGGTCCCCC AGCAAGTCCCCCTCCCGCTCACCTTCCCGYTGCTCTGCCAGCCCGCTGAGGCCAGGCCTACTGGCCCCCG ACCTGCTGTACCTGCCAGGTGCTGGCCAGCCCCGCAGGCCGGAGGCAGAACCAGGCCAGAAGCCCGTGGT GCCCACACTGTATGTGACGGAGGCCGAGGCCCACTCTCCAGCTCTGCCCGGACTCTCGGGGCCCCAGCCC AAGTGGGTGGAGGTGGAGGAGACCATTGAAGTCCGGGTGAAGAAGATGGGCCCGCAGGGTGTGTCTCCCA CCACAGAGGTGCCCAGGAGCTCATCGGGGCATCTCTTCACACTGCCCGGTGCGACCCCCGGAGGGGACCC CAATTCCAACAACTCCAACAACAAGCTGCTGGCCCAGGAGGCCTGGGCCCAGGGCACAGCCATGGTCGGC GTCAGAGAGCCCCTTGTCTTCCGCGTGGATGCCAGAGGCAGTGTGGACTGGGCTGCTTCTGGCATGGGCA GCCTGGAGGAGGAGGGCACCATGGAGGAGGCGGGAGAGGAAGAGGGGGAAGACGGAGATGCCTTTGTGAC GGAGGAGTCCCAGGACACACACAGCCTTGGGGATCGTGACCCCAAGATCCTCACGCACAACGGCCGCATG CTGACACTGGCTGACCTGGAAGATTACGTGCCTGGGGAAGGGGAGACCTTCCACTGTGGTGGCCCTGGGC CTGGCGCCCCTGATGACCCTCCCTGCGAGGTCTCGGTGATCCAGAGAGAGATCGGGGAGCCCACGGTGGG GCAGCCTGTGCTGCTCAGCGTGGGGCATGCACTGGGTCCCCGAGGCCCTCTCGGCCTCTTTAGGCCTGAG CCCCGTGGGGCGTCACCACCGGGACCCCAGGTCCGTAGCCTTGAGGGCACCTCCTTCCTCTTGCGGGAGG CCCCGGCTCGGCCTGTGGGCAGTGCTCCCTGGACGCAGTCTTTCTGCACCCGCATCCGGCGTTCTGCGGA CAGTGGCCAGAGCAGCTTCACCACAGAGCTTTCCACCCAGACCGTCAACTTCGGGACAGTGGGGGAGACG GTCACCCTTCACATCTGCCCAGACAGGGATGGGGATGAGGCGGCACAGCCCTGATGCTGCTGCCATGGTG GCTTGGGGCAGCGGGGAGAAAGGAGTGTCCTTGAGGCCTAGGACGCTGCCCGGCCTCAGCAGCAGCCCTG GGAGCCTCCTGAGGGCCCTCCCTGTCCCTGGCCACGGGCCCTTCTTACCTCACTCAACTTCAGCCAGGAG GACTGGGTGGTGCTTGCAATGTTGGAATGACCGGCTCAAAGACCTCAGCTCTGGGCTGTTTCCTGTCAGC CTGGCAGGAGCCTCAGGACTGTGGACGAAGGATGTGGCCTTGGGCATTTGTCCTGTTCCCACATGGGCCT GGTCCCTCCCTCCTGGCCCCAGCCACAGCTGCCAGGCCTGACATGGCCTTGCCTCTCCTGCAGTCTTGGT GACTGAGACCCTTGGGTGGCGCTTCCCAGCTCTGCAGGCCCTCCTGGCCTTTTCTGCAGGGTGGACACAG GGTCTGTGTGTGGGCAGCAGCCCCTGTCTCTCAGCAAGAATAAAGCAGCTTCCTGTGCAAAAAGG
Amino Acid Sequence
GenBank: CAC44768.1
SEQ ID NO: 18
MDQPQFSGAPRFLTRPKAFWSVGKDATLSCQIVGNPTPQVSWEKDQQPVTAGARFRLAQDGDLYRLTIL DLALGDSGQYVCRARNAIGEAFAAVGLQVDAEAACAEQAPHFLLRPTSIRVREGSEATFRCRVGGSPRPA VSWSKDGRRLGEPDGPRVRVEELGEASALRIRAARPRDGGTYEVRAENPLGAASAAAALWDSDAADTAS RPGTSTAALLAHLQRRREAMRAEGAPASPPSTGTRTCTVTEGKHARLSCYVTGEPKPETVWKKDGQLVTE GRRHWYEDAQENFVLKILFCKQSDRGLYTCTASNLVGQTYSSVLVWREPAVPFKKRLQDLEVREKESA TFLCEVPQPSTEAAWFKEETRLWASAKYGIEEEGTERRLTVRNVSADDDAVYICETPEGSRTVAELAVQG NLLRKLPRKTAVRVGDTAMFCVELAVPVGPVHWLRNQEEWAGGRVAISAEGTRHTL ISQCCLEDVGQV AFMAGDCQTSTRFCVSAPRKPPLQPPVDPWKARMESSVILSWSPPPHGERPVTIDGYLVEKKKLGTYTW IRCHEAEWVATPELTVADVAEEGNFQFRVSALNSFGQSPYLEFPGTVHLAPKLAVRTPLKAVQAVEGGEV TFSVDLTVASAGEWFLDGQALKASSVYEIHCDRTRHTLTIREVPASLHGAQLKFVANGIESSIRMEVRAA PGLTANKPPAAAAREVLARLHEEAQLLAELSDQAAAVTWLKDGRTLSPGPKYEVQASAGRRVLLVRDVAR DDAGLYECVSRGGRIAYQLSVQGLARFLHKDMAGSCVDAVAGGPAQFECETSEAHVHVHWYKDGMELGHS GERFLQEDVGTRHRLVAATVTRQDEGTYSCRVGEDSVDFRLRVSEPKWFAKEQLARRKLQAEAGASATL SCEVAQAQTEVTWYKDGKKLSSSSKVCMEATGCTRRLWQQAGQADAGEYSCEAGGQRLSFHLDVKEPKV VFAKDQVAH S E VQAE AGANAT L S CE VAQAQAE VMWYKDGKKL S S S LKVHVE AKGCRRRLWQQAGKT DAG DYSCEARGQRVSFRLHITEPKMMFAKEQSVHNEVQAEAGASAMLSCEVAQAQTEVTWYKDGKKLSSSSKV GMEVKGCTRRLVLPQAGKADAGEYSCEAGGQRVSFHLHITEPKGVFAKEQSVHNEVQAEAGTTAMLSCEV AQPQTEVTWYKDGKKLSSSSKVRMEVKGCTRRLWQQVGKADAGEYSCEAGGQRVSFQLHITEPKAVFAK EQLVHNEVRTEAGASATLSCEVAQAQTEVTWYKDGKKLSSSSKVRIEAAGCMRQLWQQAGQADAGEYTC EAGGQRLSFHLDVSEPKAVFAKEQLAHRKVQAEAGAIATLSCEVAQAQTEVTWYKDGKKLSSSSKVRMEA VGCTRRLWQQACQADTGEYSCEAGGQRLSFSLDVAEPKWFAKEQPVHREVQAQAGASTTLSCEVAQAQ TEVMWYKDGKKLSFSSKVRMEAVGCTRRLWQQAGQADAGEYSCEAGSQRLSFHLHVAEPKAVFAKEQPA SREVQAEAGTSATLSCEVAQAQTEVTWYKDGKKLSSSSKVRMEAVGCTRRLWQEAGQADAGEYSCKAGD QRLSFHLHVAEPKWFAKEQPAHREVQAEAGASATLSCEVAQAQTEVTWYKDGKKLSSSSKVRVEAVGCT RRLWQQAGQADAGEYSCEAGGQRLSFRLHVAELEPQISERPCRREPLWKEHEDI ILTATLA PSAATV TWLKDGVEIRRSKRHETASQGDTHTLTVHGAQVLDSAIYSCRVGAEGQDFPVQVEEVAAKFCRLLEPVCG ELGGTVTLACELSPACAEWWRCGNTQPRVGKRFQMVAEGPVRSLTVLGLRAEDAGEYVCESRDDHTSAQ LTVSVPRWKFMSGLSTWAEEGGEATFQCWSPSDVAWWFRDGALLQPSEKFAISQSGASHSLTISDL VLEDAGQITVEAEGASSSAALRVREAPVLFKKKLEPQTVEERSSVTLEVELTRPWPELRWTRNATALAPG KNVEIHAEGARHRLVLHNVGFADRGFFGCETPDDKTQAKLTVEMRQVRLVRGLQAVEAREQGTATMEVQL SHADVDGSWTRDGLRFQQGPTCHLAVRGPMHTLTLSGLRPEDSGLMVFKAEGVHTSARLWTELPVSFSR PLQDWTTEKEKVTLECELSRPNVDVRWLKDGVELRAGKTMAIAAQGACRSLTIYRCEFADQGVYVCDAH DAQSSASVKVQGRTYTLIYRRVLAEDAGEIQFVAENAESRAQLRVKELPVTLVRPLRDKIAMEKHRGVLE CQVSRASAQVRWFKGSQELQPGPKYELVSDGLYRKLI ISDVHAEDEDTYTCDAGDVKTSAQFFVEEQSI IVRGLQDVTVME PAPAWFECE S I PSVRPPKWLLGKTVLQAGGNVGLEQEGTVHRLMLRRTCS MTGPVH FTVGKSRSSARLWSDIPWLTRPLEPKTGRELQSWLSCDFRPAPKAVQWYKDDTPLSPSEKFKMSLEG QMAELRILRLMPADAGVYRCQAGSAHSSTEVTVEAREVTVTGPLQDAEATEEGWASFSCELSHEDEEVEW SLNGMPLYNDSFHEISHKGRRHTLVLKSIQRADAGIVRASSLKVSTSARLEVRVKPWFLKALDDLSAEE RGTLALQCEVSDPEAHWWRKDGVQLGPSDKYDFLHTAGTRGLWHDVSPEDAGLYTCHVGSEETRARVR VHDLHVGITKRLKTMEVLEGESCSFECVLSHESASDPAMWTVGGKTVGSSSRFQATRQGRKYILWREAA PSDAGEWFSVRGL SKASLIVRERPAAI IKPLEDQWVAPGEDVELRCELSRAG PVHWLKDRKAIRKSQ KYDWCEGTMAMLVIRGASLKDAGEYTCEVEASKSTASLHVEEKANCFTEELTNLQVEEKGTAVFTCKTE HPAATVTWRKGLLELRASGKHQPSQEGLTLRL ISALEKADSDTYTCDIGQAQSRAQLLVQGRRVHI IED LEDVDVQEGSSATFRCRISPANYEPVHWFLDKTPLHANELNEIDAQPGGYHVLTLRQLALKDSGTIYFEA GDQRASAALRVTEKPSVFSRELTDATITEGEDLTLVCETSTCDIPMCWTKDGKTLRGSARCQLSHEGHRA QLLITGATLQDSGRYKCEAGGACSSSIVRVHARPVRFQEALKDLEVLEGGAATLRCVLSSVAAPVKWCYG NNVLRPGDKYSLRQEGAMLELWRNLRPQDSGRYSCSFGDQTTSATLTVTALPAQFIGKLRNKEATEGAT ATLRCELSKTAPVEWRKGSETLRDGDRYCLRQDGAMCELQIRGLAMVDAAEYSCVCGEERTSASLTIRPM PAHFIGRLRHQESIEGATATLRCELSKAAPVEWRKGRESLRDGDRHSLRQDGAVCELQICGLAVADAGEY SCVCGEERTSATLTVKALPAKFTEGLRNEEAVEGATAMLWCELSKVAPVEWRKGPENLRDGDRYILRQEG TRCELQICGLAMADAGEYLCVCGQERTSATLTIRALPARFIEDVKNQEAREGATAVLQCELNSAAPVEWR KGSETLRDGDRYSLRQDGTKCELQIRGLAMADTGEYSCVCGQERTSAMLTVRALPIKFTEGLRNEEATEG ATAVLRCELSKMAPVEWWKGHETLRDGDRHSLRQDGARCELQIRGLVAEDAGEYLCMCGKERTSAMLTVR AMPSKFIEGLRNEEATEGDTATLWCELSKAAPVEWRKGHETLRDGDRHSLRQDGSRCELQIRGLAWDAG EYSCVCGQERTSATLTVRALPARFIEDVKNQEAREGATAVLQCELSKAAPVEWRKGSETLRGGDRYSLRQ DGTRCELQIHGLSVADTGEYSCVCGQERTSATLTVRAPQPVFREPLQSLQAEEGSTATLQCELSEPTATV VWSKGGLQLQANGRREPRLQGCTAELVLQDLQREDTGEYTCTCGSQATSATLTVTAAPVRFLRELQHQEV DEGGTAHLCCELSRAGASVEWRKGSLQLFPCAKYQMVQDGAAAELLVRGVEQEDAGDYTCDTGHTQSMAS LSVRVPRPKFKTRLQSLEQETGDIARLCCQLSDAESGAWQWLKEGVELHAGPKYEMRSQGATRELLIHQ LEAKDTGEYACVTGGQKTAASLRVTEPEVTIVRGLVDAEVTADEDVEFSCEVSRAGATGVQWCLQGLPLQ SNEVTEVAVRDGRIHTLRLKGVTPEDAGTVSFHLGNHASSAQLTVRAPEVTILEPLQDVQLSEGQDASFQ CRLSRASGQEARWALGGVPLQANEMNDITVEQGTLHLLTLHKVTLEDAGTVSFHVGTCSSEAQLKVTAKN TWRGLENVEALEGGEALFECQLSQPEVAAHTWLLDDEPVRTSENAEWFFENGLRHLLLLKNLRPQDSC RVTFLAGDMVTSAFLTVRGWRLEILEPLKNAAVRAGAQARFTCTLSEAVPVGEASWYINGAAVQPDDSDW TVTADGSHQALLLRSAQPHHAGEVTFACRDAVASARLTVLGLPDPPEDAEWAHSSHTVTLSWAAPMSDG GGGLCGYRVEVKEGATGQWRLCHELVPGPECWDGLAPGETYRFRVAAVGPVGAGEPVHLPQTVRLAEPP KPVPPQPSAPESRQVAAGEDVSLELEWAEAGEVIWHKGMERIQPGGRFEWSQGRQQMLVIKGFTAEDQ GEYHCGLAQGSICPAAATFQVALSPASVDEAPQPSLPPEAAQEGDLHLLWEALARKRRMSREPTLDSISE LPEEDGRSQRLPQEAEEVAPDLSEGYSTADELARTGDADLSHTSSDDESRAGTPSLVTYLKKAGRPGTSP LASKVGAPAAPSVKPQQQQEPLAAVRPPLGDLSTKDLGDPSMDKAAVKIQAAFKGYKVRKEMKQQEGPMF SHTFGDTEAQVGDALRLECWASKADVRARWLKDGVELTDGRHHHIDQLGDGTCSLLIAGLDRADAGCYT CQVSNKFGQVTHSACVWSGSESEAESSSGGELDDAFRRAARRLHRLFRTKSPAEVSDEELFLSADEGPA EPEEPADWQTYREDEHFICIRFEALTEARQAVTRFQEMFATLGIGVEIKLVEQGPRRVEMCISKETPAPV VPPEPLPSLLTSDAAPVFLTELQNQEVQDGYPVSFDCWTGQPMPSVRWFKDGKLLEEDDHYMINEDQQG GHQLI ITAWPADMGVYRCLAENSMGVSSTKAELRVDLTSTDYDTAADATESSSYFSAQGYLSSREQEGT ESTTDEGQLPQWEELRDLQVAPGTRLAKFQLKVKGYPAPRLYWFKDGQPLTASAHIRMTDKKILHTLEI ISVTREDSGQYAAYISNAMGAAYSSARLLVRGPDEPEEKPASDVHEQLVPPRMLERFTPKKVKKGSSITF SVKVEGRPVPTVHWLREEAERGVLWIGPDTPGYTVASSAQQHSLVLLDVGRQHQGTYTCIASNAAGQALC SASLHVSGLPKVEEQEKVKEALISTFLQGTTQAISAQGFQTASFADLGGQRKEEPLAAKEALGHLSLAEV GTEEFLQKLTSQITEMVSAKITQAKLQVPGGDSDEDSKTPSASPRHGRSRPSSSIQESSSESEDGDARGE IFDIYWTADYLPLGAEQDAITLREGQYVEVLDAAHPLRWLVRTKPTKSSPSRQGWVSPAYLDRRLKLSP EWGAAEAPEFPGEAVSEDEYKARLSSVIQELLSSEQAFVEELQFLQSHHLQHLERCPHVPIAVAGQKAVI FRNVRDIGRFHSSFLQELQQCDTDDDVAMCFIKNQAAFEQYLEFLVGRVQAESVWSTAIQEFYKKYAEE ALLAGDPSQPPPPPLQHYLEQPVERVQRYQALLKELIRNKARNRQNCALLEQAYAWSALPQRAENKLHV SLMENYPGTLQALGEPIRQGHFIVWEGAPGARMPWKGHNRHVFLFRNHLVICKPRRDSRTDTVSYVFRNM MKLSSIDLNDQVEGDDRAFEVWQEREDSVRKYLLQARTAI IKSSWVKEICGIQQRLALPVWRPPDFEEEL ADCTAELGETVKLACRVTGTPKPVISWYKDGKAVQVDPHHILIEDPDGSCALILDSLTGVDSGQYMCFAA SAAGNCSTLGKILVQVPPRFVNKVLASPFVEGEDAQFTCTIEGAPYPQIRWYKDGALLTTGNKFQTLSEP RSGLLVLVIRAASKEDLGLYECELVNRLGSARASAELRIQSPMLQAQEQCHREQLVAAVEVTEQETKVPK KTVI IEE I TWKSPRGQRRSPSKSPSRSPSRCSASPLRPGLLAPDLLYLPGAGQPRRPEAEPGQKPW PTLYVTEAEAHSPALPGLSGPQPKWVEVEETIEVRVKKMGPQGVSPTTEVPRSSSGHLFTLPGATPGGDP NSNNSNNKLLAQEAWAQGTAMVGVREPLVFRVDARGSVDWAASGMGSLEEEGTMEEAGEEEGEDGDAFVT EESQDTHSLGDRDPKILTHNGRMLTLADLEDYVPGEGETFHCGGPGPGAPDDPPCEVSVIQREIGEPTVG QPVLLSVGHALGPRGPLGLFRPEPRGASPPGPQVRSLEGTSFLLREAPARPVGSAPWTQSFCTRIRRSAD SGQSSFTTELSTQTVNFGTVGETVTLHICPDRDGDEAAQP
Biomarker: Actin binding LIM protein 1 (AbLIMl)
mRNA Sequence
NCBI Reference Sequence: NM_001003408.1
SEQ ID NO: 19
GTCATTGAGCTGATGTGCTTTTGCAAGGGGTCTCTTTAGAAGTGGTAGGGGCAGGGTCGAGAGAAGAAGG GGATGCAGAATCTGTACATGAGGAAGGAGCCAGAGTCACTCAGGAAACTCCTCATTCTTCCTGCGCTGGC TGTGGCACACCCACCGGTGGCACACCCCTCCCTCGGGTCCAGTTCAGCCCACAGACTCCTGGGAGGAGAG CCAGGGCCCCAGCCTCTCCTTTGAGGAGTGTTGCAGCCTTTCTGTGTGCCAGATGCCTAGTGAAGAAAAG CTGAAACCCTGGAGCTTTGTGCATGTTAATGACCTTGGAAATGACTGAGCTCACGGACCCTCATCACACC ATGGGAGACTACAAAGTGGCCCACCCTCAGGACCCTCACCACCCATCAGAGAAGCCTGTCATTCACTGCC ATAAATGTGGGGAGCCTTGCAAGGGTGAAGTGCTTCGGGTCCAGACCAAACATTTCCACATCAAGTGTTT CACCTGCAAAGTGTGTGGCTGTGACCTGGCACAAGGGGGCTTCTTCATAAAGAACGGAGAGTATCTCTGC ACCCTGGACTACCAGCGGATGTACGGGACACGCTGCCATGGCTGTGGGGAGTTCGTGGAGGGCGAAGTGG TGACTGCTCTGGGCAAGACCTACCATCCCAATTGCTTTGCTTGTACTATCTGCAAGCGCCCGTTTCCACC CGGAGACCGAGTCACATTCAATGGGAGAGACTGCCTTTGTCAACTCTGTGCACAGCCGATGTCGTCCAGT CCGAAAGAAACCACCTTCTCCAGCAATTGTGCCGGCTGCGGAAGAGATATCAAGAATGGGCAGGCGCTGC TGGCGCTGGATAAGCAGTGGCACTTGGGGTGCTTTAAATGCAAGTCCTGCGGGAAGGTCCTCACCGGGGA GTACATCAGCAAGGATGGTGCTCCGTACTGTGAAAAGGACTACCAGGGACTCTTTGGGGTGAAATGTGAG GCGTGTCACCAGTTTATCACAGGGAAAGTCCTGGAGGCAGGTGACAAACATTACCACCCCAGCTGTGCAC GATGCAGCAGATGCAACCAGATGTTCACAGAAGGAGAGGAAATGTATCTTCAAGGCTCCACCGTTTGGCA TCCCGACTGTAAGCAATCTACGAAGACCGAGGAAAAGCTGCGGCTGCCAAACATTCGGCGTTCTTCATCC GATTTCTTTTATTCCAAAAGTCTGATTCGACGCACAGGACGGTCTCCGTCTCTGCAGCCTACCAGGACAT CCTCGGAAAGTATTTATTCTAGGCCAGGCTCCAGTATTCCTGGCTCACCAGGTCATACTATCTATGCAAA AGTAGACAATGAGATCCTGGATTACAAGGATTTAGCAGCCATTCCGAAGGTCAAGGCAATTTATGACATT GAACGTCCAGATCTTATTACCTATGAGCCTTTCTACACTTCGGGCTATGATGACAAACAGGAGAGACAGA GCCTTGGAGAGTCTCCGAGGACTTTGTCTCCTACTCCATCAGCAGAAGGGTACCAGGATGTTCGGGATCG GATGATCCATCGGTCCACGAGCCAGGGCTCCATCAACTCCCCTGTGTACAGCCGCCACAGCTACACTCCA ACCACGTCCCGCTCTCCCCAGCATTTCCACAGACCTGGCAATGAGCCGTCCAGCGGCCGGAACTCCCCTC TCCCTTACCGGCCAGACAGCCGCCCTCTAACTCCAACTTACGCTCAGGCCCCTAAACATTTCCATGTTCC AGATCAAGGAATCAACATTTACCGAAAGCCACCCATCTACAAACAGCATGCTGCCTTGGCAGCCCAGAGC AAGTCCTCAGAAGATATCATCAAGTTTTCCAAGTTCCCAGCAGCCCAGGCACCAGACCCCAGCGAGACAC CAAAGATTGAGACGGACCACTGGCCTGGTCCCCCCTCATTTGCTGTCGTAGGACCTGACATGAAACGCAG ATCTAGTGGCAGAGAGGAAGATGATGAGGAACTTCTGAGACGTCGGCAGCTTCAAGAAGAGCAATTAATG AAGCTTAACTCAGGCCTGGGACAGTTGATCTTGAAAGAAGAGATGGAGAAAGAGAGCCGGGAAAGGTCAT CTCTGTTAGCCAGTCGCTACGATTCTCCCATCAACTCAGCTTCACATATTCCATCATCTAAAACTGCATC TCTCCCTGGCTATGGAAGAAATGGGCTTCACCGGCCTGTTTCTACCGACTTCGCTCAGTATAACAGCTAT GGGGATGTCAGCGGGGGAGTGCGAGATTACCAGACACTCCCAGATGGCCACATGCCTGCAATGAGAATGG ACCGAGGAGTGTCTATGCCCAACATGTTGGAACCAAAGATATTTCCATATGAAATGCTCATGGTGACCAA CAGAGGGCGAAACAAAATCCTCAGAGAGGTGGACAGAACCAGGCTGGAGCGCCACTTAGCCCCTGAAGTG TTTCGGGAAATCTTTGGAATGTCCATACAGGAGTTTGACAGGTTACCTCTTTGGAGACGCAACGACATGA AGAAAAAAGCAAAACTCTTCTAAGTCCCACTCGTGAATGGCAATTAGAGAAAGGACTGACAGTGGCGGTG CCCCATAGGATGTCATATTGAGGCCCAAACTTGATTGGAGAATTTGCAAACTACCGTCGCTCAGCAACAC CAAAAAGAGAAAGTCTGGTTAAAACACCATGAGTCAAATGTCGGGCCAGCCAACAGTAACACTTGCCAAG AAGCATGGCGTAGAAATTTCTATGTTCCGAAACACAGAGTAGTGTCCACATGTGAGCTTTTCACATTACC TTATGTATACAACAGGAGCTGCGTTGTTTTCTTCTTTTTCTTTTCCTTTATCTGTTGTCCAACAACAGTG CTGACTGTCCGGATAAGAGCTGGCAAGTGCCCTTAGGATGCCGCATGGGAAAAATCGGTTATCATAATTT CAAAGTATAAATATATTTATTAAGTAGCGCTGCGGCTAAGAAGGAAGAGTGAGGGGTCTGTCCATGGGGT GGCAGTGATTTCACACCCGCCTTTCTTGAATGGCTTCGTGTTACTCAGCCGTGCCCTGGCAGGAATGGAA CTCCATCAGGGAACAGGGCAGCTCTGTTTGAATGGGGTGAAAAACAGCAAAATTAATTCTTAGCAAGCTC CTTGTTCTCTACTGTTCACCGGGACCTGGCTGGCAAATGACTTAGCTATTAGTATATTTAAAAACTGTTC AAAAGCAAAAGAGAAGGAACAGGAATGAAAGGAAACCTTCCCTTAACTCTTTCTGCTTCCCATAGCAGGT CTGGTTCCTGCTCCCCGCCTAGGACAGGAGCTGTCTGGAGCTCCACCTTTGCAAAGAAAGGAAAAACACA AATTGCCCCCAGATTGGCCCAGTGGCATGCCATTAGGTGATTTTGCATAGGGGCCATATCATTCATGGCC AAAAAAAACCAAAAAGCAAAAAACAAAAACAAGTCCCATGTCCTACTGACTCTAGTTCTTCCAGTCGTTA TCCTCTGCTGTCTCTCTGGCATCCTATGAATCAGATCAAGCCCATGCTGTTGTTGGTTTTTAAGGTTTCT TTCAATAATAGGCTAAGGAAAGACATGTTTTTCTCTTTTAAATCTCTGCAACTCCAAAGTAGACTCCTTC GAACGTATTTAATTTGGCCTTTTCAGCTTTCTTCTTGCCCAGCTCTGTCGAATTCATGGGTGTGTGTGCA CATGTTGGTTTCACTCACCCAGAGTAATTTTGTGAGCATGCATGTGCTTTTTAATTTCTGCTTGAATGTT TGTCCTGCTGTGTTGCAGCTTCTAAAGACATTGTCACCGAGTGTGTGTGACTCAAAGATCAAGAGTGAGG CTAAACTGCGACCCCAACATCACTTCCTTCATGGAAAGGTGTTGGCGCTGATGTAGCTCATGGAAGGATC AGACTGGGAACCACAGAGGAAGGATGAGGTGGGATGTGGAGGTCAGAGCATCTCACGAGAGTCCCTCAGG CTCTGCAACACTAGATTCGAATATGAGCCTAGGGTTTCCCCAACACGTGCGTGCATATGAAACCACATCA TGATTCCCGACGTGAGTTAGATGAGGGAGAGTTTGTGTAACTCCACACCCAGGAGGGTAAGCCACTTTAT TATCATGCTTTGGGGCTGTGCTTAAGAATTTAAGCTCTGTTTTAAAGCCGAAGAAGAAAACATTTGAATT TACCTTCATATGCCACTAAGAACAAAAGCTCTGAAGTTCTTCCTGCTGGCACCAAATATTAGGATTACAA GCTGTCTCTCTGACACCAGTGGTAAAAAAGCAAGTAGAATAGTTTCTGAGTCTGTGTTGCAGCCTCTGAC ACGTGGCTCACCGCTGTCATTTGGGGGATGGAATGAACAGTCACTGTTCTATAGCATCCTCCTGTGCTCT TACCTAAGGAATCTAAATTGTCCTAGCCTTCCATTGCTATGCACTGAATGAGGACTACTCTCGTGTGTGT GTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTTGGATCTACTAACATGTTTAGCAAATGG CTCTGACAGGGTGGCCATGAGCAGATGAGACCAGCTCTTGCTTGGTTGGAAGCCACACTGCAGTTTGCAG TTTAGCCTTTGGTGTCTCAGTGGCTTCTTGCTTATTCCATGCTTTTTCTATCCCATCCTCTCTCTGTGCC CTCCTGCAATGGCAGCCTTGGCACAATGCCTGGGTGTCCTCATCCTTGCTTTACTCTGGAAAGTGTGGGG ATGGAGAGGATGGTGCATTTCCAGTGCTTCCTTACCTGTCAATAACAATGGTGGCCACCTGGGTCCCAGC ACTTGGCTGGTGGAGAGATTTTTTTCATTATGGAGCTAATAAAGGGTCCCTACGTTTTACATTGATTAAT GAATTATCACCCAGACATGTGCAGATGAAATGTCAATATAAGAAAAATCAAGGTATTTGGGAAACATGGG CATAATCAGGTAGTTTTGCTAAAATTCCTCTTCCTTGGTCTACATAAAGAAGACATGGGTCTGTCATAGA ATGGGGACATTAGACTGAAAACTGGGACAACCCCCTTCCTTATTTTATAGAGACTAGGAGTTAAGGGGCC TGCCTAAGATTGCAGTGATAAAGAACTGAACCCTAAATCAGCTCCCACTAATGAAACCCTGCTTTCCTTG TATCTTTTAAACTCAGTTCTGCATCCAACTTGAGAAGAAAAAAAGCTTCTTTCCATATCAAGTACCATAT GAGTTCATTTAAACTGCACCTTGCAGAAATGCATTGCCAGAAAGCACCAGTAGCCTCCTATCTGCAAGCA GAGTAGTGCTCTGCTCTGGGGAGGGGTCATTGGAAACCATAATGCAGAGTGGGCCCCCTACTCCATTTCC CAGCAAAAGGCTCCAGCTGGAGGGATGGGTTGTGGGGCAACCTGGTTCCTGCTAACTGCCAGATTGAATG TGTGGGCTAGAATGCCTGCACATTTAGTTAAACTGGGCTCAGCATGCTTGTCCTCAAAATGTCCATCCTG GTCACAGCACACAAGATGGCTATTGGTCTGCTTTTACCCTACCCTGTACTATACATGAAAATTCCAGTTA TTAACACACTCAAACTGGTGGAGCTTGTTCACCCTAGGAAGGGGATTGTATATATGGCAGGCTTCCCTGG TGCCGATGTAAAGGGCTACATTTGGGAACATTTGACTTCCTTGGGACTCTTAAGTGCATAATGATGGCAT GAAGTAAAAGGGGCCTCAATGATGATAGGAAAATCAGTTCTTTTAAAATTTCTTCAAGAAAATCCAGGCT ATCACATAGTCTTTCTGTGTGACTTATTAGGAGATAGGAAGAGCATTGGGAAACTTGCACAGCTAGCTAT GCATCTACATTTTGGTTTGGGGTTAGTTATGAAATGTTCTTAATATGACGTGTTCAATAACTTCACATAA ACTTCCTGTTCTCCAAAACCTCAAAGAGATAGAGTTAATGAGTTGTTGTTTTTTTTTAAATGGGGGTAGT TTTCTATCTGTCATGGGCTCTAGCATCTACTCCGCTACCCAATTCTGTCATCTCCAAGCTGAGTTTCCTC TTCTGAGGCAGAGGCTGGAGCAGTTCTTTTTCAGTTCTCATCCTCTCCATCCCAATCCAGTATATCAATC AACTCTAACTCGGAGACGTCTAGCTGGCAATGTTTCTAAAACTTTCACTGGATTTCTTTAGACATTGAAG CAAACATTTTTTTCTAAGAATTGCTTCTCAGATGATGATATCAAATGTATATGCTTTTGCAAGTTTGAAA AGTTCAAATTAACCACTTTTGACTAGGTAAGTCTTTCTAAAAACCATTTAAAGCTAACTGGGTCTTAGCA TCCTCCTGTGTATGGAAGAGACAGGTGACCGCTCCAGGTTGGGTGCTCACAGAACCCTTTTCCTGACTCT CATGGAAGATGGTGGAAGGAAAATAGACTGTCTCATCAACCCTCCTGTGTCCTCTGAAGCAATCTCAGTT TTTATTAACCACCTCTTCTGTGTTTCTGGTAGCTATTTAACCTGTATTTAATCTGTACTTCCTATGCCAG CCTCAATTTTATTTGATTTTTACAATTATTCTCTTCTAACCAATGAAGTGTTTGTCAGTATGCCCCAAAG CTTGCTCTTTTGTGCTCCCTTTTGAATAACTTTCTATCCAGAAAAAGAGATTATTTGGGACTTGAGATTT GCAGTGA ACCAACTTATAGCAA GATG AC TTAAGGGAAC ACCCAACTATGTTG GA AGAAGAAAG AGAAACCTTCACTTTGGCATTTTTTTTTAATCACTGTTTATTTTTCTGTTTGCGGCCCAGGAAGCAGTGG GAGGTGGTGGCAGATATGCTTTGCATATGGATTGTTATGTTTTTATTTGGGCAAGTTTAATCATGGAAAA CTCAAAAAGAAGGGGGGAAATGGTCAGTTTAAGCCAAAAGAAACTTTCTAAACAATGTATAGGTACACAG CAAAATTAAACAAATCCAACAATTTCTGAAGCTTAGTGTAATTGAGTGGTGGTTGTTATTCAATAAAATT ATTCCCAAAAGTGTTTCTCCTAAGAGTGCAGTTCCCATGAGTCACTTCCTGAACCCATTGACCAAAGGTG GACAGAGACAATCCTGTAGACCTTGACATTCAGAAAGATGTGAGCTGCTTACTGATCATATATGCATACG TTTCTTTACAGCAGAGGAAACCATTGTCCACAAAACTGATGTTCTTTTGGGGTTTTATGTACAGACTTGT CCAATCATGTGTGTGGTTCCTGCGAGTTGCTGATGACTCCGCATTGAAGCTCTCTGAGTTCTTTGATTTT AAGTTGGGTTTATGGAATTTTTTCAAATGTTGGAAGGTGTGTGGTTCTTCCTGCCCTCCCTCCCCTTTTG GAAATATGAAAGCAAATGTTTAGAAGAATTCCTTTTGAAAAGCTGTGTCGTGTTCCCTGTGAAACTGAGC AGGTGTGTGTTGGCGCGCTAAGTGCCACATGCTTGTGTGTAGAGGAGGAGGTGGCCCTGCCGGCTCCGCG CTGCTGTGCCTGTGATCCCTACCTGCTCCCCGCTCCTGTTGCCAGCAGCACTCACTGCACTCCTTTGTCA TATACTCTGCATCACTGTCATACTCACAACTTCGTGAATAAAGTTGTGTGCTTTATTCGTAGAAAAAAAA AAAA
Amino Acid Sequence
GenBank: AAH02448.1
SEQ ID NO: 20
MFTEGEEMYLQGSTVWHPDCKQSTKTEEKLRAKVDNEILDYKDLAAIPKVKAIYDIERPDLITYEPFYTS GYDDKQERQSLGESPRTLSPTPSAEGYQDVRDRMIHRSTSQGSINSPVYSRHSYTPTTSRSPQHFHRPDQ GI IYRKPPIYKQHAALAAQSKSSEDI IKFSKFPAAQAPDPSE PKIETDHWPGPPS AWGPDMKRRSS GREEDDEELLRRRQLQEEQLMKLNSGLGQLILKEEMEKESRERSSLLASRYDSPINSASHIPSSKTASLP GYGRNGLHRPVSTDFAQYNSYGDVSGGVRDYQTLPDGHMPAMRMDRGVSMPNMLEPKIFPYEMLMVTNRG RNKILREVDRTRLERHLAPEVFREIFGMSIQEFDRLPLWRRNDMKKKAKLF
Biomarker: Tensin-1
mRNA Sequence
NCBI Reference Sequence: NM_022648.4
SEQ ID NO: 21
CTCAGTTGCTCCCAGCAGTGGCCCTGGGACCAGCTCTGCTCCTTGCACCCCGCTCCCTGCCTGGACACAG GCTCACTCGCTGCCTTCTTCTGGGGGAAACCAGCTTCTTGCCAGCCACAGCTGCTGCCTCCGCCACTGGC CACCGCCCCTGTCCTGGGAGTCCCTTGGCCCAGACACCCACCTGACTTAGTGGCTCCTCTGCAGGAAAGG TGGCTGCCCCCTGCGTTCCTCCATCCAACCATGAGCTGGTGCCCATCACCACTGAGAATGCACCAAAGAA TGTAGTGGACAAGGGAGAAGGAGCCTCCCGGGGTGGAAACACACGGAAAAGCCTCGAGGACAACGGCTCC ACCAGGGTCACCCCGAGTGTCCAGCCCCACCTCCAGCCCATCAGAAACATGAGTGTGAGCCGGACCATGG AGGACAGCTGTGAGCTGGACCTGGTGTACGTCACAGAGAGGATCATCGCTGTCTCCTTCCCCAGCACAGC CAATGAGGAGAACTTCCGGAGCAACCTCCGTGAGGTGGCGCAGATGCTCAAGTCCAAACATGGAGGCAAC TACCTGCTGTTCAACCTCTCTGAGCGGAGACCTGACATCACGAAGCTCCATGCCAAGGTACTGGAATTTG GCTGGCCCGACCTCCACACCCCAGCCCTGGAGAAGATCTGCAGCATCTGTAAGGCCATGGACACATGGCT CAATGCAGACCCTCACAATGTCGTTGTTCTACACAACAAGGGAAACCGAGGCAGGATAGGAGTTGTCATC GCGGCTTACATGCACTACAGCAACATTTCTGCCAGTGCGGACCAGGCTCTGGACCGGTTTGCAATGAAGC GGTTCTATGAGGATAAGATTGTGCCCATTGGCCAGCCATCCCAAAGAAGGTACGTGCATTACTTCAGTGG CCTGCTCTCCGGCTCCATCAAAATGAACAACAAGCCCTTGTTTCTGCACCACGTGATCATGCACGGCATC CCCAACTTTGAGTCTAAAGGAGGATGTCGGCCATTTCTCCGCATCTACCAGGCCATGCAACCTGTGTACA CATCTGGCATCTACAACATCCCAGGAGACAGCCAGACTAGCGTCTGCATCACCATCGAGCCAGGACTGCT CTTGAAGGGAGACATCTTGCTGAAGTGCTACCACAAGAAGTTCCGAAGCCCAGCCCGAGACGTCATCTTC CGTGTGCAGTTCCACACCTGTGCCATCCATGACCTGGGGGTTGTCTTTGGGAAGGAGGACCTTGATGATG CTTTCAAAGATGATCGATTTCCAGAGTATGGCAAAGTGGAGTTTGTATTTTCTTATGGGCCAGAGAAAAT TCAAGGCATGGAGCACCTGGAGAACGGGCCGAGCGTGTCTGTGGACTATAACACCTCCGACCCCCTCATC CGCTGGGACTCCTACGACAACTTCAGTGGGCATCGAGATGACGGCATGGAGGAGGTGGTGGGACACACGC AGGGGCCACTAGATGGGAGCCTGTATGCTAAGGTGAAGAAGAAAGACTCCCTGCACGGCAGCACCGGGGC TGTTAATGCCACACGTCCTACACTGTCGGCCACCCCCAACCACGTGGAACACACGCTTTCTGTGAGCAGC GACTCGGGCAACTCCACAGCCTCCACCAAGACCGACAAGACCGACGAGCCTGTCCCCGGGGCCTCCAGTG CCACTGCTGCCTTGAGTCCCCAGGAGAAGCGGGAGCTGGACCGCCTGCTGAGTGGCTTTGGCTTAGAGCG AGAGAAGCAAGGCGCCATGTACCACACCCAGCACCTCAGGTCCCGCCCAGCAGGGGGCTCGGCTGTGCCC TCCTCTGGACGCCACGTTGTCCCAGCCCAGGTTCATGTCAATGGTGGGGCGTTAGCATCTGAGCGGGAGA CAGACATCCTGGACGATGAATTGCCAAACCAGGATGGTCACAGTGCGGGCAGCATGGGCACACTCTCTTC TCTGGACGGGGTCACCAACACCAGTGAGGGGGGCTACCCAGAGGCCCTGTCCCCACTGACCAACGGTCTG GACAAGTCCTACCCCATGGAGCCTATGGTCAATGGAGGAGGCTACCCCTACGAGTCTGCCAGCCGGGCGG GGCCTGCCCATGCTGGCCACACGGCCCCCATGCGGCCCTCCTACTCTGCACAGGAGGGTTTAGCTGGCTA CCAGAGGGAGGGGCCCCACCCAGCCTGGCCACAGCCAGTGACCACCTCCCACTATGCCCATGACCCCAGC GGTATGTTCCGCTCTCAATCCTTTTCGGAAGCTGAACCCCAGCTGCCCCCAGCTCCGGTCCGAGGGGGAA GCAGCCGGGAGGCTGTGCAAAGGGGACTGAATTCGTGGCAGCAGCAGCAGCAGCAGCAGCAGCAGCCTCG CCCACCTCCACGCCAGCAGGAAAGAGCCCACTTGGAGAGTCTTGTAGCCAGCAGGCCCAGCCCTCAGCCA TTGGCAGAGACCCCCATCCCCAGTCTCCCTGAGTTCCCGCGAGCAGCCTCCCAGCAGGAGATTGAACAGT CCATCGAAACACTCAATATGCTGATGCTGGACCTGGAGCCAGCCTCCGCTGCTGCCCCACTACACAAGTC CCAGAGTGTCCCCGGGGCCTGGCCAGGGGCTTCTCCACTCTCCTCCCAGCCCCTCTCTGGATCCTCCCGT CAGTCCCATCCACTGACCCAGTCCAGATCTGGCTATATCCCCAGTGGGCATTCGTTGGGAACCCCTGAGC CAGCCCCACGGGCCTCTCTGGAGTCTGTCCCTCCTGGCAGGTCTTACTCACCTTATGACTATCAGCCATG TTTGGCTGGGCCTAACCAGGATTTCCATTCAAAGAGCCCAGCCTCTTCCTCCTTGCCTGCCTTCCTTCCG ACCACCCACAGCCCTCCAGGGCCTCAGCAACCCCCAGCCTCTCTCCCTGGCCTCACTGCTCAGCCTCTGC TCTCACCAAAGGAAGCGACTTCAGACCCCTCCCGGACTCCAGAGGAGGAGCCATTGAATTTAGAAGGGCT GGTGGCCCACAGGGTAGCAGGGGTACAGGCTCGGGAGAAGCAGCCTGCAGAGCCCCCAGCCCCTCTGCGG AGGCGGGCGGCCAGTGATGGACAGTATGAGAACCAGTCTCCAGAAGCCACATCCCCTCGTAGCCCTGGGG TTCGCTCCCCTGTCCAGTGTGTCTCCCCGGAGCTGGCTCTTACCATCGCTCTCAATCCTGGAGGGCGGCC CAAAGAGCCCCATTTGCACAGCTACAAGGAGGCCTTCGAGGAGATGGAGGGAACCTCCCCGAGCAGCCCA CCACCCAGTGGGGTGCGGTCCCCCCCGGGTCTGGCCAAGACACCCCTGTCTGCTCTGGGCCTGAAACCTC ACAACCCAGCGGACATCCTGTTGCACCCCACAGGAGTTACCAGAAGACGGATCCAGCCAGAGGAAGATGA GGGGAAGGTGGTGGTCAGGCTGTCAGAAGAGCCCCGGAGCTATGTGGAGTCTGTGGCACGGACAGCGGTG GCTGGACCCCGAGCTCAGGACTCTGAGCCCAAGAGCTTTAGTGCTCCAGCCACCCAGGCCTATGGCCATG AGATACCCCTGAGGAACGGGACCCTGGGTGGCTCCTTTGTCTCCCCCAGCCCCCTCTCCACCAGCAGCCC CATCCTCAGTGCTGACAGCACTTCAGTGGGGAGTTTCCCGTCGGGAGAGAGCAGTGACCAGGGTCCCCGG ACGCCCACCCAGCCTCTGTTGGAGTCTGGCTTCCGCTCAGGCAGCCTGGGACAGCCCAGCCCGTCTGCCC AGAGAAACTACCAGAGCTCTTCTCCTCTCCCGACTGTGGGCAGTAGCTACAGCAGCCCCGACTACTCACT TCAGCATTTCAGCTCCTCTCCGGAAAGCCAGGCTCGAGCTCAGTTCAGTGTGGCTGGCGTCCACACGGTG CCTGGGAGCCCTCAGGCGCGCCACAGAACAGTGGGCACCAACACTCCCCCTAGTCCTGGCTTCGGCTGGC GGGCCATCAATCCCAGCATGGCTGCCCCCAGCAGTCCCAGTTTGAGCCATCACCAGATGATGGGTCCACC AGGCACTGGCTTCCATGGTAGCACTGTCTCCAGCCCCCAGAGCAGTGCAGCGACCACCCCGGGGAGCCCC AGCCTGTGTCGGCACCCAGCAGGGGTCTACCAGGTTTCTGGCCTCCACAACAAAGTGGCCACCACCCCGG GGAGTCCCAGCCTGGGCCGGCACCCTGGGGCTCACCAAGGCAACCTGGCCTCCGGTCTTCATAGCAATGC AATAGCCAGCCCTGGAAGCCCCAGCCTGGGCCGTCACCTCGGAGGGTCTGGATCTGTGGTTCCCGGCAGC CCCTGCTTGGACCGGCATGTGGCCTATGGTGGCTATTCTACCCCGGAGGATCGGAGACCCACACTGTCCC GGCAGAGCAGTGCCTCTGGCTACCAGGCTCCTTCCACGCCCTCCTTCCCTGTCTCCCCTGCCTACTACCC TGGCCTGAGCAGCCCTGCCACCTCCCCGTCACCAGACTCCGCAGCCTTCCGGCAAGGGAGCCCAACACCA GCCTTGCCAGAGAAGCGAAGGATGTCAGTGGGAGACCGGGCAGGCAGCCTCCCCAACTATGCCACCATCA ATGGGAAGGTGTCTTCGCCTGTCGCCAGCGGCATGTCCAGTCCCAGTGGGGGCAGCACCGTCTCCTTCTC CCACACTCTGCCCGACTTCTCCAAGTACTCCATGCCAGACAACAGCCCGGAGACGCGGGCTAAAGTGAAG TTTGTCCAGGACACTTCTAAGTATTGGTACAAGCCTGAGATCTCCAGGGAGCAGGCCATCGCGCTCCTCA AGGACCAGGAGCCGGGGGCCTTCATCATCCGCGACAGTCACTCCTTCCGAGGCGCGTACGGGCTGGCCAT GAAGGTGTCTTCGCCACCTCCAACCATCATGCAGCAGAATAAAAAAGGAGACATGACCCATGAGCTGGTC AGGCATTTTCTGATAGAGACTGGCCCCAGAGGAGTCAAGCTCAAGGGCTGCCCCAATGAGCCAAACTTCG GATCGCTGTCTGCCCTGGTCTACCAGCACTCCATCATCCCATTGGCCCTGCCTTGCAAGCTGGTCATTCC AAACCGAGACCCCACAGATGAATCGAAAGATAGCTCCGGCCCTGCCAACTCAACTGCAGACCTGCTGAAA CAAGGGGCAGCCTGCAATGTGCTCTTCGTCAACTCTGTGGACATGGAGTCACTCACTGGGCCACAGGCCA TCTCTAAAGCCACATCTGAGACGTTGGCTGCAGACCCCACGCCAGCTGCCACCATCGTTCACTTCAAAGT CTCTGCCCAGGGAATCACTCTGACTGACAACCAGAGAAAGCTCTTTTTCAGACGCCACTACCCTCTCAAC ACTGTCACCTTCTGTGACCTGGATCCACAGGAAAGAAAGTGGATGAAAACAGAGGGTGGTGCCCCTGCTA AGCTCTTCGGCTTCGTGGCCCGGAAGCAGGGCAGCACCACGGACAACGCCTGCCACCTCTTTGCTGAGCT TGACCCCAACCAGCCGGCCTCTGCCATCGTCAACTTCGTCTCCAAGGTCATGCTGAATGCCGGCCAAAAG AGATGAACCCTGCCCCTTGCCCAGGGCCAGTGCCATGGGGAAGGGGCTTGTGGGGAGGGGACCCATGAAT CCTGACCACTCTTGAACCCAGAAGGAGGACTTTGGGCCAATTTCGGAGGAGAGAAGAAAGTGCAACGTGG G G AG AG G G AAG T G AAT TGCAGAGGGGAGGGG GAAAAG AG AG AG AG AG AG AG AG AG AG AG AG AG AG AG AAA GATGGAGGAGAAGAACTTGGATTCCCCTGGGTAGATGGAAACTGCAAAAACCCAAAGCCTCCAAAACTAA CCAGGTCCACCTAACACCCCCTCCCTCCCCTAAGAAGATGGATGTCCTCAAAAGAGAAGGAACAAACCTC CTTGGGAATCCACATTTTTTGGGGGAATGGAAAAGCTCTGTCTCCCTAACTCAACTGCTTTGCAAGGGGA AATCAAGCTGGGAGAATCTTTTTCTGGCCACCTGTGGGGTAGGTTGTCAAACCAAACAGAGCCACCGTGG GACATCAAGTGGAAGAACTTGTTTGCTTGAAAGTATCTCAGACCCAAGGCACCTCAGGTCTCTTTGCTGT GCCTCCACTATATTGTCGTGTGGGTGTGTGTCTGCACCCACATCCTCACACATTGATCTAGATCTGCCTT TATCCACTCGAATTATAAACAGCTCGGCTTGTCCTTGTCCCATGTGTTTGTAGACACACATGCATACTGT CCAAAGATTAGGGTTGGTGGTGGCAGTGCAGCAGGGGAGGGACAAACAACCAAGCTATGGGTGACAGAGG CTCTCTCCTGGTGCCTGCACCTGCACTCTAGTGACCCTGGGTGCCGCCAGACCCTTCTCTTCTACAAAGA CCCCAGCAGGAGTGGGAGGGTCTGCAATGGCATCGCCCTGTCCTGCCTTGGCCAGAAGCCTGGAGCTTTG GTTTGAGGAGGTAGAGATATGTGTATCCATAGGAAGAGATCTGTCAGAACAGGCAGCTGTTGAGCTCGGG GTGTCTTCCCCAAGGCATGTGGCTCAGCAGCAAGAAAGGCAAGTTGCTCCTGCTGGGGCCCTGGACTCTG CCTTAGCTCCCACCTCTCAGCCTTGTTATTGGGTTTCATGCCCCTGGACCAGCCTTATCTCAGACCTGCT TACCTGCATGATGCCTTTTTGGGGGCTGGGGATTGAGTCTTGCTGCTCTGCCCAGCCCTGTTCTATTCTG CAGGGTCCCTGTGTTGGAATTCTCCCTGGGGAACCTACTTTCTGCTCAGTGAGGCTCCGGCCAGAAACCT GGAGTCCTTATCCTCCCCTCTGTAAGTGTTTTAGGGTCTGGCTTTTGCAGGCACCCTCTGACCTCAGCAG AGCTCCTGGGCCTGCTGCCTGCACACCACATCGCCTACCTACAATGCCAAAGCCTCACTGTCACCCTTTC TGCCTTGGTTTCCCTAGCTGAGCCACGCTGCCCATGCAGCAGAGGGCAGAAGGCTTGCACTTGGGCCAAA GGGCCTAAGGTCCACTGGACAGTTGGGAAAACACCTGACCACCATTTAAGGACTCTAAGCCAGAATGGAA AATTCACCAGGACTCCATTCTTAAGCCTATGCGAGTCCCCTAGAGAGAGGCATTGTACTGATATATAAAT ATTATATAATATATACATGAGACATACTGACAGAATCTGTAAGCTAATAAAATGTAAGAAAAGGTTAAAA AAAGAATAGGTAAATTGACAAGAAGTATTTATTGTTTTTCCATATTGCTTTATTGCCTTCCTTGGGGATA AACCAATTCCTATCCTTTTTTATATGTGTAAGTAAAGCCTGAAGTGTAGGGGGCCTTTGTTCTTGAAGCA GCCAGGGTCTCCTTGCCCTGGCCTTGGCCTTCCCTAGACTGTGTGGGGCTCAGCATTGGGAGGGGTTGCA CATGTCCCAGCCTTTGGCCCCCTTACTTTTCAGCAAGCCAGGGGCCCAGCAGTCAGCTCCCAGGATGTGT GGGGAGCTGTCCCTGACTCTGCAGGCCTGAGCGAGTGTGTGAGCATGCGGGGACATGGGTGTGTATGGCA CACATAGGTGCGTGTGTGTCTTTTGTATTTTTTCTCCTCCAAGGAGCTGTGTCAGTGTGGACGTTCTGTT TCAGGGAGTTGGAAAGGAGGGTGTCTGCAGAAGGTGGAGAGCAGGGGCAGAGGCCCCACTGGCCACCCCC TGCTTCCCAGAGTGAAACCTTGTGCCTGGTGACCAAAGTCCCTCCAAAGTGCTCTTCCTTCTGGGTTATT CAAGCCAAATATCTGGGTTTCCCCCTCTCCTCATTCCCTAGCAAACCCCAATTATCTTCCAAGATAGGAG ATATTTCCCATCCCCTTCCTTTGTAAATATCTCATCTCCCACTGGAGAGCCCAGGAGCCTATTCCTGGCA TGGATGTTCTGTCCACACTTGAGGCTGGGCGGTGTATCAGACCCTTCAAGCAGCCTGGCTGGGGCCCAGG ACTGAGTCTGGGGTCAGCTTTCACGGTCGCTTTTCCCTTCCTCACCACCCACCACAGCCCACCTTGCATG CATGGCCAGCCCCTCCACTCCAGCCTGAGCCATGTGTGCCCCTGCGGGAGGACCCATTCATGCCAGAAAG CTGGTAACTCCCTCCCAGCATCCCTGCGGAAGGAGTCAGTTTCTGAGAGTGTGACTTTTCAAGGCGAATG ATGGGGAAGGGTTCCCCAGTCCCCACAGTGGCCCCACCTCTGGGCCCTGCACCAGAGCCCTTCTGTGTCA CGGCGGGCTGTGCACCCATGCACACACCTACGCACACACAACACTCCGCACTGCAGTATATTCTTGCCAA AGATTTCCTTTAAAAGCAAGCACTTTTACTAATTATTATTTTGTAAATGTTTATCTTCTTCTGTCTTCTC CCTCCCTGAATCTATTTTACTGTTGTTTATTGTTGAATCTGTGTGTCAGCCAGGAGAGCGCTGTCTGGCC TTGAACATGGGCTGGGATGGGAAAGGGTCTGGGAGAAGATGGGCAACAAAGAGCCAGGGAGTCATGGACA TCGCAGCGACGCAGACCCCAGCAGGTTCAGTCCCGTGCTGCCACCAGCTGTCCAGCTGGGTGTCTGGAGG GAAGAGGGCAGAGGAGGGTCATGTCCCTTCAGCTGGGGGAGGGGCCCAGTGAGCTCCACGTGGCTTTTTC CCAAAGGGAGCAAGAGGGAAGGATTGGGCGAGAAAACAATGGAGAGGGGACCTGCGAAGGAAAACAGGGA GGAAGTGAGCGGTTTGATCAGCCTGCTATCACGGTGTTCTGGCTCTCTTATTTAGCCAGGCGCTTAAGGG ACAGATACATCACATCCTAAGTTTGGGAAAGGCCTTTGACCCATGTCATCTGAGCGTCTCCTCCAGTAGC TCTGAAAGCTGTGGACACCAATGGCCAGGATTCCTTCTCCCCTGGTTTTTGAGGATCCCTGGGTCTTCTG AGACTGGCCAGGAGAGGGATGGTGGGGCCAGTGGTTGTGTGAAAGCAGGAGGGGCAGCCCTCCTGGACAA GTGTGATCCCCCTATAAACGGCTCTCAGGAGGTTAGTGAGTAGGAGATTCTGCCTTGTTCTGATGAGCCT GTGCAGGGGCTCCAGGGGAGCATGCTGTCCAGGGGGCACAGAAGGGTGGTGAGTGTGATCAAATCTAGTC TCACTCCCACTTTTTAGTCTCACTCCTACTTTTGTCCACCACCCCTGCCTCCTGGATCTTCTCCCACTTT TTTTTTCAGCTTTAGGACCTGGGGAGATCCTGTGAGTCAAGGCAGACACCCAATCCTGCCCCCACACTCG GGGTCCTCCAAGAGGTTGGGGGGCAGAGTCCCAGAGCAGCCCTTTACCCCAGGTCCAGGCCCTGGAATCC TGAGACTCGCGTTTCCTTGGCCAGTGGTAACACAGGACGTGTGTGCGCATGTGCAAGTGTGGATGTATGT GTGTGCGTGTGTTTTGCTCATTTCTTTAGGGAACTTGGGAGTCGGGGTTGGAGGTGCTGGGCAATGGAAC TTCAAATTCAATGTCGCCCAGCAGTGAGGGGAGTCGGGAGGTGAGGCCTGTAGGCCAACCAATTGGTGGA GTCTCAGCGATAGCCCAGGTGAGAAGTGGTTCACCCAGAGGGGCAGGGTGGGGGCCTCGGGCAGATCTGT CCCTCTTGGCACCTCTGTCCTCAAATGTCCAAAATGTTGGAGGACCTCTGTTCATATCCCACGCCTGGGC TCTTGCCAGCAGTGGAGTTACTGTAGAGGGATGTCCCAAGCTTGTTTTCCAATCAGTGTTAAGCTGTTTG AAACTCTCCTGTGTCTGTGTTTTGTTTGTGCGTGTGTGTGAGAGCACATCAGTGTGTGCAGGCTGTGTTT CCCCATTTCTCTCCTCCCTTCAGACCCATCATTGAGAACAAATGTAAGAAATCCCTTCCCACCACCCTCC CTGCCTCCCAGGCCCTCTGCGGGGGAAACAAGATCACCCAGCATCCTTCCCCACCCCAGCTGTGTATTTA TATAGATGGAAATATACTTTATATTTTGTATCATCGTGCCTATAGCCGCTGCCACCGTGTATAAATCCTG GTGTATGCTCCTTATCCTGGACATGAATGTATTGTACACTGACGCGTCCCCACTCCTGTACAGCTGCTTT GTTTCTTTGCAATGCATTGTATGGCTTTATAAATGATAAAGTTAAAGAAAACTCTG
Amino Acid Sequence
NCBI Reference Sequence: NP_072174.3
SEQ ID NO: 22
MSVSRTMEDSCELDLVYVTERI IAVS PS ANEENFRSNLREVAQMLKSKHGGNYLLFNLSERRPDI KL HAKVLEFGWPDLHTPALEKICSICKAMDTWLNADPHNVWLHNKGNRGRIGWIAAYMHYSNISASADQA LDRFAMKRFYEDKIVPIGQPSQRRYVHYFSGLLSGSIKMNNKPLFLHHVIMHGIPNFESKGGCRPFLRIY QAMQPVYTSGIYNIPGDSQTSVCITIEPGLLLKGDILLKCYHKKFRSPARDVIFRVQFHTCAIHDLGWF GKEDLDDAFKDDRFPEYGKVEFVFSYGPEKIQGMEHLENGPSVSVDYNTSDPLIRWDSYDNFSGHRDDGM EEWGHTQGPLDGSLYAKVKKKDSLHGSTGAVNATRPTLSATPNHVEHTLSVSSDSGNSTASTKTDKTDE PVPGASSATAALSPQEKRELDRLLSGFGLEREKQGAMYHTQHLRSRPAGGSAVPSSGRHWPAQVHVNGG ALASERETDILDDELPNQDGHSAGSMGTLSSLDGVTNTSEGGYPEALSPLTNGLDKSYPMEPMVNGGGYP YESASRAGPAHAGHTAPMRPSYSAQEGLAGYQREGPHPAWPQPVTTSHYAHDPSGMFRSQSFSEAEPQLP PAPVRGGSSREAVQRGLNSWQQQQQQQQQPRPPPRQQERAHLESLVASRPSPQPLAE PIPSLPEFPRAA SQQEIEQSIETLNMLMLDLEPASAAAPLHKSQSVPGAWPGASPLSSQPLSGSSRQSHPLTQSRSGYIPSG HSLGTPEPAPRASLESVPPGRSYSPYDYQPCLAGPNQDFHSKSPASSSLPAFLPTTHSPPGPQQPPASLP GLTAQPLLSPKEATSDPSRTPEEEPLNLEGLVAHRVAGVQAREKQPAEPPAPLRRRAASDGQYENQSPEA TSPRSPGVRSPVQCVSPELALTIALNPGGRPKEPHLHSYKEAFEEMEGTSPSSPPPSGVRSPPGLAKTPL SALGLKPHNPADILLHPTGVTRRRIQPEEDEGKVWRLSEEPRSYVESVARTAVAGPRAQDSEPKSFSAP ATQAYGHEIPLRNGTLGGSFVSPSPLSTSSPILSADSTSVGSFPSGESSDQGPRTPTQPLLESGFRSGSL GQPSPSAQRNYQSSSPLPTVGSSYSSPDYSLQHFSSSPESQARAQFSVAGVHTVPGSPQARHRTVGTNTP PSPGFGWRAINPSMAAPSSPSLSHHQMMGPPGTGFHGSTVSSPQSSAATTPGSPSLCRHPAGVYQVSGLH NKVATTPGSPSLGRHPGAHQGNLASGLHSNAIASPGSPSLGRHLGGSGSWPGSPCLDRHVAYGGYSTPE DRRPTLSRQSSASGYQAPSTPSFPVSPAYYPGLSSPATSPSPDSAAFRQGSPTPALPEKRRMSVGDRAGS LPNYATINGKVSSPVASGMSSPSGGSTVSFSHTLPDFSKYSMPDNSPETRAKVKFVQDTSKYWYKPEISR EQAIALLKDQEPGAFI IRDSHSFRGAYGLAMKVSSPPP IMQQNKKGDMTHELVRHFLIETGPRGVKLKG CPNEPNFGSLSALVYQHSI IPLALPCKLVIPNRDPTDESKDSSGPANS ADLLKQGAACNVLFVNSVDME SLTGPQAISKATSETLAADPTPAATIVHFKVSAQGITLTDNQRKLFFRRHYPLNTVTFCDLDPQERKWMK TEGGAPAKLFGFVARKQGSTTDNACHLFAELDPNQPASAIVNFVSKVMLNAGQKR Biomarker: Thyroid hormone receptor-associated protein 3 (THRAP-3) mRNA Sequence
NCBI Reference Sequence: NM_005119.3
SEQ ID NO: 23
GGAGACGCTGGGCGCGCTGTGGGGCGGGGGCGAGGTTCGGGCTGGTTGTTCCGTTGCGAGCTGCAGCTGC GATCTCTGTGGTAGGCCCAGAAGTGTATGCTGACTTGTAAAGTGAAGAAGCCAGTGGTGCTGCGGGTGTT CTTTTGGGGTAGTGTCTGGGATCCAGTACGAGTTGAATCATTGTTCAAATAAGGTGTAATTGAAAAGTGA TCCTCTCTTCAGAGATGTCAAAAACAAACAAATCCAAGTCTGGATCTCGCTCTTCTCGCTCAAGATCTGC ATCAAGATCTCGTTCTCGTTCATTTTCGAAGTCTCGGTCCCGAAGCCGATCTCTCTCTCGTTCAAGGAAG CGCAGGCTGAGTTCTAGGTCTCGTTCCAGATCATATTCTCCAGCTCATAACAGAGAAAGAAACCACCCAA GAGTATATCAGAATCGGGATTTCCGAGGTCACAACAGAGGCTATAGAAGGCCCTATTATTTCCGTGGGCG TAACAGAGGCTTTTATCCATGGGGCCAATATAACCGAGGAGGCTATGGAAACTACCGCTCAAATTGGCAG AATTACCGGCAAGCATACAGTCCTCGTCGAGGCCGTTCAAGATCCCGGTCCCCAAAGAGAAGGTCCCCTT CACCAAGGTCCAGGAGCCATTCTAGAAACTCTGATAAGTCGTCTTCTGACCGGTCAAGGCGCTCCTCATC CTCCCGTTCTTCCTCCAACCATAGCCGAGTTGAATCTTCTAAGCGCAAGTCTGCAAAGGAGAAAAAGTCC TCTTCTAAGGATAGCCGGCCATCTCAGGCTGCCGGGGATAACCAGGGAGATGAGGCCAAGGAGCAGACAT TCTCTGGAGGCACCTCTCAAGATACAAAAGCATCTGAGAGCTCGAAGCCATGGCCAGATGCCACCTACGG CACTGGTTCTGCATCACGGGCCTCAGCAGTTTCTGAGCTGAGTCCTCGGGAGCGAAGCCCAGCTCTCAAA AGCCCCCTCCAGTCTGTGGTGGTGAGGCGGCGGTCACCCCGTCCTAGCCCCGTGCCAAAACCTAGTCCTC CACTTTCCAGCACATCCCAGATGGGCTCAACTCTGCCGAGTGGTGCCGGGTATCAGTCTGGGACACACCA AGGTCAGTTCGACCATGGTTCTGGGTCCCTGAGTCCATCCAAAAAGAGCCCTGTGGGTAAGAGTCCACCA TCCACTGGCTCCACATATGGCTCATCTCAGAAGGAGGAGAGTGCTGCTTCAGGAGGAGCAGCCTATACAA AGAGGTATCTAGAAGAGCAGAAGACAGAGAATGGAAAAGATAAGGAACAGAAACAAACAAATACCGATAA AGAAAAAATAAAAGAGAAAGGGAGCTTCTCTGACACAGGCTTGGGTGATGGAAAAATGAAATCTGATTCT TTTGCTCCCAAAACTGATTCTGAGAAGCCTTTTCGGGGCAGTCAGTCTCCCAAAAGGTATAAGCTCCGAG ATGACTTTGAGAAGAAGATGGCTGACTTCCACAAGGAGGAGATGGATGATCAAGATAAGGACAAAGCTAA GGGAAGAAAGGAATCTGAGTTTGATGATGAACCCAAATTTATGTCTAAAGTCATAGGTGCAAACAAAAAC CAGGAGGAGGAGAAGTCAGGCAAATGGGAGGGCCTGGTATATGCACCTCCAGGGAAGGAAAAGCAGAGAA AAACAGAGGAGCTGGAGGAGGAGTCTTTCCCAGAGAGATCCAAAAAGGAAGATCGGGGCAAGAGAAGCGA AGGTGGGCACAGGGGCTTTGTGCCTGAGAAGAATTTCCGAGTGACTGCTTATAAAGCAGTCCAGGAGAAA AGCTCATCACCTCCCCCAAGAAAGACCTCTGAGAGCCGAGACAAGCTGGGAGCGAAAGGAGATTTTCCCA CAGGAAAGTCTTCCTTTTCCATTACTCGAGAGGCACAGGTCAATGTCCGGATGGACTCTTTTGATGAGGA CCTCGCACGACCCAGTGGCTTATTGGCTCAGGAACGCAAGCTTTGCCGAGATCTAGTCCATAGCAACAAA AAGGAACAGGAGTTTCGTTCCATTTTCCAGCACATACAATCAGCTCAGTCTCAGCGTAGCCCCTCAGAAC TGTTTGCCCAACATATAGTGACCATTGTTCACCATGTTAAAGAGCATCACTTTGGGTCCTCAGGAATGAC ATTACATGAACGCTTTACTAAATACCTAAAGAGAGGAACTGAGCAGGAGGCAGCCAAAAACAAGAAAAGC C C AG AG A AC AC AGG AG AA AG AC AT T T C C C C C AG AC AT T C AG AAAAC AT GG T TTGGCTCAT GAT G AAA TGAAAAGTCCCCGGGAACCTGGCTACAAGGCTGAGGGAAAATACAAAGATGATCCTGTTGATCTCCGCCT T G AT AT T G AAC G T C G T AAAAAAC AT AAGG AG AG AG AT C T TAAAC G AGG T AAAT C GAG AG AAT C AG T GG AT TCCCGAGACTCCAGTCACTCAAGGGAAAGGTCAGCTGAAAAAACAGAGAAAACTCATAAAGGATCAAAGA AAC AG AAG AAGC AT C GG AG AGC AAG AG AC AGG T C C AG AT CCTCCTCCTCTTCCTCCCAGTCATCTCACTC CTACAAAGCAGAAGAGTACACTGAAGAGACAGAGGAAAGAGAGGAGAGCACCACGGGCTTTGACAAATCA AGACTGGGGACCAAAGACTTTGTGGGTCCAAGTGAAAGAGGAGGTGGCAGAGCTCGAGGAACCTTTCAGT TTCGAGCCAGAGGAAGAGGCTGGGGCAGAGGCAACTACTCTGGGAACAATAACAACAACAGCAACAACGA TTTTCAAAAAAGAAACCGGGAAGAGGAGTGGGACCCAGAGTACACACCCAAAAGCAAGAAGTATTACTTG CATGATGACCGTGAAGGCGAAGGCAGTGACAAGTGGGTGAGCCGGGGCCGGGGCCGAGGAGCCTTTCCTC GGGGTCGGGGCCGGTTCATGTTCCGGAAATCAAGTACCAGCCCCAAGTGGGCCCATGACAAGTTCAGTGG GGAGGAAGGGGAGATTGAAGACGACGAGAGTGGGACAGAGAACCGAGAAGAGAAGGACAATATACAGCCC ACAACCGAGTAGGGGCCACCCTTGACGGGATTCCTGCCCAGGGGAGAGAGGCGCTGGGAAGATGGCTGGT GAGGAGCTTAACAGAGGAACCTCAAGAAGATTCTGAAAATCCTACCCCCACCCCCCACCAGCCGCACAGA TTGTACTACCGCGAGAGGCATCCCTGGCGCTGTCTCCCACTGGACAGAGGAGGCTGGCCATGGGGCCCAG GGGTCAGGCCCAGCTTTTGAGCAGAATACAACGCATTGGGCTTTAGCTGTTTTTCTCATTTGTTGGTGTG TGGGGTGGGGGCAGGGGTAGGGCGGGAGAGCGATGCTTGGATTTTTGTTTCCTATTAGAAACCAACAGTT TTGTTCTAATTTCATTTCATTTGGAGCTAAGATGACTAATTTGATGATTTTCGATCTCTTTTCCCCTGTC CTGATTTTAAAAGCCCCCTCCTTTTTTTTTTTTTTTTTCTTTTTTTAGGCATATGTAGTAATATTAGAAA C AT T T AAT T T GGG AAAC T T T G AT T C T T G AAAG AG AAAAC AAAAGC AT G T G AAT AAAC T T T G AAG T G T T C A CCTCAGTTTGGGACCAAACTGCTTGGATCTTTGTAAAAACCGGTTTTGTATGTCAAGGAGGAGTTTAAGG CCTTTCCGACCACCTTGTGTTCCCCTTTTCTGCGCAGCCATGTATCACGTGGAGTTGCTCCTTACCACAC CTCACGTGCCCCTGAGCCCTATTTCCTGATTTCTTCTGGGCTGGACTTCCCCGTTCTCCACCAGCAGCTC CAGTATCCCAAACTTTCTAGTCCTGCTGATCCTCCCAGCAACGGGGTGGAAACTGGAGGGCAGTGTCTGG TCTGTTTTCTAAGAAACTTATGAATTCTATTATCTTTACAAATATGAGAAAATTTTTTCAATATTTTTTA TTAATCTTTTTATAAAATGAAAAGAAACTCCTATGATCGATTAAGGAAGGTGGTTATGGCTGGGTGGTTC AGGGGTTTTTTTGGGTTTCTTTTTTTTTTTCTTTGTCTTTTTAACCTTAAGCTGTTTAAGTTGAAGCATT CTCAGATGTTTGGGGGGAAACATCCTCTTAAAATGGGTCCTTGTGCTTGCCTTCTGGGGAGGCGGTCCTG AGCAGGTGAATCATAAGGCATTTATGCATATGTTATATGCGGACTGCACCCACCTCTCCCCCCCAGCCTT TGCCTCTTGCGTTGTTGTGCTGCTTTCCCCTTACTTTGCTACATTTCTATAGTTAAGTTGGTTTTACTTG AATGATTCATGTTTAGGGGGAAAATGAAAATCTCCCTTAAAATTTGTTTCAACTCCTCCTGCAAATAAAA TAAATGAAGTGGCAGATGTAAAAAAAAAAAAAAAAAA
Amino Acid Sequence
GenBank: AAI12331.1
SEQ ID NO: 24
MSKTNKSKSGSRSSRSRSASRSRSRSFSKSRSRSRSLSRSRKRRLSSRSRSRSYSPAHNRERNHPRVYQN RDFRGHNRGYRRPYYFRGRNRGFYPWGQYNRGGYGNYRSNWQNYRQAYSPRRGRSRSRSPKRRSPSPRSR SHSRNSDKSSSDRSRRSSSSRSSSNHSRVESSKRKSAKEKKSSSKDSRPSQAAGDNQGDEAKEQTFSGGT SQDTKASESSKPWPDATYGTGSASRASAVSELSPRERSPALKSPLQSVWRRRSPRPSPVPKPSPPLSST SQMGSTLPSGAGYQSGTHQGQFDHGSGSLSPSKKSPVGKSPPSTGSTYGSSQKEESAASGGAAYTKRYLE EQKTENGKDKEQKQTNTDKEKIKEKGSFSDTGLGDGKMKSDSFAPKTDSEKPFRGSQSPKRYKLRDDFEK KMADFHKEEMDDQDKDKAKGRKESEFDDEPKFMSKVIGANKNQEEEKSGKWEGLVYAPPGKEKQRKTEEL EEESFPERSKKEDRGKRSEGGHRGFVPEKNFRVTAYKAVQEKSSSPPPRKTSESRDKLGAKGDFPTGKSS FSITREAQVNVRMDSFDEDLARPSGLLAQERKLCRDLVHSNKKEQEFRSIFQHIQSAQSQRSPSELFAQH IVTIVHHVKEHHFGSSGMTLHERFTKYLKRGTEQEAAKNKKSPEIHRRIDISPSTFRKHGLAHDEMKSPR EPGYKAEGKYKDDPVDLRLDIERRKKHKERDLKRGKSRESVDSRDSSHSRERSAEKTEKTHKGSKKQKKH RRARDRSRSSSSSSQSSHSYKAEEYTEETEEREESTTGFDKSRLGTKDFVGPSERGGGRARGTFQFRARG RGWGRGNYSGNNNNNSNNDFQKRNREEEWDPEYTPKSKKYYLHDDREGEGSDKWVSRGRGRGAFPRGRGR FMFRKSSTSPKWAHDKFSGEEGEIEDDESGTENREEKDNIQPTTE Biomarker: Nestin
mRNA Sequence
NCBI Reference Sequence: NM_006617.1
SEQ ID NO: 25
GCTACTCCCACCCCGCCCCGCCCCGTCATTGTCCCCGTCGGTCTCTTTTCTCTTCCGTCCTAAAAGCTCT GCGAGCCGCTCCCTTCTCCCGGTGCCCCGCGTCTGTCCATCCTCAGTGGGTCAGACGAGCAGGATGGAGG GCTGCATGGGGGAGGAGTCGTTTCAGATGTGGGAGCTCAATCGGCGCCTGGAGGCCTACCTGGCCCGGGT CAAGGCGCTGGAGGAGCAGAATGAGCTGCTCAGCGCGGAGCTCGGGGGGCTCCGGGCACAATCCGCGGAC ACCTCCTGGCGGGCGCATGCCGACGACGAGCTGGCGGCCCTGCGGGCCCTCGTTGACCAACGCTGGCGGG AGAAGCACGCGGCCGAGGTGGCGCGCGACAACCTGGCTGAAGAGCTGGAGGGCGTGGCAGGCCGATGCCA GCAGCTGCGGCTGGCCCGGGAGCGGACGACGGAGGAGGTAGCCCGCAACCGGCGCGCCGTCGAGGCAGAG AAATGCGCCCGGGCCTGGCTGAGTAGCCAGGTGGCAGAGCTGGAGCGCGAGCTAGAGGCTCTACGCGTGG CGCACGAGGAGGAGCGCGTCGGCCTGAACGCGCAGGCTGCCTGTGCCCCCCGCTGCCCCGCGCCGCCCCG CGGGCCTCCCGCGCCGGCCCCGGAGGTAGAGGAGCTGGCAAGGCGACTGGGCGAGGCGTGGCGCGGGGCA GTGCGCGGCTACCAGGAGCGCGTGGCACACATGGAGACGTCGCTGGGCCAGGCCCGCGAGCGGCTGGGCC GGGCGGTGCAGGGTGCCCGCGAGGGCCGCCTGGAGCTGCAGCAGCTCCAGGCTGAGCGCGGAGGCCTCCT GGAGCGCAGGGCAGCGTTGGAACAGAGGTTGGAGGGCCGCTGGCAGGAGCGGCTGCGGGCTACTGAAAAG TTCCAGCTGGCTGTGGAGGCCCTGGAGCAGGAGAAACAGGGCCTACAGAGCCAGATCGCTCAGGTCCTGG AAGGTCGGCAGCAGCTGGCGCACCTCAAGATGTCCCTCAGCCTGGAGGTGGCCACGTACAGGACCCTCCT GGAGGCTGAGAACTCCCGGCTGCAAACACCTGGCGGTGGCTCCAAGACTTCCCTCAGCTTTCAGGACCCC AAGCTGGAGCTGCAATTCCCTAGGACCCCAGAGGGCCGGCGTCTTGGATCTTTGCTCCCAGTCCTGAGCC CAACTTCCCTCCCCTCACCCTTGCCTGCTACCCTTGAGACACCTGTGCCAGCCTTTCTTAAGAACCAAGA ATTCCTCCAGGCCCGTACCCCTACCTTGGCCAGCACCCCCATCCCCCCCACACCTCAGGCACCCTCTCCT GCTGTAGATGCAGAGATCAGAGCCCAGGATGCTCCTCTCTCTCTGCTCCAGACACAGGGTGGGAGGAAAC AGGCTCCAGAGCCCCTGCGGGCTGAAGCCAGGGTGGCCATTCCTGCCAGCGTCCTGCCTGGACCAGAGGA GCCTGGGGGCCAGCGGCAAGAGGCCAGTACAGGCCAGTCCCCAGAGGACCATGCCTCCTTGGCACCACCC CTCAGCCCTGACCACTCCAGTTTAGAGGCTAAGGATGGAGAATCCGGTGGGTCTAGAGTGTTCAGCATAT GCCGAGGGGAAGGTGAAGGGCAAATCTGGGGGTTGGTAGAGAAAGAAACAGCCATAGAGGGCAAAGTGGT AAGCAGCTTGCAGCAGGAAATATGGGAAGAAGAGGATCTAAACAGGAAGGAAATCCAGGACTCCCAGGTT C C T T T G G AAAAAG AAAC C C T G AAG TCTCTGGGAGAGGAGATT C AAG AG T C AC T G AAG AC T C T G G AAAAC C AG AGC C AT GAG AC AC T AG AAAGGG AG AAT C AAG AAT G T C C G AGG T C T T TAG AAG AAG AC T TAG AAAC AC T AAAAAGTCTAGAAAAGGAAAATAAAGAGCTATTAAAGGATGTGGAGGTAGTGAGACCTCTAGAAAAAGAG GCTGTAGGCCAACTTAAGCCTACAGGAAAAGAGGACACACAGACATTGCAATCCCTGCAAAAGGAGAATC AAG AAC T AAT G AAAT C T C T T G AAGG T AAT C TAG AG AC AT TTTTATTTC C AGG AAC GG AAAAT C AAG AAT T AGTAAGTTCTCTGCAAGAGAACTTAGAGTCATTGACAGCTCTGGAAAAGGAGAATCAAGAGCCACTGAGA TCTCCAGAAGTAGGGGATGAGGAGGCACTGAGACCTCTGACAAAGGAGAATCAGGAACCCCTGAGGTCTC T T G AAG AT G AG AAC AAAG AGGC C T T TAG AT C T C TAG AAAAAG AG AAC C AGG AGC C AC T G AAG AC T C TAG A AG AAG AGG AC C AG AG T AT T G T GAG AC C T C TAG AAAC AG AG AAT C AC AAAT C AC T G AGG T C T T TAG AAG AA CAGGACCAAGAGACATTGAGAACTCTTGAAAAAGAGACTCAACAGCGACGGAGGTCTCTAGGGGAACAGG AT C AG AT G AC AT T AAG AC C C C C AG AAAAAG T GG AT C TAG AAC C AC T G AAG T C T C T T G AC C AGG AG AT AGC TAG AC C T C T T G AAAAT GAG AAT C AAG AG T T C T T AAAG T C AC T C AAAG AAG AG AGC G TAG AGGC AG TAAAA T C T T TAG AAAC AG AG AT C C TAG AAT C AC T G AAG T C T GC GGG AC AAG AG AAC C T GG AAAC AC T G AAAT C T C CAGAAACTCAAGCACCACTGTGGACTCCAGAAGAAATAAATCAGGGGGCAATGAATCCTCTAGAAAAGGA AAT T C AAG AAC C AC T GG AG T C T G T GG AAG T G AAC C AAG AG AC AT T C AG AC T C C T GG AAG AGG AG AAT C AG GAATCATTGAGATCTCTGGGAGCATGGAACCTGGAGAATTTGAGATCTCCAGAGGAGGTAGACAAGGAAA GTCAAAGGAATCTGGAAGAGGAAGAGAACCTGGGAAAGGGAGAGTACCAAGAGTCACTGAGGTCTCTGGA GGAGGAGGGACAGGAGCTGCCGCAGTCTGCAGATGTGCAGAGGTGGGAAGATACGGTGGAGAAGGACCAA GAACTGGCTCAGGAAAGCCCTCCTGGGATGGCTGGAGTGGAAAATGAGGATGAGGCAGAGCTGAATCTGA GGGAGCAGGATGGCTTCACTGGGAAGGAGGAGGTGGTAGAGCAGGGAGAGCTGAATGCCACAGAGGAGGT CTGGATCCCAGGCGAGGGGCACCCAGAGAGCCCTGAGCCCAAAGAGCAGAGAGGCCTGGTTGAGGGAGCC AGTGTGAAGGGAGGGGCTGAGGGCCTCCAGGACCCTGAAGGGCAATCACAACAGGTGGGGGCCCCAGGCC TCCAGGCTCCCCAGGGGCTGCCAGAGGCGATAGAGCCCCTGGTGGAAGATGATGTGGCCCCAGGGGGTGA CCAAGCCTCCCCAGAGGTCATGTTGGGGTCAGAGCCTGCCATGGGTGAGTCTGCTGCGGGAGCTGAGCCA GGCCCGGGGCAGGGGGTGGGAGGGCTGGGGGACCCAGGCCATCTGACCAGGGAAGAGGTGATGGAACCAC CCCTGGAAGAGGAGAGTTTGGAGGCAAAGAGGGTTCAGGGCTTGGAAGGGCCTAGAAAGGACCTAGAGGA GGCAGGTGGTCTGGGGACAGAGTTCTCCGAGCTGCCTGGGAAGAGCAGAGACCCTTGGGAGCCTCCCAGG GAGGGTAGGGAGGAGTCAGAGGCTGAGGCCCCCAGGGGAGCAGAGGAGGCGTTCCCTGCTGAGACCCTGG GCCACACTGGAAGTGATGCCCCTTCACCTTGGCCTCTGGGGTCAGAGGAAGCTGAGGAGGATGTACCACC AGTGCTGGTCTCCCCCAGCCCAACGTACACCCCGATCCTGGAAGATGCCCCTGGGCCTCAGCCTCAGGCT GAAGGGAGTCAGGAGGCTAGCTGGGGGGTGCAGGGGAGGGCTGAAGCCCTGGGGAAAGTAGAGAGCGAGC AGGAGGAGTTGGGTTCTGGGGAGATCCCCGAGGGCCCCCAGGAGGAAGGGGAGGAGAGCAGAGAAGAGAG CGAGGAGGATGAGCTCGGGGAGACCCTTCCAGACTCCACTCCCCTGGGCTTCTACCTCAGGTCCCCCACC TCCCCCAGGTGGGACCCCACTGGAGAGCAGAGGCCACCCCCTCAAGGGGAGACTGGAAAGGAGGGCTGGG ATCCTGCTGTCCTGGCTTCCGAGGGCCTTGAGGCCCCACCCTCAGAAAAGGAGGAGGGGGAGGAGGGAGA AGAGGAGTGTGGCCGTGACTCTGACCTGTCAGAAGAATTTGAGGACCTGGGGACTGAGGCACCTTTTCTT CCTGGGGTCCCTGGGGAGGTGGCAGAACCTCTGGGCCAGGTGCCCCAGCTGCTACTGGATCCTGCAGCCT GGGATCGAGATGGGGAGTCCGATGGGTTTGCAGATGAGGAAGAAAGTGGGGAGGAGGGAGAGGAGGATCA GGAGGAGGGGAGGGAGCCAGGGGCTGGGCGGTGGGGGCCAGGGTCTTCTGTTGGCAGCCTCCAGGCCCTG AGTAGCTCCCAGAGAGGGGAATTCCTGGAGTCTGATTCTGTGAGTGTCAGTGTCCCCTGGGATGACAGCT TGAGGGGTGCAGTGGCTGGTGCCCCCAAGACTGCCCTGGAAACGGAGTCCCAGGACAGTGCTGAGCCTTC TGGCTCAGAGGAAGAGTCTGACCCTGTTTCCTTGGAGAGGGAGGACAAAGTCCCTGGCCCTCTAGAGATC CCCAGTGGGATGGAGGATGCAGGCCCAGGGGCAGACATCATTGGTGTTAATGGCCAGGGTCCCAACTTGG AGGGGAAGTCACAGCATGTGAATGGGGGAGTGATGAACGGGCTGGAGCAGTCTGAGGAAGTGGGGCAAGG AATGCCGCTAGTCTCTGAGGGAGACCGAGGGAGCCCCTTTCAGGAGGAGGAGGGGAGTGCTCTGAAGACC TCTTGGGCAGGGGCTCCTGTTCACCTGGGCCAGGGTCAGTTCCTGAAGTTCACTCAGAGGGAAGGAGATA GAGAGTCCTGGTCCTCAGGGGAGGACTAGGAAAAGACCATCTGCCCGGCACTGGGGACTTAGGGGTGCGG GGAGGGGAAGGACGCCTCCAAGCCCGCTCCCTGCTCAGGAGCAGCACTCTTAACTTACGATCTCTTGACA TATGGTTTCTGGCTGAGAGGCCTGGCCCGCTAAGGTGAAAAGGGGTGTGGCAAAGGAGCCTACTCCAAGA ATGGAGGCTGTAGGAATATAACCTCCCACCCTGCAAAGGGAATCTCTTGCCTGCTCCATCTCATAGGCTA AGTCAGCTGAATCCCGATAGTACTAGGTCCCCTTCCCTCCGCATCCCGTCAGCTGGAAAAGGCCTGTGGC CCAGAGGCTTCTCCAAAGGGAGGGTGACATGCTGGCTTTTGTGCCCAAGCTCACCAGCCCTGCGCCACCT CACTGCAGTAGTGCACCATCTCACTGCAGTAGCACGCCCTCCTGGGCCGTCTGGCCTGTGGCTAATGGAG GTGACGGCACTCCCATGTGCTGACTCCCCCCATCCCTGCCACGCTGTGGCCCTGCCTGGCTAGTCCCTGC C GAA AAAG AA GCC CCGC CAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA
Amino Acid Sequence
NCBI Reference Sequence: NP_006608.1
SEQ ID NO: 26
MEGCMGEESFQMWELNRRLEAYLARVKALEEQNELLSAELGGLRAQSADTSWRAHADDELAALRALVDQR WREKHAAEVARDNLAEELEGVAGRCQQLRLARERTTEEVARNRRAVEAEKCARAWLSSQVAELERELEAL RVAHEEERVGLNAQAACAPRCPAPPRGPPAPAPEVEELARRLGEAWRGAVRGYQERVAHME SLGQARER LGRAVQGAREGRLELQQLQAERGGLLERRAALEQRLEGRWQERLRATEKFQLAVEALEQEKQGLQSQIAQ VLEGRQQLAHLKMSLSLEVATYRTLLEAENSRLQTPGGGSKTSLSFQDPKLELQFPRTPEGRRLGSLLPV LSPTSLPSPLPATLETPVPAFLKNQEFLQARTPTLASTPIPPTPQAPSPAVDAEIRAQDAPLSLLQTQGG RKQAPEPLRAEARVAIPASVLPGPEEPGGQRQEASTGQSPEDHASLAPPLSPDHSSLEAKDGESGGSRVF SICRGEGEGQIWGLVEKETAIEGKWSSLQQEIWEEEDLNRKEIQDSQVPLEKETLKSLGEEIQESLKTL ENQSHETLERENQECPRSLEEDLETLKSLEKENKELLKDVEWRPLEKEAVGQLKPTGKEDTQTLQSLQK ENQELMKSLEGNLETFLFPGTENQELVSSLQENLESLTALEKENQEPLRSPEVGDEEALRPLTKENQEPL RSLEDENKEAFRSLEKENQEPLKTLEEEDQSIVRPLETENHKSLRSLEEQDQETLRTLEKETQQRRRSLG EQDQMTLRPPEKVDLEPLKSLDQEIARPLENENQEFLKSLKEESVEAVKSLETEILESLKSAGQENLETL KSPETQAPLWTPEEINQGAMNPLEKEIQEPLESVEVNQETFRLLEEENQESLRSLGAWNLENLRSPEEVD KESQRNLEEEENLGKGEYQESLRSLEEEGQELPQSADVQRWEDTVEKDQELAQESPPGMAGVENEDEAEL NLREQDGFTGKEEWEQGELNATEEVWIPGEGHPESPEPKEQRGLVEGASVKGGAEGLQDPEGQSQQVGA PGLQAPQGLPEAIEPLVEDDVAPGGDQASPEVMLGSEPAMGESAAGAEPGPGQGVGGLGDPGHLTREEVM EPPLEEESLEAKRVQGLEGPRKDLEEAGGLGTEFSELPGKSRDPWEPPREGREESEAEAPRGAEEAFPAE TLGHTGSDAPSPWPLGSEEAEEDVPPVLVSPSPTYTPILEDAPGPQPQAEGSQEASWGVQGRAEALGKVE SEQEELGSGEIPEGPQEEGEESREESEEDELGETLPDSTPLGFYLRSPTSPRWDPTGEQRPPPQGETGKE GWDPAVLASEGLEAPPSEKEEGEEGEEECGRDSDLSEEFEDLGTEAPFLPGVPGEVAEPLGQVPQLLLDP AAWDRDGESDGFADEEESGEEGEEDQEEGREPGAGRWGPGSSVGSLQALSSSQRGEFLESDSVSVSVPWD DSLRGAVAGAPKTALETESQDSAEPSGSEEESDPVSLEREDKVPGPLEIPSGMEDAGPGADI IGVNGQGP NLEGKSQHVNGGVMNGLEQSEEVGQGMPLVSEGDRGSPFQEEEGSALKTSWAGAPVHLGQGQFLKFTQRE GDRESWSSGED Biomarker: Sorbin and SH3 domain-containing protein 2 (SORB-2) mRNA Sequence
NCBI Reference Sequence: NM_001270771.1
SEQ ID NO: 27
ATGCATTAAACTCCAGGTATTTTCAGGGAGGAATCATGAAGCCGCTCCTGTTAAATCATATCCATTTTAT TACGTCTCTCCTTGTGGATTACGTAGAACTTTGGAAAATTAAACATTAGACATCTTCATAGAAAATCATG TCACTGATTTTTAAAGCCAGCTCAATTTAACCTGCCACTGATTCCCTAACCAGCTCTTCTCTGACTGTAT CCCGAGGGAAAATGTATTGAACGCAGCAGAAGGTTAAGGCAGAGGAGTTCGGCTAGACCGGGTAGCAGCC AATCAGATGGGGAGGGTCCAAGGATCTGATGCCTCCAGGCGAGCAGCGCCAAGCTGAGCTGGTTAGTCTC CTTTTATTGTGGATGAACACCACGTACATGCCGTAAATGCTCCCTAGGCTGGCTGCGGCTACAAGGCAGA CCCGGGAGCTGAGAATAGAGGAATACCTGCCCAAGCAACTCATTCTGCACAGCAGCCGACAAGACAGCGA TTTTCCTGTGCTGGAATGTGCCCATCCAAAGCCAACCAATTTGTTGGAAAATCAGTCAAAATCTAATTTT GATGTTCTGCAAGTTCCCTGGGGGCCATGAAGCACCTGACACTTGGATTGGACTTAATCTTAAACCTCTG GAGTTCAAGACCTTTTAAAAAGGGCTAAATAAACAATCTCTACATGTAAAAGGCCACTGACTCCTACTTC CTCTGTATAGAGCAACTGTTGAACTCAGCTGCCTGTAGGAAAACTGAAGACTTTAATAACAAACTCTCCA AGGTGAAAATGAACACAGATAGCGGTGGGTGTGCTCGCAAACGTGCCGCCATGTCTGTTACGCTAACATC TGTGAAGAGAGTGCAAAGTTCTCCAAACCTATTGGCTGCAGGGCGTGATTCTCAGTCACCAGACTCAGCT TGGAGATCTTACAATGATGGCAATCAGGAGACACTGAACGGAGATGCTACATATTCCTCTCTTGCAGCAA AAGGTTT AGAAGCGTTCGACCAAACCTACAAGATAAAAGATCACCAACTCAGAGCCAGATAACAGTGAA TGGAAACTCAGGAGGTGCCGTGAGTCCCATGAGTTACTATCAGAGGCCGTTTTCCCCCTCGGCATATTCT CTCCCAGCCTCACTCAACTCCAGCATTGTCATGCAGCACGGCACATCCCTCGATTCCACAGACACATATC CCCAGCATGCGCAGTCTCTGGATGGCACCACCAGCAGCTCTATCCCCCTGTACCGATCCTCAGAGGAAGA GAAGAGAGTGACAGTCATCAAAGCCCCGCATTACCCAGGGATCGGGCCCGTGGATGAATCCGGAATCCCC ACAGCAATTAGAACGACAGTCGACCGGCCCAAGGACTGGTACAAGACGATGTTTAAGCAAATTCACATGG TGCACAAGCCGGATGATGACACAGACATGTATAATACTCCTTATACATACAATGCAGGTCTGTACAACCC ACCCTACAGTGCTCAGTCACACCCTGCTGCAAAGACCCAAACCTACAGACCTCTTTCCAAAAGCCACTCC GACAACAGCCCCAATGCCTTTAAGGATGCGTCCTCCCCAGTGCCTCCCCCACATGTTCCACCTCCAGTCC CGCCGCTTCGACCAAGAGATCGGTCTTCAACAGAAAAGCATGACTGGGATCCTCCAGACAGAAAAGTGGA CACAAGAAAATTTCGGTCTGAGCCAAGGAGTATTTTTGAATATGAACCTGGCAAGTCATCAATTCTTCAG CAT G AAAG AC CAGCCTCCTTGTATCAG CCTC T AT AG AC AG AAGC C T GG AAAG AC C C AT GAG T T C T GC AA GCATGGCCAGTGACTTCAGGAAGCGGAGGAAGAGCGAGCCTGCAGTGGGTCCACCACGGGGCTTGGGAGA TCAAAGTGCGAGCAGGACTAGCCCAGGCCGAGTGGACCTCCCAGGATCAAGCACCACTCTTACAAAGTCT TTCACTAGCTCTTCTCCTTCTTCCCCATCAAGAGCAAAAGGTGGGGATGATAGCAAAATATGTCCATCCC TTTGCAGTTACTCAGGGCTCAACGGCAACCCCTCCAGTGAGTTAGATTACTGTAGCACTTACAGACAGCA CTTGGATGTCCCTCGGGACTCACCAAGAGCCATTAGTTTCAAGAACGGCTGGCAAATGGCCCGGCAAAAC GCAGAAATCTGGAGCAGCACGGAAGAAACCGTCTCTCCCAAAATCAAATCCCGGAGCTGTGACGATCTCC TAAACGACGACTGCGATAGCTTCCCGGACCCGAAAGTCAAGTCGGAAAGCATGGGCTCCCTGTTATGTGA GGAGGACTCCAAGGAGAGCTGCCCCATGGCGTGGGGGTCCCCCTACGTCCCGGAGGTTCGCAGTAACGGC AGATCAAGGATCAGACACAGGTCAGCCCGCAACGCCCCGGGGTTCCTGAAGATGTACAAGAAAATGCACC GCATCAACCGCAAGGACCTGATGAACTCCGAGGTGATCTGCTCCGTGAAGTCCAGGATTCTGCAGTACGA GAGCGAGCAGCAGCACAAGGACCTGCTGCGCGCCTGGAGCCAGTGCTCCACGGAGGAGGTGCCCCGGGAC ATGGTGCCCACACGCATCTCCGAATTCGAAAAGCTGATCCAGAAGTCCAAATCCATGCCAAATTTAGGGG ATGACATGCTGTCTCCCGTAACCCTAGAACCACCGCAAAATGGCCTATGTCCCAAGAGGCGGTTTTCCAT TGAGTATTTGCTGGAGGAAGAAAATCAAAGCGGCCCCCCCGCTCGGGGCCGGCGAGGCTGCCAGTCTAAC GCCCTGGTGCCCATTCACATTGAAGTCACCAGCGATGAGCAGCCCCGAGCACACGTGGAGTTTTCCGACA GCGACCAGGACGGGGTTGTGTCCGACCACAGTGACTACATTCACCTAGAGGGGTCATCCTTCTGCAGTGA AAGTGACTTTGATCACTTTTCCTTCACATCCTCCGAAAGCTTTTACGGATCCAGCCACCACCACCACCAT CACCACCACCACCACCACCGCCACCTCATCAGCTCCTGCAAAGGCAGGTGCCCGGCCTCCTACACTCGAT TTACCACAATGTTAAAACACGAAAGAGCCAGACACGAAAACACCGAGGAGCCCAGAAGGCAAGAAATGGA CCCTGGCCTTTCTAAACTTGCTTTTCTAGTCAGTCCTGTGCCTTTCCGGAGGAAAAAAAATTCGGCTCCT AAGAAACAGACTGAAAAGGCAAAATGTAAAGCATCTGTGTTTGAGGCTCTGGACTCTGCCCTTAAAGACA TCTGTGACCAAATTAAAGCTGAAAAGAAAAGGGGGAGCTTGCCGGACAACAGCATCCTGCACCGCCTCAT CAGTGAGCTGCTGCCAGATGTTCCCGAGAGGAACTCATCCCTGAGAGCGCTGAGGAGGAGCCCCCTGCAC CAGCCTCTCCACCCACTGCCTCCCGATGGTGCTATTCATTGTCCACCCTACCAGAATGACTGCGGGAGAA TGCCCCGCAGTGCCTCTTTCCAAGACGTGGACACAGCCAACAGCAGCTGCCACCACCAAGACCGTGGCGG TGCACTCCAAGACCGTGAGTCCCCTAGAAGTTACTCATCCACTTTGACTGACATGGGGAGAAGTGCACCA AGGGAAAGAAGAGGAACTCCAGAAAAAGAGAAATTGCCTGCAAAAGCTGTTTATGATTTTAAGGCTCAGA CATCTAAGGAGTTGTCATTTAAGAAAGGAGATACTGTCTACATCCTCAGGAAAATTGATCAAAATTGGTA TGAGGGAGAACACCACGGGAGAGTGGGCATCTTCCCGATCTCATACGTAGAGAAACTCACACCTCCTGAG AAAGCACAGCCTGCAAGACCACCTCCGCCAGCCCAGCCCGGAGAAATCGGAGAAGCTATAGCCAAATACA ACTTCAACGCAGACACAAATGTGGAGCTGTCACTGAGAAAGGGAGATAGAGTTATTCTTCTTAAAAGAGT TGATCAAAACTGGTATGAAGGTAAAATCCCAGGAACCAACAGACAAGGCATCTTCCCTGTTTCCTATGTG GAGGTCGTCAAGAAGAACACAAAAGGTGCTGAGGACTACCCTGACCCTCCAATACCCCACAGCTATTCTA GTGATAGGATTCACAGCTTGAGCTCAAATAAGCCACAGCGTCCTGTGTTTACTCATGAAAATATTCAAGG TGGGGGGGAACCGTTTCAGGCTCTGTATAACTATACTCCCAGGAATGAAGATGAGCTGGAGCTCAGAGAA AGTGATGTCATTGATGTCATGGAAAAGTGTGATGACGGCTGGTTTGTGGGGACCTCAAGAAGAACCAAAT TCTTTGGTACTTTCCCCGGAAACTACGTCAAGAGGCTGTGAATTGCGCTCCCTCCTTCTGTAGAGGCCGC CTGCCAGCCATGCACCTGCGTCAACGCGCCTGAAACACCCCGCGGGCCTCCCGTTGTCATGCCTTACGGT TTCCAATGCGCCGTCACCATCTCCACCTGCCACCAAACCACCAGCAGAGTAGCCGCCGCTGCTGTGAGCC TGGGGACGACATGGCAGGCTGGTCCCCCTCCGTGAAAGTGTGGATTCCTACTTCCTGCTCTAAGCTTTGA CACGTCAAAATGTGGGATCAGAAAGAAAAAAATCATGATATTTAAAAATGGTCAAATATTTGAGGCAAAA AAAAAAAAAAAAAGTGTCTCCAGGAGGCTGTCCAGCCTCGTGGCTCCATTTCAACATCTCCCCCCAGGCG ATGTTCTCCCCCAAGACGACCAGAAAATTGTTTATTGGGGAATGCTGTGGTTTGCATTTTCATATTCTTC GCTTGGCAGTGTGTATTCTTTTCACAAGTTTGCCTAGTGTCTTGGTTTACACAATATGACAACTGTAACT GTACTTTAGCTATTGTTTGCCTGCACATACATGTTGTAATATGCACAGTGATTACAACCTTTAAAGCAAG AGGAGGCGAGTTAATTTGGATGAGTGTGATTTTGCTGATTGAATGTGAGTTTTCAAATAGGAGTCTTTTT CTGCAATTTGTTTGCATTTTTTAGAAGTGCAAACAGTAAGTAAATAAAAGCCTTCGGTAATAATCATGAC AATACAAGAGGCTGAGCTAGGCTACAGGGGAAAACTATTGTGTTGTAAAGTTGCATCGCTATTTTATATT AAAATGTAATGATCAGCATCATGAACAATGAGCTCTTAGTGTTTTATTTATCTGAAAAAATGTAAGTAAA AGCAGTGTTAGTGAGAGGTGCAAAGAATTATTCTAACAGACACCTGAAAGTATCTGTAAGCAATACTAGT AGGTAAGATTTCACTGTGTGTACACATACACATTTGAGATTGTATGAGAACATATAATCCATATGATATG TTGTACATTTTATGGAAATGTAATAGAATCTCACACATTATTTTATAGAAATAGAGTACAGAAGCATCAC AAGTATTAAAGTTGGTTTTAGCAAGGATTAGGATTACTACAATTATTTTTACTATAATAATTATTTTTCT TCTATGGAACTCAGAATCCTGGCTACATTTGAGGACAGGAATATGTTGAGCCTGATTTTCCTGGTGTGTG TAAAATATTTCCTAGGAATAAATTAGGCACTTTTTAGAAGCAATGTTAAATCATTCAGGTTTTATTTTCT GCCCTGAAGCAGAAATTTAAAAAATGATATTGGGACCTGGAAGGTTTAATATGGTTCACAGTGCCTGAAT TACACCTGCTCCGAAAACTAGCTTTGTATTTCTTATGACTTTGCATAAGAACTGTTCATCTTTGGATCTT GAGCACCTTACAGATAAAACTTTTTATGGCATCTCTTTCATGGACAGTGATATTGATCTTTTCACAATGT ACGTAGCCTCTAGAATTTTGTACATATGTTTGCTCTTTTTTTGTACAGACTAGTTGTTGAGAAAACAGGG GCGTTTCCTAATTTGGTCTTTATCCTGCGACAACACTTTCAGCAGACTAGCCTCTCCTTATCTCACAGAT CACAAGCACCCCTAGATAGTGTGATTCTGTCAGATAGCATTTATGCAAAAATCTATGAAGTTAAAAGATC GTAGAAGCCAAATGAAATGTACATATCTACTGACTGATGACAAGGGAATTTCATTAGGAAGAAGGTAAAG AAACATCGTTGAGTAGCCTACCTTGATTTCTGTCAAGTTCATAACCAGCTTCATATTTTAAAGGCTTCAG GTTTGAAATTAAGTCAACTGCATGCAGCTTTGCTGATAAATGAATAATTCTCTTTGATGCCATTTATGAG AAAAGACTTCAATATCTGTTGCCTGTCATATTTAAGAAAAATTACTGTTTCTACTCTCTGTATCTGATTT TAAAAGAAAAAACTATTCATACCTGGCTTCCAGGTAATTGACTTTGAATTCTTACAAGCAAAGGTCATTG TGTTTTTCTTGAATAGACATCTAATAAATTTTGCTTGGAAGTATACTTCAGTTTTTCTTCTTGACATTTA TCTTTATAAAAATTGTGTATTTTATTCCAACTTGTTAAACTAAGAGAAAATGCA
Amino Acid Sequence
NCBI Reference Sequence: NP_001257700.1
SEQ ID NO: 28
MNTDSGGCARKRAAMSVTLTSVKRVQSSPNLLAAGRDSQSPDSAWRSYNDGNQETLNGDATYSSLAAKGF RSVRPNLQDKRSPTQSQITVNGNSGGAVSPMSYYQRPFSPSAYSLPASLNSSIVMQHGTSLDSTDTYPQH AQSLDGTTSSSIPLYRSSEEEKRVTVIKAPHYPGIGPVDESGIPTAIRTTVDRPKDWYKTMFKQIHMVHK PDDDTDMYNTPYTYNAGLYNPPYSAQSHPAAKTQTYRPLSKSHSDNSPNAFKDASSPVPPPHVPPPVPPL RPRDRSSTEKHDWDPPDRKVDTRKFRSEPRSIFEYEPGKSSILQHERPASLYQSSIDRSLERPMSSASMA SDFRKRRKSEPAVGPPRGLGDQSASRTSPGRVDLPGSSTTLTKSFTSSSPSSPSRAKGGDDSKICPSLCS YSGLNGNPSSELDYCSTYRQHLDVPRDSPRAISFKNGWQMARQNAEIWSSTEETVSPKIKSRSCDDLLND DCDSFPDPKVKSESMGSLLCEEDSKESCPMAWGSPYVPEVRSNGRSRIRHRSARNAPGFLKMYKKMHRIN RKDLMNSEVICSVKSRILQYESEQQHKDLLRAWSQCSTEEVPRDMVPTRISEFEKLIQKSKSMPNLGDDM LSPVTLEPPQNGLCPKRRFSIEYLLEEENQSGPPARGRRGCQSNALVPIHIEVTSDEQPRAHVEFSDSDQ DGWSDHSDYIHLEGSSFCSESDFDHFSF SSESFYGSSHHHHHHHHHHHRHLISSCKGRCPASYTRFTT MLKHERARHENTEEPRRQEMDPGLSKLAFLVSPVPFRRKKNSAPKKQTEKAKCKASVFEALDSALKDICD QIKAEKKRGSLPDNSILHRLISELLPDVPERNSSLRALRRSPLHQPLHPLPPDGAIHCPPYQNDCGRMPR SASFQDVDTANSSCHHQDRGGALQDRESPRSYSSTLTDMGRSAPRERRGTPEKEKLPAKAVYDFKAQTSK ELSFKKGDTVYILRKIDQNWYEGEHHGRVGIFPISYVEKLTPPEKAQPARPPPPAQPGEIGEAIAKYNFN ADTNVELSLRKGDRVILLKRVDQNWYEGKIPGTNRQGIFPVSYVEWKKNTKGAEDYPDPPIPHSYSSDR IHSLSSNKPQRPVFTHENIQGGGEPFQALYNYTPRNEDELELRESDVIDVMEKCDDGWFVGTSRRTKFFG TFPGNYVKRL
Biomarker: LIM domain only protein 7 (LMO-7)
mRNA Sequence
NCBI Reference Sequence: NM_005358.5
SEQ ID NO: 29
GGAAAGAAGTGGAATAATTAGGAACCTAGGGTGGGGTAGGGTAGCAGGACATTTCAAACATTAATGAGCA TATGAGATTCCAGGTCTTGTTAAAATGCAAATTCTGATTCAGCTGGTAGGTGAGGTCTGAGATTGTGCAT TTCTAACAAGCACTCAGATAATCTTAAGGCTGTTGGCCCCAGGGTCACACTTATAGTGATTTTCTAGAAC CCAGTTGGGGAAGTGAATCTTGGGCAGGAGAAATACACACCTCTTGCATTGAGTTTGGAGATCTCATCTG ATATAACTTTTTAAGAAAGAAAAATAATTTTCCAAATATCCAATTGATAAGCTTTCCCACTAAGTGGCTT TCCCACTAAGTGGCTGCGTTATGAAAATTGCTTCACTTTGAAACTTCTGGTCTTGGTAATATAGAATTTC TGTGTTCTCACAGTGCTTGATTGAGAATATGATATTGAGATTATGGCATAAAATATAGTGGCTGTACAAA AAAAAATACATTATTAGGATCTCTAACAATTATGTAAAAGTCATTGCTTCATGGGTAGAGCTCAAACTTT GGTGTGAGACCTGGTTTTATTCTTGGCACTTACTCTGAGTTGTCTTAGGCAAATTAATACCTTAAGCAAA AATATTCTCATGTACATTTTACATGAGAATTATAAATGAAGTACATAAAGTCCAGCAGTCACAAATGTTA TCTATTATTACCATCGTCCTAAGACTGCAATCAGCTATAGTGAAAGTAGTCTCAAAGATTGTTTCATAAA TCATCAGATTCACCTAATTTTCTAAAGAATTTAAATAAGGAGATGGAATGAATAGATTGCATTTTGTTTC CATGCACAGGGGAACTGTGCATATTTCTTCTGTGACTCGGAAATGGTTTAACTTTTAAAAATCCCAAAAT AGCTGAAGTTAGCAGACATGCAATTTACCAAGGATGATTGGAATTTTTATCTTTCCTGTAATAATACTAT ACCCAAGCACACTGCTCATGAGGAAAACATTTTTATGTGAATCTTTTACTCTTGGGGGCAAAGAATGCTG TTTTTCTTTTTGATAACTATGTTTATAGAATCTAAATCACCCTGAGCAATTATTTCAACATCTAAAGTTA TTATTACCATTCATGTTTCATTTATAGCTATTTGAATTTTGATGAATTTCAATATGGTGCTACAGTGATA GGGCAAGTGCAAATAAGTTCAATATATGGGTACGGTCTAAAGCTATTTTAATTTTTTTATTACAACTGCT ATGAAGAAAATTAGGATATGCCATATTTTCACGTTTTACAGTTGGATGTCCTATGATGTTCTCTTCCAGA GAACAGAGCTCGGAGCTCTGGAAATTTGGAGGCAACTGATATGTGCTCATGTCTGCATCTGTGTGGGTTG GCTGTATCTCAGGGACAGAGTCTGCAGCAAAAAAGATATAATTTTGAGGACTGAACAAAATTCAGGAAGG AC ATTCTCAT T AAGGC AG T AAC AG AG AAG AAT T T T G AAAC AAAAG AT T T T C G AGC C T C T C T AG AAAAT G GTGTTCTGCTGTGTGATTTGATTAATAAGCTTAAACCTGGCGTCATTAAGAAGATCAATAGACTGTCTAC ACCAATAGCAGGATTGGATAATATAAACGTTTTCTTGAAAGCTTGTGAACAGATTGGATTGAAAGAAGCC CAGCTTTTCCATCC T GG AG AT C T AC AGG AT T T AT C AAAT C GAG T C AC T G T C AAGC AAG AAG AG AC T G AC A GGAGAGTGAAAAATGTTTTGATAACATTGTACTGGCTGGGAAGAAAAGCACAAAGCAACCCGTACTATAA TGGTCCCCATCTTAATTTGAAAGCGTTTGAGAATCTTTTAGGACAAGCACTGACGAAGGCACTCGAAGAC TCCAGCTTCCTGAAAAGAAGTGGCAGGGACAGTGGCTACGGTGACATCTGGTGTCCTGAACGTGGAGAAT TTCTTGCTCCTCCAAGGCACCATAAGAGAGAAGATTCCTTTGAAAGCTTGGACTCTTTGGGCTCGAGGTC ATTGACAAGCTGCTCCTCTGATATCACGTTGAGAGGGGGGCGTGAAGGTTTTGAAAGTGACACAGATTCG GAATTTACATTTAAGATGCAGGATTATAATAAAGATGATATGTCGTATCGAAGGATTTCGGCTGTTGAGC C AAAG AC T GC G T T AC C C T T C AAT C G T T T T T T AC C C AAC AAAAG T AG AC AGC C AT C C T AT G T AC C AGC AC C TCTGAGAAAGAAAAAGCCAGACAAACATGAGGATAACAGAAGAAGTTGGGCAAGCCCGGTTTATACAGAA GCAGATGGAACATTTTCAAGACTCTTTCAAAAGATTTATGGTGAGAATGGGAGTAAGTCCATGAGTGATG TCAGCGCAGAAGATGTTCAAAACTTGCGTCAGCTGCGTTACGAGGAGATGCAGAAAATAAAATCACAATT AAAAGAACAAGATCAGAAATGGCAGGATGACCTTGCAAAATGGAAAGATCGTCGAAAAAGTTACACTTCA GATCTGCAGAAGAAAAAAGAAGAGAGAGAAGAAATTGAAAAGCAGGCACTTGAGAAGTCTAAGAGAAGCT CTAAGACGTTTAAGGAAATGCTGCAGGACAGGGAATCCCAAAATCAAAAGTCTACAGTTCCGTCAAGAAG GAGAATGTATTCTTTTGATGATGTGCTGGAGGAAGGAAAGCGACCCCCTACAATGACTGTGTCAGAAGCA AG T T AC C AG AG T GAG AG AG TAG AAG AG AAGGG AGC AAC TTATCCTT C AG AAAT T C C C AAAG AAG AT T C T A CCACTTTTGCAAAAAGAGAGGACCGTGTAACAACTGAAATTCAGCTTCCTTCTCAAAGTCCTGTGGAAGA ACAAAGCCCAGCCTCTTTGTCTTCTCTGCGTTCACGGAGCACACAAATGGAATCAACTCGTGTTTCAGCT TCTCTCCCCAGAAGTTACCGGAAAACTGATACAGTCAGGTTAACATCTGTGGTCACACCAAGACCCTTTG GCTCTCAGACAAGGGGAATCTCATCACTCCCCAGATCTTACACGATGGATGATGCTTGGAAGTATAATGG AGATGTTGAAGACATTAAGAGAACTCCAAACAATGTGGTCAGCACCCCTGCACCAAGCCCGGACGCAAGC CAACTGGCTTCAAGCTTATCTAGCCAGAAAGAGGTAGCAGCAACAGAAGAAGATGTGACAAGGCTGCCCT CTCCTACATCCCCCTTCTCATCTCTTTCCCAAGACCAGGCTGCCACTTCTAAAGCCACATTGTCTTCCAC ATCTGGTCTTGATTTAATGTCTGAATCTGGAGAAGGGGAAATCTCCCCACAAAGAGAAGTCTCAAGATCC CAGGATCAGTTCAGTGATATGAGAATCAGCATAAACCAGACGCCTGGGAAGAGTCTTGACTTTGGGTTTA CAATAAAATGGGATATTCCTGGGATCTTCGTAGCATCAGTTGAAGCAGGTAGCCCAGCAGAATTTTCTCA GCTACAAGTAGATGATGAAATTATTGCTATTAACAACACCAAGTTTTCATATAACGATTCAAAAGAGTGG GAGGAAGCCATGGCTAAGGCTCAAGAAACTGGACACCTAGTGATGGATGTGAGGCGCTATGGAAAGGCTG GTTCACCTGAAACAAAGTGGATTGATGCAACTTCTGGAATTTACAACTCAGAAAAATCTTCAAATCTATC TGTAACAACTGATTTCTCCGAAAGCCTTCAGAGTTCTAATATTGAATCCAAAGAAATCAATGGAATTCAT GATGAAAGCAATGCTTTTGAATCAAAAGCATCTGAATCCATTTCTTTGAAAAACTTAAAAAGGCGATCAC AATTTTTTGAACAAGGAAGCTCTGATTCGGTGGTTCCTGATCTTCCAGTTCCAACCATCAGTGCCCCGAG TCGCTGGGTGTGGGATCAAGAGGAGGAGCGGAAGCGGCAGGAGAGGTGGCAGAAGGAGCAGGACCGCCTA CTGCAGGAAAAATATCAACGTGAGCAGGAGAAACTGAGGGAAGAGTGGCAAAGGGCCAAACAGGAGGCAG AGAGAGAGAATTCCAAGTACTTGGATGAGGAACTGATGGTCCTAAGCTCAAACAGCATGTCTCTGACCAC ACGGGAGCCCTCTCTTGCCACCTGGGAAGCTACCTGGAGTGAAGGGTCCAAGTCTTCAGACAGAGAAGGA ACCCGAGCAGGAGAAGAGGAGAGGAGACAGCCACAAGAGGAAGTTGTTCATGAGGACCAAGGAAAGAAGC CGCAGGATCAGCTTGTTATTGAGAGAGAGAGGAAATGGGAGCAACAGCTTCAGGAAGAGCAAGAGCAAAA GCGGCTTCAGGCTGAGGCTGAGGAGCAGAAGCGTCCTGCGGAGGAGCAGAAGCGCCAGGCAGAGATAGAG CGGGAAACATCAGTCAGAATATACCAGTACAGGAGGCCTGTTGATTCCTATGATATACCAAAGACAGAAG AAGCATCTTCAGGTTTTCTTCCTGGTGACAGGAATAAATCCAGATCTACTACTGAACTGGATGATTACTC CACAAATAAAAATGGAAACAATAAATATTTAGACCAAATTGGGAACATGACCTCTTCACAGAGGAGATCC AAGAAAGAACAAGTACCATCAGGAGCAGAATTGGAGAGGCAACAAATCCTTCAGGAAATGAGGAAGAGAA CACCCCTTCACAATGACAACAGCTGGATCCGACAGCGCAGTGCCAGTGTCAACAAAGAGCCTGTTAGTCT TCCTGGGATCATGAGAAGAGGCGAATCTTTAGATAACCTGGACTCCCCCCGATCCAATTCTTGGAGACAG CCTCCTTGGCTCAATCAGCCCACAGGATTCTATGCTTCTTCCTCTGTGCAAGACTTTAGTCGCCCACCAC CTCAGCTGGTGTCCACATCAAACCGTGCCTACATGCGGAACCCCTCCTCCAGCGTGCCCCCACCTTCAGC TGGCTCCGTGAAGACCTCCACCACAGGTGTGGCCACCACACAGTCCCCCACCCCGAGAAGCCATTCCCCT TCAGCTTCACAGTCAGGCTCTCAGCTGCGTAACAGGTCAGTCAGTGGGAAGCGCATATGCTCCTACTGCA ATAACATTCTGGGCAAAGGAGCCGCCATGATCATCGAGTCCCTGGGTCTTTGTTATCATTTGCATTGTTT TAAGTGTGTTGCCTGTGAGTGTGACCTCGGAGGCTCTTCCTCAGGAGCTGAAGTCAGGATCAGAAACCAC CAACTGTACTGCAACGACTGCTATCTCAGATTCAAATCTGGACGGCCAACCGCCATGTGATGTAAGCCTC CATACGAAAGCACTGTTGCAGATAGAAGAAGAGGTGGTTGCTGCTCATGTAGATCTATAAATATGTGTTG TATGTCTTTTTTGCTTTTTTTTTAAAAAAAAGAATAACTTTTTTTGCCTCTTTAGATTACATAGAAGCAT TGTAGTCTTGGTAGAACCAGTATTTTTGTTGTTTATTTATAAGGTAATTGTGTGTGGGGAAAAGTGCAGT ATTTACCTGTTGAATTCAGCATCTTGAGAGCACAAGGGAAAAAATAAGAACCTACGAATATTTTTGAGGC AGATAATGATCTAGTTTGACTTTCTAGTTAGTGGTGTTTTGAAGAGGGTATTTTATTGTTTTTTAAAAAA AGGTTCTTAAACATTATTTGAAATAGTTAATATAAATACATAATTGCATTTGCTCTGTTTATTGTAATGT ATTCTAAATTAATGCAGAACCATATGGAAAATTTCATTAAAATCTATCCCCAAATGTGCTTTCTGTATCC TTCCTTCTACCTATTATTCTGATTTTTAAAAATGCAGTTAATGTACCATTTATTTGCTTGATGAAGGGAG CTCTATTTTCTTTACCAGAAATGTTGCTAAGTAATTCCCAATAGAAAGCTGCTTATTTTCATTAATGAAA AATAACCATGGTTTGTATACTAGAAGTCTTCTTCAGAAACTGGTGAGCCTTTCTGTTCAATTGCATTTGT AAATAAACTTGCTGATGCATTTAACGAGTGGGTCGTCTTTTTCTTAGGTGTATGTGTCTGACCTCAGGCC TTTTAGCCATATTTCAGTATGTGGCCTTTTTTGATGTTATGTTTTATCCAGTAGCTTTACTAAGGTATAA TTGATGTAATAAACTGCATATATTTAAAGTGTATACTTTGACAAATTTTGACATGGTGTATACCTTCGAA ACTATGCCACAGTCTGGATGTGTTTACTGAAACATTTTAATAAGGAAGTTTATTTTTGATAAAGTTATGT T T T T GG AT AC AAT AT AT T T G T AT GG T G AG AG T G AT G AAT T G T T GG AT C AT T T G AAT AAAAT C T T T T AC T A AC C C C AT G AT AAAAGG AG AAG AC AAC AG T GAGCT TAG AAT AT C T AT AAAGC AAAAAAT GTAGTCTCTTGT TTAAAAAATCTGGAGCGGGAATGCAAGGATACAAAACTTTAGCATGCTTTGAGCAAAAATTTAAACTTAC TGGAATCTTTTATAATAATGTAAGTGGAATGGAGGATTCTAGGAACTGAGAACTGTATTGGAATAGGTTC AAAATATGTAAGAAATGCTAATGTGGGAGATAAAAATTTTATTTAGTACTTATTCTGATTATTATTAAAG TAATAATGTGTTCCTTGAGGATAACTTGTCAAATGCCCCAAAGCATAAAGAATATAATTCTGAATCCCAA ATTCCAAAGACAAGAACTCTGTGTTTGAATTCATTCTGCATATAATTATTTATAAGTATAGATTGTGAAT TTTTCCATGTTCTTAAAATTATTTTTATCTTTTTTCATGGTTGCATAGTGCTCCATTGTTTGGCCTTGGT AATATTTAGTTGATAATTCCATTACTGTGTATTTTTCACTTGTTTCTAAGATCAAACATTTTAATATGTG CATGTTATATATAAATATGTAAATTCTGTGATACTCTATGATCATCTCTTTCTTTATATTATTTTCATAG ACATGAAATAGTTGCTCAGAGATTATGCATTTTAAGACACTCATAGTATATATTGCCAAAGTGGTTTCCA GAAAGGCACTGCTGGCTTCGACTCCTATAAGCAGCACGTGGGCTTGTTCATCTCACTGCATGTTTATGAA GATACAGTTCTTTTGCCTTGTTCTCTGCCTGATGTGTATGCAGAGGCAGCCCTCAATATGCAGTGGTTGA ATAAATGAATGAAGAAACCACTATCAAAAAAAAAAAAAAAAAA Amino Acid Sequence
GenBank: ACV60543.1
SEQ ID NO: 30
MEGLEEAEANCSVAFAEAQRWVEAVTEKNFETKDFRASLENGVLLCDLINKLKPGVIKKINRLSTPIAGL DNINVFLKACEQIGLKEAQLFHPGDLQDLSNRVTVKQEETDRRVKNVLITLYWLGRKAQSNPYYNGPHLN LKAFENLLGQALTKALEDSSFLKRSGRDSGYGDIWCPERGEFLAPPRHHKREDSFESLDSLGSRSLTSCS SDITLRGGREGFESDTDSEFTFKMQDYNKDDMSYRRISAVEPKTALPFNRFLPNKSRQPSYVPAPLRKKK PDKHEDNRRSWASPVYTEADGTFSSNQRRIWGTNVENWPTVQGTSKSSCYLEEEKAKTRSIPNIVKDDLY VRKLSPVMPNPGNAFDQFLPKCWTPEDVNWKRIKRETYKPWYKEFQGFSQFLLLQALQTYSDDILSSETH TKIDPTSGPRLITRRKNLSYAPGYRRDDLEMAALDPDLENDDFFVRKTGVFHANPYVLRAFEDFRKFSEQ DDSVERDI ILQCREGELVLPDLEKDDMIVRRIPAQKKEVPLSGAPDRYHPVPFPEPWTLPPEIQAKFLCV FERTCPSKEKSNSCRILVPSYRQKKDDMLTRKIQSWKLGTTVPPISFTPGPCSEADLKRWEAIREASRLR HKKRLMVERLFQKIYGENGSKSMSDVSAEDVQNLRQLRYEEMQKIKSQLKEQDQKWQDDLAKWKDRRKSY TSDLQKKKEEREEIEKQALEKSKRSSKTFKEMLQDRESQNQKSTVPSRRRMYSFDDVLEEGKRPPTMTVS EASYQSERVEEKGATYPSEIPKEDSTTFAKREDRVTTEIQLPSQSPVEEQSPASLSSLRSRSTQMESTRV SASLPRSYRKTDTVRLTSWTPRPFGSQTRGISSLPRSYTMDDAWKYNGDVEDIKRTPNNWSTPAPSPD ASQLASSLSSQKEVAATEEDVTRLPSPTSPFSSLSQDQAATSKATLSSTSGLDLMSESGEGEISPQREVS RSQDQFSDMRISINQTPGKSLDFGFTIKWDIPGIFVASVEAGSPAEFSQLQVDDEI IAINNTKFSYNDSK EWEEAMAKAQETGHLVMDVRRYGKAGSPETKWIDATSGIYNSEKSSNLSVTTDFSESLQSSNIESKEING IHDESNAFESKASESISLKNLKRRSQFFEQGSSDSWPDLPVPTISAPSRWVWDQEEERKRQERWQKEQD RLLQEKYQREQEKLREEWQRAKQEAERENSKYLDEELMVLSSNSMSLTTREPSLATWEATWSEGSKSSDR EGTRAGEEERRQPQEEWHEDQGKKPQDQLVIERERKWEQQLQEEQEQKRLQAEAEEQKRPAEEQKRQAE IERETSVRIYQYRRPVDSYDIPKTEEASSGFLPGDRNKSRSTTELDDYSTNKNGNNKYLDQIGNMTSSQR RSKKEQVPSGAELERQQILQEMRKRTPLHNDNSWIRQRSASVNKEPVSLPGIMRRGESLDNLDSPRSNSW RQPPWLNQPTGFYASSSVQDFSRPPPQLVSTSNRAYMRNPSSSVPPPSAGSVKTSTTGVATTQSPTPRSH SPSASQSGSQLRNRSVSGKRICSYCNNILGKGAAMI IESLGLCYHLHCFKCVACECDLGGSSSGAEVRIR NHQLYCNDCYLRFKSGRPTAM
Biomarker: LIM domain-binding protein 3 (LDB3/Cypher)
mRNA Sequence
NCBI Reference Sequence: NM_001171610.1
SEQ ID NO: 31
ACAGTCCATTTATGGGCGCAGCCGAAGGCAGATATCAGTGTCGATGGCATTCGCTCCCAGCTATTCTTAG GAGCCTCTCAAGAGCTCCACGCAGCCCGGCTGGGCAGCAAGGGACAGAACAGAGGCGGCCGCTGACAGCA CCAGCATGTCTTACAGTGTGACCCTGACTGGGCCCGGGCCCTGGGGCTTCCGTCTGCAGGGGGGCAAGGA CTTCAACATGCCCCTCACTATCTCCCGGATCACACCAGGCAGCAAGGCAGCCCAGTCCCAGCTCAGCCAG GGTGACCTCGTGGTGGCCATTGACGGCGTCAACACAGACACCATGACCCACCTGGAAGCCCAGAACAAGA TCAAGTCTGCCAGCTACAACTTGAGCCTCACCCTGCAGAAATCAAAGCGTCCCATTCCCATCTCCACGAC AGCACCTCCAGTCCAGACCCCTCTGCCGGTGATCCCTCACCAGAAGGACCCCGCTCTGGACACGAACGGC AGCCTGGTGGCACCCAGCCCCAGCCCTGAGGCGAGGGCCAGCCCAGGCACCCCAGGCACCCCGGAGCTCA GGCCCACCTTTAGCCCTGCCTTCTCCCGGCCCTCCGCCTTCTCCTCACTCGCCGAGGCCTCTGACCCTGG CCCTCCGCGGGCCAGCCTGAGGGCCAAGACCAGCCCAGAGGGGGCCCGGGACCTACTCGGCCCAAAAGCC CTGCCGGGCTCGAGCCAGCCGAGGCAATATAACAACCCCATTGGCCTGTACTCGGCAGAGACCCTGAGGG AGATGGCTCAGATGTACCAGATGAGCCTCCGAGGGAAGGCCTCGGGTGTCGGACTCCCAGGAGGCGCCGA CTACCAGGAACGCTTCAACCCCAGTGCCCTGAAGGACTCGGCCCTGTCCACCCACAAGCCCATCGAGGTG AAGGGGCTGGGCGGCAAGGCCACCATCATCCATGCGCAGTACAACACGCCCATCAGCATGTATTCCCAGG ATGCCATCATGGATGCCATCGCTGGGCAGGCCCAAGCCCAAGGCAGTGACTTCAGTGGGAGCCTCCCTAT TAAGGACCTTGCCGTAGACAGCGCCTCTCCCGTCTACCAGGCTGTGATTAAGAGCCAGAACAAGCCAGAA GATGAGGCTGACGAGTGGGCACGCCGTTCCTCCAACCTGCAGTCTCGCTCCTTCCGCATCCTGGCCCAGA TGACGGGGACAGAATTCATGCAAGACCCTGATGAAGAAGCTCTGCGAAGGTCAAGGCCCCAGGCCTCTTC CTACAGCCCCGCAGTGGCCGCCTCTTCAGCACCTGCCACCCACACCAGCTACAGTGAGGGCCCCGCCGCC CCTGCACCCAAGCCCCGGGTTGTCACCACTGCCAGCATCCGGCCTTCTGTCTACCAGCCAGTGCCTGCAT CTACCTACAGCCCGTCCCCAGGGGCCAATTACAGTCCCACTCCCTACACCCCCTCCCCTGCCCCTGCCTA CACCCCCTCCCCTGCCCCTGCCTACACCCCCTCACCTGTCCCCACCTACACTCCATCCCCAGCACCAGCC TATACCCCCTCACCTGCCCCCAACTATAACCCTGCACCCTCGGTGGCCTACAGCGGGGGCCCTGCGGAGC CTGCCAGCCGTCCACCCTGGGTGACAGATGATAGCTTCTCCCAGAAGTTTGCCCCGGGCAAGAGCACCAC CTCCATCAGCAAGCAGACCCTGCCCCGGGGAGGCCCAGCCTACACCCCAGCGGGTCCTCAGGTGCCACCA CTTGCCAGGGGGACCGTCCAGAGGGCTGAGCGATTCCCAGCCAGCAGCCGGACTCCACTCTGCGGTCACT GCAACAATGTCATCCGGGGCCCATTTCTGGTAGCCATGGGCCGTTCTTGGCACCCTGAAGAGTTCACCTG TGCCTACTGCAAGACTTCCCTGGCAGATGTGTGCTTTGTGGAAGAGCAGAACAACGTTTACTGTGAGCGA TGTTATGAGCAATTCTTTGCCCCGCTGTGTGCCAAGTGCAACACCAAAATTATGGGGGAAGTAATGCATG CCTTGAGACAGACATGGCACACCACCTGCTTCGTCTGTGCGGCCTGCAAGAAGCCTTTTGGGAACAGCCT CTTCCACATGGAAGACGGGGAGCCCTACTGCGAGAAAGACTACATCAATCTGTTCAGCACCAAGTGCCAT GGCTGCGATTTCCCCGTGGAGGCTGGCGACAAGTTTATCGAAGCCCTGGGCCACACTTGGCACGACACCT GCTTCATTTGCGCAGTCTGCCATGTGAATCTGGAGGGGCAGCCGTTCTACTCCAAGAAGGACAGACCCCT GTGCAAGAAGCACGCACACACCATCAACTTGTAGGCGGCCAAGGCCGCCTGTGCTGACGAGGCCCGGAGC TGCTCCTGCTGCTGGCAACAAAGGATTCGGGAGGCTGATGTTTCTTCTGAGGGGAATGGGGAGAGAGAGG AAGCGACTGAGCCCTTTGGAAGTATAATTTTAGGTTTTTTCTTCTGTACACAGATCGTGCATTTGCATAG T T C AG AC TAGGAGCCAAA G AAG AC CAAAAC C AAGC AG TATTAATC C AAG AC GG AAT TG AC TCA GACATTTAGAGCAGAATTCCAAGAACTCAAAAGTGAAAAGCAACAAGCAGCTTTCCCAAAGCGATACACT TGCTTTGGTCACCAGAGGAGGACAGAGCTTAGAGCAGCTGTGGAGAATCTGAAGCATTCTGCGGAGTTCT TAAGCGCTCCCCTGGCAAACAAATTGAAGTGCCAAACAGCACTCGCTGCAGGGTATTTTTAGAGTCATAG CTGAGAGCTTGTTAGCTAAGACCCATTGGGCTTTCCTCACCAAAAAAGGAAGTGTTATTCCATTACTAGC GTCATGGAGCTACCTCTGCGCATCAGACTTCAGACCTTGAACAAACTTAAAACCTTCTTGGGAGCCCGGA CGTCCAAAGAGATGTCTTCTGGGAGCCACTGGGCAATTGCCAGGGCTCCAGGAAGGGCTCTGGCTCAGGT TGCAGACAGCTGAGAAAAGATGGCCCTGTCAGCCACCCTCTCTCAGTCTGAAACATCCAACATCCCCAGA AGGCTTAGCTCCTTTTTGAATTGTGATGGGAAAGTAGAGTTGGGTTTTTCCAGTTTTGCTCTGTGGTGTG TGAGAGATTTTTTTAAAGGCTTTGGGTTGTCTTTGGCCTTTGTTTAGCTTTAAGGGTTCGTTAGCATGAG TGTCCAGTCGTGTGCATGAATTTCACCCCAACTTGTGACTGCTCACTTATGACGTCTCCCCCAGTACCCT CCATCTCAAATAGGCTTGGTGGCCTGTGGAAAAGAAGAGAGACAGAGAGACAGTGTCTGAAACAGGATGG CAGAATAGGCTCACATGCCCAAACTCTGGGTGGGGAAGAGGAAACTTACTTTCTGCCACCCTCAGTAAGA ACACACGAGGAGGCAGGACCTCCCACCTTCAGGTCTGCATCATCCTTTTCAAATGTTCCTTTAAATGCAG CACACTGAGTTTGTACAATTGTGTTAACTGCTGGAAGGGACAGATGCACTGATATATATGCATTTGCTGT TTTGGCCAATATTTTGAAAATGTATGAGCTGAGTTGATCTAGCTATTATTTAAGTATTTATTGAAGTAGA GGGGCCTTCAAACTACTTTATACTAGTGATAGTTTGAGTTAGGTAAGCATCTTAAAGCTGTTTGGTGATA AAGAAGGCAGCTTAGATTCTGTGGTTGGAAACAGTGTAGTCGCTTCCCTTTTTAGGAAGCCCTGTTAATA TGCTCATTGAAAACATGGCATTGAAGCAGGCACTTGCGTGGATGTTTCTCACTTGAGCACGATATTTAGG CTCTCTTCCAACTCACTCTATTCTGTCCTCACTCCTGTTTTGGATTTTTCTCTTTGCATGTTTGAAATGT TTTATGGGAATGTATTAGAACTCTTTTCTTCTAAGGACTGAGACTTCCAGGGGATTGCCATCTTACCTGT CTCTTCTCCATGAGGGAGAAGGAAGCAGCTAGCTATGTCCCTAGCTGCAGGAAGCCCCTATTTTTTCCAA GCACGAAGCCACCAGTCTCCCCCAGGGAGCATCAGGAAGGGACATGGATGTGCTCCTGCCACAGGGCCCT TCCTACCTTTGGATCTGTGAGAAGGTGAATACAAAGCAGCAGGCAGAGTAAAATCTGCTGGGACTGCCTG GAGATTTGTCAGGAGCTGCAGACAAGTACCTTGGAGCATTCTGTTATTTTTGGAAAGTTCAAATATGCAG GGACAAGGAGGTTGCTGACTGTACTGACAGGCTCTAAGTCATTTTCTCCAAAAACTATCTATTCAATTAT CAGGGGCTGGTCTTGAGGAAGGAAAAAAAAAAAAAAACGTTCCCAGAATTCAGTTTCCAAAATCTCTTTT TAAAGGGTTTACACACACACACACACACACACACACACACACACACACACACACACACGATCATTAAAAA GTGTATGCTCTTTAAGAAGAAAAGTAAAATATCTCAAAGGACGGTTTCACCACCGTCCTTTATTGAATCA ATTTTTCTACATTTCAGAGCAAGTGTAGATTCTGAGGGACTCCTATTTGCCAAAAAGACAAAACTAGCAA AAAAAAAACAAAAAAACAAAAAAAAAACCACTTAAAAGGTAGCAGGAAAAGAAGGTAGTTTTGAGTGTGG TTCACTCAGTGTCTGTGAGTCTGGTGTAGTGTCAGGAGTAAGGCCGTGTCTAGCTCAAGTTTACATTTGG ATGTCCTACAACACTAAACAAAATTTTTCATAATCCATGGTGGGGAGCACACTTTGGAGCTACATTTCTT GTCTCCTCATTGTTGACATTAATTAAACATTTATAGGCCAGGCACAGTGGCTCACGCCTGTTATCCCAGC ACTTTGGGAGGCCGAGGCAGGTGAATCACCTGAGGTCAGGAGTTTGAAACCAGCCTGGCCAATATGGTGA AACCCCATCTCTACTAAAAATACACAAAATTAGCCAGGTGTGGTGGCAGGCGCCTGTAGTTCCAGCTACT TGGGAGGCTGAGGCAGGAATCTCCTGAATCCTGGAGGCGGAGGTTGCAGTGAGCCGAGATTATGCCATTG CACTCCAGCCTGGGCAACAGGAGCGAAACTCCGTTGCAAAAAAAAAAAAAAAAAAAAAATTATAATCACA ACTTTTTGCAATGGAGTGACTTATATCTGCAGCTTATATCTGCAGTGTTTGTGTTAGGAACCTAGCTTTT ATAATGTGTTAACTTTTTAACTCAGTATTCTGGCTTTGGGATTTTTTGTTTTGTTTTTGGAAACATTTCA GAAGTGGAATGTAGCCTGTTAAAGGTGTGCACAAAAATATTTTGCATGTGTTTTTTTTTTTGCCTGTGTG AATTCTACTTTTTAGCAAAAATAAAGCCCCCCAAAGGATGTGCAAA
Amino Acid Sequence
NCBI Reference Sequence: NP_001165081.1
SEQ ID NO: 32
MSYSVTLTGPGPWGFRLQGGKDFNMPLTISRITPGSKAAQSQLSQGDLWAIDGVNTDTMTHLEAQNKIK SASYNLSLTLQKSKRPIPISTTAPPVQTPLPVIPHQKDPALDTNGSLVAPSPSPEARASPGTPGTPELRP TFSPAFSRPSAFSSLAEASDPGPPRASLRAKTSPEGARDLLGPKALPGSSQPRQYNNPIGLYSAETLREM AQMYQMSLRGKASGVGLPGGADYQERFNPSALKDSALSTHKPIEVKGLGGKATI IHAQYNTPISMYSQDA IMDAIAGQAQAQGSDFSGSLPIKDLAVDSASPVYQAVIKSQNKPEDEADEWARRSSNLQSRSFRILAQMT GTEFMQDPDEEALRRSRPQASSYSPAVAASSAPATHTSYSEGPAAPAPKPRWTTASIRPSVYQPVPAST YSPSPGANYSPTPYTPSPAPAYTPSPAPAYTPSPVPTYTPSPAPAYTPSPAPNYNPAPSVAYSGGPAEPA SRPPWVTDDSFSQKFAPGKSTTSISKQTLPRGGPAYTPAGPQVPPLARGTVQRAERFPASSRTPLCGHCN NVIRGPFLVAMGRSWHPEEFTCAYCKTSLADVCFVEEQNNVYCERCYEQFFAPLCAKCNTKIMGEVMHAL RQTWHTTCFVCAACKKPFGNSLFHMEDGEPYCEKDYINLFSTKCHGCDFPVEAGDKFIEALGHTWHDTCF ICAVCHVNLEGQPFYSKKDRPLCKKHAHTINL
Biomarker: Striated muscle preferentially expressed kinase (SPEG) mRNA Sequence
GenBank: AY603755.1
SEQ ID NO: 33
GACTCAGAGACGGCTGAGGATGACATCAGCGATGTGCAGGGAACCCAGCGCCTGGAGCTTCGGGATGACG GGGCCTTCAGCACCCCCACGGGGGGTTCTGACACCCTGGTGGGCACCTCCCTGGACACACCCCCGACCTC CGTGACAGGCACCTCAGAGGAGCAAGTGAGCTGGTGGGGCAGCGGGCAGACGGTCCTGGAGCAGGAAGCG GGCAGTGGGGGTGGCACCCGCCGCCTCCCGGGCAGCCCAAGGCAAGCACAGGCAACCGGGGCCGGGCCAC GGCACCTGGGGGTGGAGCCGCTGGTGCGGGCATCTCGAGCTAATCTGGTGGGCGCAAGCTGGGGGTCAGA GGATAGCCTTTCCGTGGCCAGTGACCTGTACGGCAGCGCATTCAGCCTGTACAGAGGACGGGCGCTCTCT ATCCACGTCAGCGTCCCTCAGAGCGGGTTGCGCAGGGAGGAGCCCGACCTTCAGCCTCAACTGGCCAGCG AAGCCCCACGCCGCCCTGCCCAGCCGCCTCCTTCCAAATCCGCGCTGCTCCCCCCACCGTCCCCTCGGGT CGGGAAGCGGTCCCCGCCGGGACCCCCGGCCCAGCCCGCGGCCACCCCCACGTCGCCCCACCGTCGCACT CAGGAGCCTGTGCTGCCCGAGGACACCACCACCGAAGAGAAGCGAGGGAAGAAGTCCAAGTCGTCCGGGC CCTCCCTGGCGGGCACCGCGGAATCCCGACCCCAGACGCCACTGAGCGAGGCCTCAGGCCGCCTGTCGGC GTTGGGCCGATCGCCTAGGCTGGTGCGCGCCGGCTCCCGCATCCTGGACAAGCTGCAGTTCTTCGAGGAG CGACGGCGCAGCCTGGAGCGCAGCGACTCGCCGCCGGCGCCCCTGCGGCCCTGGGTGCCCCTGCGCAAGG CCCGCTCTCTGGAGCAGCCCAAGTCGGAGCGCGGCGCACCGTGGGGCACCCCCGGGGCCTCGCAGGAAGA ACTGCGGGCGCCAGGCAGCGTGGCCGAGCGGCGCCGCCTGTTCCAGCAGAAAGCGGCCTCGCTGGACGAG CGCACGCGTCAGCGCAGCCCGGCCTCAGACCTCGAGCTGCGCTTCGCCCAGGAGCTGGGCCGCATCCGCC GCTCCACGTCGCGGGAGGAGCTGGTGCGCTCGCACGAGTCCCTGCGCGCCACGCTGCAGCGTGCCCCATC CCCTCGAGAGCCCGGCGAGCCCCCGCTCTTCTCTCGGCCCTCCACCCCCAAGACATCGCGGGCCGTGAGC CCCGCCGCCGCCCAGCCGCCCTCTCCGAGCAGCGCGGAGAAGCCGGGGGACGAGCCTGGGAGGCCCAGGA GCCGCGGGCCGGCGGGCAGGACAGAGCCGGGGGAAGGCCCGCAGCAGGAGGTTAGGCGTCGGGACCAATT CCCGCTGACCCGGAGCAGAGCCATCCAGGAGTGCAGGAGCCCTGTGCCGCCCCCCGCCGCCGATCCCCCA GAGGCCAGGACGAAAGCACCCCCCGGTCGGAAGCGGGAGCCCCCGGCGCAGGCCGTGCGCTTCCTGCCCT GGGCCACGCCGGGCCTGGAGGGCGCTGCTGTACCCCAGACCTTGGAGAAGAACAGGGCGGGGCCTGAGGC AGAGAAGAGGCTTCGCAGAGGGCCGGAGGAGGACGGTCCCTGGGGGCCCTGGGACCGCCGAGGGGCCCGC AGCCAGGGCAAAGGTCGCCGGGCCCGGCCCACCTCCCCTGAGCTCGAGTCTTCGGATGACTCCTACGTGT CCGCTGGAGAAGAGCCCCTAGAGGCCCCTGTGTTTGAGATCCCCCTGCAGAATGTGGTGGTGGCACCAGG GGCAGATGTGCTGCTCAAGTGTATCATCACTGCCAACCCCCCGCCCCAAGTGTCCTGGCACAAGGATGGG TCAGCGCTGCGCAGCGAGGGCCGCCTCCTCCTCCGGGCTGAGGGTGAGCGGCACACCCTGCTGCTCAGGG AGGCCAGGGCAGCAGATGCCGGGAGCTATATGGCCACCGCCACCAACGAGCTGGGCCAGGCCACCTGTGC CGCCTCACTGACCGTGAGACCCGGTGGGTCTACATCCCCTTTCAGCAGCCCCATCACCTCCGACGAGGAA TACCTGAGCCCCCCAGAGGAGTTCCCAGAGCCTGGGGAGACCTGGCCGCGAACCCCCACCATGAAGCCCA GTCCCAGCCAGAACCGCCGTTCTTCTGACACTGGCTCCAAGGCACCCCCCACCTTCAAGGTCTCACTTAT GGACCAGTCAGTAAGAGAAGGCCAAGATGTCATCATGAGCATCCGCGTGCAGGGGGAGCCCAAGCCTGTG GTCTCCTGGCTGAGAAACCGCCAGCCCGTGCGCCCAGACCAGCGGCGCTTTGCGGAGGAGGCTGAGGGTG GGCTGTGCCGGCTGCGGATCCTGGCTGCAGAGCGTGGCGATGCTGGTTTCTACACTTGCAAAGCGGTCAA TGAGTATGGTGCTCGGCAGTGCGAGGCCCGCTTGGAGGTCCGAGCACACCCTGAAAGCCGGTCCCTGGCC GTGCTGGCCCCCCTGCAGGACGTGGACGTGGGGGCCGGGGAGATGGCGCTGTTTGAGTGCCTGGTGGCGG GGCCCACTGACGTGGAGGTGGATTGGCTGTGCCGTGGCCGCCTGCTGCAGCCTGCACTGCTCAAATGCAA GATGCATTTCGATGGCCGCAAATGCAAGCTGCTACTTACATCTGTACATGAGGACGACAGTGGCGTCTAC ACCTGCAAGCTCAGCACGGCCAAAGATGAGCTGACCTGCAGTGCCCGGCTGACCGTGCGGCCCTCGTTGG CACCCCTGTTCACACGGCTGCTGGAAGATGTGGAGGTGTTGGAGGGCCGAGCTGCCCGTTTCGACTGCAA GATCAGTGGCACCCCGCCCCCTGTTGTTACCTGGACTCATTTTGGCTGCCCCATGGAGGAGAGTGAGAAC TTGCGGCTGCGGCAGGACGGGGGTCTGCACTCACTGCACATTGCCCATGTGGGCAGCGAGGACGAGGGGC TCTATGCGGTCAGTGCTGTTAACACCCATGGCCAGGCCCACTGCTCAGCCCAGCTGTATGTAGAAGAGCC CCGGACAGCCGCCTCAGGCCCCAGCTCGAAGCTGGAGAAGATGCCATCCATTCCCGAGGAGCCAGAGCAG GGTGAGCTGGAGCGGCTGTCCATTCCCGACTTCCTGCGGCCACTGCAGGACCTGGAGGTGGGACTGGCCA AGGAGGCCATGCTAGAGTGCCAGGTGACCGGCCTGCCCTACCCCACCATCAGCTGGTTCCACAATGGCCA CCGCATCCAGAGCAGCGACGACCGGCGCATGACACAGTACAGGGATGTCCATCGCTTGGTGTTCCCTGCC GTGGGGCCTCAGCACGCCGGTGTCTACAAGAGCGTCATTGCCAACAAGCTGGGCAAAGCTGCCTGCTATG CCCACCTGTATGTCACAGATGTGGTCCCAGGCCCTCCAGATGGCGCCCCGCAGGTGGTGGCTGTGACGGG GAGGATGGTCACACTCACATGGAACCCCCCCAGGAGTCTGGACATGGCCATCGACCCGGACTCCCTGACG TACACAGTGCAGCACCAGGTGCTGGGCTCGGACCAGTGGACGGCACTGGTCACAGGCCTGCGGGAGCCAG GGTGGGCAGCCACAGGGCTGCGTAAGGGGGTCCAGCACATCTTCCGGGTCCTCAGCACCACTGTCAAGAG CAGCAGCAAGCCCTCACCCCCTTCTGAGCCTGTGCAGCTGCTGGAGCACGGCCCAACCCTGGAGGAGGCC CCTGCCATGCTGGACAAACCAGACATCGTGTATGTGGTGGAGGGACAGCCTGCCAGCGTCACCGTCACAT TCAACCATGTGGAGGCCCAGGTCGTCTGGAGGAGCTGCCGAGGGGCCCTCCTAGAGGCACGGGCCGGTGT GTACGAGCTGAGCCAGCCAGATGATGACCAGTACTGTCTTCGGATCTGCCGGGTGAGCCGCCGGGACATG GGGGCCCTCACCTGCACCGCCCGAAACCGTCACGGCACACAGACCTGCTCGGTCACATTGGAGCTGGCAG AGGCCCCTCGGTTTGAGTCCATCATGGAGGACGTGGAGGTGGGGGCTGGGGAAACTGCTCGCTTTGCGGT GGTGGTCGAGGGAAAACCACTGCCGGACATCATGTGGTACAAGGACGAGGTGCTGCTGACCGAGAGCAGC CATGTGAGCTTCGTGTACGAGGAGAATGAGTGCTCCCTGGTGGTGCTCAGCACGGGGGCCCAGGATGGAG GCGTCTACACCTGCACCGCCCAGAACCTGGCGGGTGAGGTCTCCTGCAAAGCAGAGTTGGCTGTGCATTC AGCTCAGACAGCTATGGAGGTCGAGGGGGTCGGGGAGGATGAGGACCATCGAGGAAGGAGACTCAGCGAC TTTTATGACATCCACCAGGAGATCGGCAGGGGTGCTTTCTCCTACTTGCGGCGCATAGTGGAGCGTAGCT CCGGCCTGGAGTTTGCGGCCAAGTTCATCCCCAGCCAGGCCAAGCCAAAGGCATCAGCGCGTCGGGAGGC CCGGCTGCTGGCCAGGCTCCAGCACGACTGTGTCCTCTACTTCCATGAGGCCTTCGAGAGGCGCCGGGGA CTGGTCATTGTCACCGAGCTCTGCACAGAGGAGCTGCTGGAGCGAATCGCCAGGAAACCCACCGTGTGTG AGTCTGAGATCCGGGCCTATATGCGGCAGGTGCTAGAGGGAATACACTACCTGCACCAGAGCCACGTGCT GCACCTCGATGTCAAGCCTGAGAACCTGCTGGTGTGGGATGGTGCTGCGGGCGAGCAGCAGGTGCGGATC TGTGACTTTGGGAATGCCCAGGAGCTGACTCCAGGAGAGCCCCAGTACTGCCAGTATGGCACACCTGAGT TTGTAGCACCCGAGATTGTCAATCAGAGCCCCGTGTCTGGAGTCACTGACATCTGGCCTGTGGGTGTTGT TGCCTTCCTCTGTCTGACAGGAATCTCCCCGTTTGTTGGGGAAAATGACCGGACAACATTGATGAACATC CGAAACTACAACGTGGCCTTCGAGGAGACCACATTCCTGAGCCTGAGCAGGGAGGCCCGGGGCTTCCTCA TCAAAGTGTTGGTGCAGGACCGGCTGAGACCTACCGCAGAAGAGACCCTAGAACATCCTTGGTTCAAAAC TCAGGCAAAGGGCGCAGAGGTGAGCACGGATCACCTGAAGCTATTCCTCTCCCGGCGGAGGTGGCAGCGC TCCCAGATCAGCTACAAATGCCACCTGGTGCTGCGCCCCATCCCCGAGCTGCTGCGGGCCCCCCCAGAGC GGGTGTGGGTGACCATGCCCAGAAGGCCACCCCCCAGTGGGGGGCTCTCATCCTCCTCGGATTCTGAAGA GGAAGAGCTGGAAGAGCTGCCCTCAGTGCCCCGCCCACTGCAGCCCGAGTTCTCTGGCTCCCGGGTGTCC CTCACAGACATTCCCACTGAGGATGAGGCCCTGGGGACCCCAGAGACTGGGGCTGCCACCCCCATGGACT GGCAGGAGCAGGGAAGGGCTCCCTCTCAGGACCAGGAGGCTCCCAGCCCAGAGGCCCTCCCCTCCCCAGG CCAGGAGCCCGCAGCTGGGGCTAGCCCCAGGCGGGGAGAGCTCCGCAGGGGCAGCTCGGCTGAGAGCGCC CTGCCCCGGGCCGGGCCGCGGGAGCTGGGCCGGGGCCTGCACAAGGCGGCGTCTGTGGAGCTGCCGCAGC GCCGGAGCCCCGGCCCGGGAGCCACCCGCCTGGCCCGGGGAGGCCTGGGTGAGGGCGAGTATGCCCAGAG GCTGCAGGCCCTGCGCCAGCGGCTGCTGCGGGGAGGCCCCGAGGATGGCAAGGTCAGCGGCCTCAGGGGT CCCCTGCTGGAGAGCCTGGGGGGCCGTGCTCGGGACCCCCGGATGGCACGAGCTGCCTCCAGCGAGGCAG CGCCCCACCACCAGCCCCCACTCGAGAACCGGGGCCTGCAAAAGAGCAGCAGCTTCTCCCAGGGTGAGGC GGAGCCCCGGGGCCGGCACCGCCGAGCGGGGGCGCCCCTCGAGATCCCCGTGGCCAGGCTTGGGGCCCGT AGGCTACAGGAGTCTCCTTCCCTGTCTGCCCTCAGCGAGGCCCAGCCATCCAGCCCTGCACGGCCCAGCG CCCCCAAACCCAGTACCCCTAAGTCTGCAGAACCTTCTGCCACCACACCTAGTGATGCTCCGCAGCCCCC CGCACCCCAGCCTGCCCAAGACAAGGCTCCAGAGCCCAGGCCAGAACCAGTCCGAGCCTCCAAGCCTGCA CCACCCCCCCAGGCCCTGCAAACCCTAGCGCTGCCCCTCACACCCTATGCTCAGATCATTCAGTCCCTCC AGCTGTCAGGCCACGCCCAGGGCCCCTCGCAGGGCCCTGCCGCGCCGCCTTCAGAGCCCAAGCCCCACGC TGCTGTCTTTGCCAGGGTGGCCTCCCCACCTCCGGGAGCCCCCGAGAAGCGCGTGCCCTCAGCCGGGGGT CCCCCGGTGCTAGCCGAGAAAGCCCGAGTTCCCACGGTGCCCCCCAGGCCAGGCAGCAGTCTCAGTAGCA GCATCGAAAACTTGGAGTCGGAGGCCGTGTTCGAGGCCAAGTTCAAGCGCAGCCGCGAGTCGCCCCTGTC GCTGGGGCTGCGGCTGCTGAGCCGTTCGCGCTCGGAGGAGCGCGGCCCCTTCCGTGGGGCCGAGGAGGAG GATGGCATATACCGGCCCAGCCCGGCGGGGACCCCGCTGGAGCTGGTGCGACGGCCTGAGCGCTCACGCT CGGTGCAGGACCTCAGGGCTGTCGGAGAGCCTGGCCTCGTCCGCCGCCTCTCGCTGTCACTGTCCCAGCG GCTGCGGCGGACCCCTCCCGCGCAGCGCCACCCGGCCTGGGAGGCCCGCGGCGGGGACGGAGAGAGCTCG GAGGGCGGGAGCTCGGCGCGGGGCTCCCCGGTGCTGGCGATGCGCAGGCGGCTGAGCTTCACCCTGGAGC GGCTGTCCAGCCGATTGCAGCGCAGTGGCAGCAGCGAGGACTCGGGGGGCGCGTCGGGCCGCAGCACGCC GCTGTTCGGACGGCTTCGCAGGGCCACGTCCGAGGGCGAGAGTCTGCGGCGCCTTGGCCTTCCGCACAAC CAGTTGGCCGCCCAGGCCGGCGCCACCACGCCTTCCGCCGAGTCCCTGGGCTCCGAGGCCAGCGCCACGT CGGGCTCCTCAGCCCCAGGGGAAAGCCGAAGCCGGCTCCGCTGGGGCTTCTCTCGGCCGCGGAAGGACAA GGGGTTATCGCCACCAAACCTCTCTGCCAGCGTCCAGGAGGAGTTGGGTCACCAGTACGTGCGCAGTGAG TCAGACTTCCCCCCAGTCTTCCACATCAAACTCAAGGACCAGGTGCTGCTGGAGGGGGAGGCAGCCACCC TGCTCTGCCTGCCAGCGGCCTGCCCTGCACCGCACATCTCCTGGATGAAAGACAAGAAGTCCTTGAGGTC AGAGCCCTCAGTGATCATCGTGTCCTGCAAAGATGGGCGGCAGCTGCTCAGCATCCCCCGGGCGGGCAAG CGGCACGCCGGTCTCTATGAGTGCTCGGCCACCAACGTACTGGGCAGCATCACCAGCTCCTGTACCGTGG CTGTGGCCCGAGTCCCAGGAAAGCTAGCTCCTCCAGAGGTAACCCAGACCTACCAGGACACGGCGCTGGT GCTGTGGAAGCCGGGAGACAGCCGGGCACCTTGCACGTATACGCTGGAGCGGCGAGTGGATGGGGAGTCT GTGTGGCACCCTGTGAGCTCAGGCATCCCCGACTGTTACTACAACGTGACCCACCTGCCAGTTGGCGTGA CTGTGAGGTTCCGTGTGGCCTGTGCCAACCGTGCTGGGCAGGGGCCCTTCAGCAACTCTTCTGAGAAGGT CTTTGTCAGGGGTACTCAAGATTCTTCAGCTGTGCCATCTGCTGCCCACCAAGAGGCCCCTGTCACCTCA AGGCCAGCCAGGGCCCGGCCTCCTGACTCTCCTACCTCACTGGCCCCACCCCTAGCTCCTGCTGCCCCCA CACCCCCGTCAGTCACTGTCAGCCCCTCATCTCCCCCCACACCTCCTAGCCAGGCCTTGTCCTCGCTCAA GGCTGTGGGTCCACCACCCCAAACCCCTCCACGAAGACACAGGGGCCTGCAGGCTGCCCGGCCAGCGGAG CCCACCCTACCCAGTACCCACGTCACCCCAAGTGAGCCCAAGCCTTTCGTCCTTGACACTGGGACCCCGA TCCCAGCCTCCACTCCTCAAGGGGTTAAACCAGTGTCTTCCTCTACTCCTGTGTATGTGGTGACTTCCTT TGTGTCTGCACCACCAGCCCCTGAGCCCCCAGCCCCTGAGCCCCCTCCTGAGCCTACCAAGGTGACTGTG CAGAGCCTCAGCCCGGCCAAGGAGGTGGTCAGCTCCCCTGGGAGCAGTCCCCGAAGCTCTCCCAGGCCTG AGGGTACCACTCTTCGACAGGGTCCCCCTCAGAAACCCTACACCTTCCTGGAGGAGAAAGCCAGGGGCCG CTTTGGTGTTGTGCGAGCGTGCCGGGAGAATGCCACGGGGCGAACGTTCGTGGCCAAGATCGTGCCCTAT GCTGCCGAGGGCAAGCCGCGGGTCCTGCAGGAGTACGAGGTGCTGCGGACCCTGCACCACGAGCGGATCA TGTCCCTGCACGAGGCCTACATCACCCCTCGGTACCTCGTGCTCATTGCTGAGAGCTGTGGCAACCGGGA ACTCCTCTGTGGGCTCAGTGACAGGTTCCGGTATTCTGAGGATGACGTGGCCACTTACATGGTGCAGCTG CTACAAGGCCTGGACTACCTCCACGGCCACCACGTGCTCCACCTAGACATCAAGCCAGACAACCTGCTGC TGGCCCCTGACAATGCCCTCAAGATTGTGGACTTTGGCAGTGCCCAGCCCTACAACCCCCAGGCCCTTAG GCCCCTTGGCCACCGCACGGGCACGCTGGAGTTCATGGCTCCGGAGATGGTGAAGGGAGAACCCATCGGC TCTGCCACGGACATCTGGGGAGCGGGTGTGCTCACTTACATTATGCTCAGTGGACGCTCCCCGTTCTATG AGCCAGACCCCCAGGAAACGGAGGCTCGGATTGTGGGGGGCCGCTTTGATGCCTTCCAGCTGTACCCCAA TACATCCCAGAGCGCCACCCTCTTCTTGCGAAAGGTTCTCTCTGTACATCCCTGGAGCCGGCCATCCAGC TGTCTGTCTGTCTGCCACAAGGAAATAAAAATGGCAAGCAGC
Amino Acid Sequence
NCBI Reference Sequence: NP_005867.3
SEQ ID NO: 34
MQKARGTRGEDAGTRAPPSPGVPPKRAKVGAGGGAPVAVAGAPVFLRPLK AAVCAGSDVRLRVWSGTP QPSLRWFRDGQLLPAPAPEPSCLWLRRCGAQDAGVYSCMAQNERGRASCEAVLTVLEVGDSETAEDDISD VQGTQRLELRDDGAFSTPTGGSDTLVGTSLDTPPTSVTGTSEEQVSWWGSGQTVLEQEAGSGGGTRRLPG SPRQAQATGAGPRHLGVEPLVRASRANLVGASWGSEDSLSVASDLYGSAFSLYRGRALSIHVSVPQSGLR REEPDLQPQLASEAPRRPAQPPPSKSALLPPPSPRVGKRSPPGPPAQPAATPTSPHRRTQEPVLPEDTTT EEKRGKKSKSSGPSLAGTAESRPQTPLSEASGRLSALGRSPRLVRAGSRILDKLQFFEERRRSLERSDSP PAPLRPWVPLRKARSLEQPKSERGAPWGTPGASQEELRAPGSVAERRRLFQQKAASLDERTRQRSPASDL ELRFAQELGRIRRS SREELVRSHESLRATLQRAPSPREPGEPPLFSRPS PK SRAVSPAAAQPPSPSS AEKPGDEPGRPRSRGPAGRTEPGEGPQQEVRRRDQFPLTRSRAIQECRSPVPPPAADPPEARTKAPPGRK REPPAQAVRFLPWATPGLEGAAVPQTLEKNRAGPEAEKRLRRGPEEDGPWGPWDRRGARSQGKGRRARPT SPELESSDDSYVSAGEEPLEAPVFEIPLQNVWAPGADVLLKCI ITANPPPQVSWHKDGSALRSEGRLLL RAEGERHTLLLREARAADAGSYMATATNELGQATCAASLTVRPGGSTSPFSSPITSDEEYLSPPEEFPEP GETWPRTPTMKPSPSQNRRSSDTGSKAPPTFKVSLMDQSVREGQDVIMSIRVQGEPKPWSWLRNRQPVR PDQRRFAEEAEGGLCRLRILAAERGDAGFYTCKAVNEYGARQCEARLEVRAHPESRSLAVLAPLQDVDVG AGEMALFECLVAGPTDVEVDWLCRGRLLQPALLKCKMHFDGRKCKLLLTSVHEDDSGVYTCKLSTAKDEL TCSARLTVRPSLAPLFTRLLEDVEVLEGRAARFDCKISGTPPPWTWTHFGCPMEESENLRLRQDGGLHS LHIAHVGSEDEGLYAVSAVNTHGQAHCSAQLYVEEPRTAASGPSSKLEKMPSIPEEPEQGELERLSIPDF LRPLQDLEVGLAKEAMLECQVTGLPYPTISWFHNGHRIQSSDDRRMTQYRDVHRLVFPAVGPQHAGVYKS VIANKLGKAACYAHLYVTDWPGPPDGAPQWAVTGRMVTLTWNPPRSLDMAIDPDSLTYTVQHQVLGSD QWTALVTGLREPGWAATGLRKGVQHIFRVLSTTVKSSSKPSPPSEPVQLLEHGPTLEEAPAMLDKPDIVY WEGQPASVTVTFNHVEAQWWRSCRGALLEARAGVYELSQPDDDQYCLRICRVSRRDMGALTCTARNRH GTQTCSVTLELAEAPRFESIMEDVEVGAGETARFAVWEGKPLPDIMWYKDEVLLTESSHVSFVYEENEC SLWLSTGAQDGGVYTCTAQNLAGEVSCKAELAVHSAQTAMEVEGVGEDEDHRGRRLSDFYDIHQEIGRG AFSYLRRIVERSSGLEFAAKFIPSQAKPKASARREARLLARLQHDCVLYFHEAFERRRGLVIVTELCTEE LLERIARKPTVCESEIRAYMRQVLEGIHYLHQSHVLHLDVKPENLLVWDGAAGEQQVRICDFGNAQELTP GEPQYCQYGTPEFVAPEIVNQSPVSGVTDIWPVGWAFLCLTGISPFVGENDRTTLMNIRNYNVAFEETT FLSLSREARGFLIKVLVQDRLRPTAEETLEHPWFKTQAKGAEVSTDHLKLFLSRRRWQRSQISYKCHLVL RPIPELLRAPPERVWVTMPRRPPPSGGLSSSSDSEEEELEELPSVPRPLQPEFSGSRVSLTDIPTEDEAL GTPETGAATPMDWQEQGRAPSQDQEAPSPEALPSPGQEPAAGASPRRGELRRGSSAESALPRAGPRELGR GLHKAASVELPQRRSPSPGATRLARGGLGEGEYAQRLQALRQRLLRGGPEDGKVSGLRGPLLESLGGRAR DPRMARAASSEAAPHHQPPLENRGLQKSSSFSQGEAEPRGRHRRAGAPLEIPVARLGARRLQESPSLSAL SEAQPSSPARPSAPKPSTPKSAEPSATTPSDAPQPPAPQPAQDKAPEPRPEPVRASKPAPPPQALQTLAL PL PYAQI IQSLQLSGHAQGPSQGPAAPPSEPKPHAAVFARVASPPPGAPEKRVPSAGGPPVLAEKARVP TVPPRPGSSLSSSIENLESEAVFEAKFKRSRESPLSLGLRLLSRSRSEERGPFRGAEEEDGIYRPSPAGT PLELVRRPERSRSVQDLRAVGEPGLVRRLSLSLSQRLRRTPPAQRHPAWEARGGDGESSEGGSSARGSPV LAMRRRLSFTLERLSSRLQRSGSSEDSGGASGRSTPLFGRLRRATSEGESLRRLGLPHNQLAAQAGATTP SAESLGSEASATSGSSAPGESRSRLRWGFSRPRKDKGLSPPNLSASVQEELGHQYVRSESDFPPVFHIKL KDQVLLEGEAATLLCLPAACPAPHISWMKDKKSLRSEPSVI IVSCKDGRQLLSIPRAGKRHAGLYECSAT NVLGSITSSCTVAVARVPGKLAPPEVPQTYQDTALVLWKPGDSRAPCTYTLERRVDGESVWHPVSSGIPD CYYNVTHLPVGVTVRFRVACANRAGQGPFSNSSEKVFVRGTQDSSAVPSAAHQEAPV SRPARARPPDSP TSLAPPLAPAAPTPPSVTVSPSSPPTPPSQALSSLKAVGPPPQTPPRRHRGLQAARPAEPTLPSTHVTPS EPKPFVLDTGTPIPASTPQGVKPVSSSTPVYWTSFVSAPPAPEPPAPEPPPEPTKVTVQSLSPAKEWS SPGSSPRSSPRPEGTTLRQGPPQKPYTFLEEKARGRFGWRACRENATGRTFVAKIVPYAAEGKRRVLQE YEVLRTLHHERIMSLHEAYITPRYLVLIAESCGNRELLCGLSDRFRYSEDDVATYMVQLLQGLDYLHGHH VLHLDIKPDNLLLAPDNALKIVDFGSAQPYNPQALRPLGHRTGTLEFMAPEMVKGEPIGSATDIWGAGVL TYIMLSGRSPFYEPDPQETEARIVGGRFDAFQLYPNTSQSATLFLRKVLSVHPWSRPSLQDCLAHPWLQD AYLMKLRRQTLTFTTNRLKEFLGEQRRRRAEAATRHKVLLRSYPGGP
Biomarker: Filamin-C
mRNA Sequence
NCBI Reference Sequence: NM_001458.4
SEQ ID NO: 35
CCCTGGAGGGAGAGAGAGCCAGAGAGCGGCCGAGCGCCTAGGAGGCCCGCCGAGCCTCGCCGAGCCCCGC CAGCCCCGGCGCGAGAGAAGTTGGAGAGGAGAGCAGCGCAGCGCAGCGAGTCCCGTGGTCGCGCCCCAAC AGCGCCCGACAGCCCCCGATAGCCCAAACCGCGGCCCTAGCCCCGGCCGCACCCCCAGCCCGCGCCAGCA TGATGAACAACAGCGGCTACTCAGACGCCGGCCTCGGCCTGGGCGATGAGACAGACGAGATGCCGTCCAC GGAGAAGGACCTGGCGGAGGACGCGCCGTGGAAGAAGATCCAGCAGAACACATTCACGCGCTGGTGCAAT GAGCACCTCAAGTGCGTGGGCAAGCGCCTGACCGACCTGCAGCGCGACCTCAGCGACGGGCTCCGGCTCA TCGCGCTGCTCGAGGTGCTCAGCCAGAAGCGCATGTACCGCAAGTTCCATCCGCGCCCCAACTTCCGCCA AATGAAGCTGGAGAACGTGTCCGTGGCCCTCGAGTTCCTCGAGCGCGAGCACATCAAGCTCGTGTCCATA GACAGCAAGGCCATCGTGGATGGGAACCTGAAGCTGATCCTGGGCCTGATCTGGACGCTGATCCTGCACT ACTCCATCTCCATGCCCATGTGGGAGGATGAAGATGATGAGGATGCCCGCAAACAGACGCCCAAGCAGCG GCTGCTTGGCTGGATCCAGAACAAGGTGCCCCAGCTGCCCATCACCAACTTCAACCGTGACTGGCAGGAC GGCAAAGCTCTGGGCGCCCTGGTGGACAACTGCGCCCCCGGTCTCTGCCCCGACTGGGAGGCCTGGGACC CCAACCAGCCCGTGGAGAACGCCCGGGAGGCCATGCAGCAGGCCGACGACTGGCTTGGGGTGCCCCAGGT CATTGCCCCTGAGGAGATTGTGGACCCCAACGTGGATGAGCATTCTGTTATGACCTACCTGTCCCAGTTC CCCAAGGCCAAGCTCAAACCTGGTGCCCCTGTTCGATCCAAGCAGCTGAACCCCAAGAAAGCCATCGCCT ATGGGCCTGGCATCGAGCCACAGGGCAACACCGTGCTGCAGCCTGCCCACTTCACCGTGCAGACGGTGGA CGCGGGCGTGGGCGAGGTGCTGGTCTACATCGAGGACCCTGAAGGCCACACCGAGGAGGCTAAGGTGGTT CCCAACAATGACAAGGATCGCACCTATGCTGTCTCCTATGTGCCCAAGGTCGCTGGGTTACACAAGGTGA CCGTGCTCTTTGCTGGCCAGAACATTGAACGCAGTCCCTTTGAGGTGAACGTGGGCATGGCCCTGGGAGA TGCCAACAAGGTGTCAGCCCGTGGCCCTGGCCTGGAACCTGTGGGCAATGTGGCCAACAAACCCACCTAC TTTGACATCTACACTGCGGGGGCCGGCACTGGCGATGTTGCTGTGGTGATCGTGGACCCACAGGGCCGGC GGGACACAGTGGAGGTGGCCCTGGAGGACAAGGGTGACAGCACGTTCCGCTGCACATACAGACCTGCCAT GGAGGGGCCACATACCGTGCATGTGGCCTTTGCGGGTGCCCCCATCACCCGCAGTCCCTTCCCTGTCCAT GTGTCGGAAGCCTGTAACCCCAACGCCTGCCGCGCCTCTGGGCGAGGCCTGCAGCCCAAGGGTGTTCGCG TGAAAGAGGTGGCTGACTTCAAGGTGTTTACCAAGGGTGCCGGCAGCGGGGAGCTCAAGGTCACGGTCAA GGGGCCAAAGGGCACAGAGGAGCCAGTGAAGGTGCGGGAGGCTGGGGATGGTGTGTTCGAGTGCGAGTAC TACCCGGTGGTGCCTGGGAAGTATGTGGTGACCATCACGTGGGGCGGCTACGCCATCCCTCGCAGCCCCT TTGAGGTACAGGTGAGCCCAGAGGCAGGAGTGCAAAAGGTCCGGGCCTGGGGTCCTGGTTTGGAGACTGG CCAGGTGGGCAAGTCAGCCGATTTTGTGGTGGAAGCCATTGGCACCGAGGTGGGGACACTGGGCTTCTCC ATCGAGGGGCCCTCACAAGCCAAGATCGAATGTGACGACAAGGGGGATGGCTCCTGCGATGTGCGGTACT GGCCCACGGAGCCTGGGGAGTACGCTGTGCACGTCATCTGTGACGATGAGGACATCCGAGACTCACCCTT CATTGCCCACATCCTGCCCGCCCCACCTGACTGCTTCCCAGATAAGGTGAAGGCCTTTGGGCCTGGCCTG GAGCCTACCGGCTGCATCGTGGACAAGCCCGCTGAGTTCACCATTGATGCTCGTGCAGCTGGCAAGGGAG ACCTGAAGCTCTATGCCCAGGACGCCGACGGCTGTCCCATCGACATCAAGGTGATCCCCAACGGCGACGG CACCTTCCGCTGCTCCTACGTGCCCACCAAGCCCATTAAGCACACCATCATCATCTCCTGGGGAGGCGTA AACGTGCCCAAGAGCCCCTTCCGGGTGAACGTGGGCGAGGGCAGCCACCCCGAGCGGGTAAAGGTGTACG GCCCCGGAGTGGAGAAGACAGGCCTCAAGGCCAATGAGCCCACCTACTTCACGGTGGACTGCAGCGAGGC GGGGCAAGGCGACGTGAGCATCGGCATCAAGTGCGCCCCAGGCGTGGTGGGCCCTGCAGAGGCTGACATT GACTTCGACATCATCAAGAATGACAACGACACCTTCACCGTCAAGTACACGCCACCAGGGGCGGGCCGCT ACACCATCATGGTGCTGTTTGCCAACCAGGAGATCCCCGCCAGCCCCTTCCACATCAAGGTGGACCCATC CCACGATGCCAGCAAAGTCAAGGCCGAGGGCCCTGGGCTGAATCGCACAGGTGTGGAAGTCGGGAAGCCC ACCCACTTCACGGTGCTGACCAAGGGAGCCGGCAAGGCCAAGCTGGATGTGCAGTTTGCAGGGACAGCCA AGGGCGAGGTTGTGCGGGACTTTGAGATCATAGACAACCATGACTACTCCTACACTGTCAAGTACACCGC TGTCCAGCAGGGCAACATGGCAGTGACAGTGACTTATGGCGGGGACCCTGTCCCCAAGAGCCCCTTTGTG GTGAATGTGGCACCCCCGCTGGACCTCAGCAAAATCAAAGTTCAGGGCCTTAATAGCAAGGTGGCTGTGG GACAGGAACAAGCATTCTCTGTGAACACACGAGGGGCTGGCGGTCAGGGCCAACTGGATGTGCGGATGAC TTCGCCCTCTCGCCGGCCCATCCCCTGCAAGCTGGAGCCAGGCGGTGGAGCGGAAGCCCAGGCTGTGCGC TACATGCCCCCGGAGGAGGGGCCCTACAAGGTGGATATCACCTACGATGGTCACCCGGTGCCTGGCAGCC CGTTTGCTGTGGAGGGTGTCCTGCCCCCTGATCCCTCCAAGGTCTGTGCTTATGGCCCGGGTCTCAAGGG TGGACTGGTAGGCACCCCCGCGCCATTCTCCATCGACACCAAGGGGGCTGGCACAGGTGGCCTGGGGCTG ACCGTAGAGGGCCCCTGCGAGGCCAAGATCGAGTGCCAGGACAATGGTGATGGCTCATGTGCTGTCAGCT ACCTGCCCACGGAGCCTGGCGAGTACACCATCAACATCCTGTTTGCTGAGGCCCACATCCCTGGCTCGCC CTTCAAAGCCACCATTCGGCCTGTGTTTGACCCGAGCAAGGTGCGGGCCAGTGGACCGGGCCTGGAGCGC GGCAAGGTCGGTGAGGCAGCCACCTTCACTGTGGACTGCTCAGAGGCAGGCGAGGCGGAGCTGACCATTG AGATCCTGTCGGATGCCGGGGTCAAGGCCGAGGTGCTGATCCACAACAACGCGGATGGCACCTACCACAT CACCTACAGCCCTGCCTTCCCTGGCACCTACACCATTACCATCAAGTATGGCGGGCATCCCGTGCCCAAA TTCCCCACCCGTGTCCATGTGCAGCCTGCGGTCGATACCAGTGGCGTCAAGGTCTCAGGGCCTGGTGTTG AGCCACACGGTGTCCTGCGGGAGGTGACCACTGAGTTCACTGTGGATGCAAGATCCCTAACAGCCACAGG CGGCAACCACGTGACGGCTCGTGTGCTCAACCCCTCGGGGGCCAAGACAGACACCTATGTGACAGACAAT GGGGACGGCACCTACCGAGTGCAGTACACCGCCTACGAGGAGGGCGTGCATCTGGTGGAGGTCCTGTATG ATGAGGTCGCTGTGCCCAAGAGCCCCTTCCGAGTGGGCGTGACCGAGGGCTGTGATCCCACCCGCGTCCG AGCCTTCGGGCCAGGCCTGGAGGGTGGCTTGGTCAACAAGGCCAACCGATTCACTGTGGAGACCAGGGGA GCGGGCACCGGGGGCCTTGGCCTAGCCATCGAGGGTCCCTCGGAAGCCAAGATGTCCTGCAAGGACAACA AGGATGGTAGCTGCACCGTGGAGTACATCCCCTTCACTCCTGGAGACTATGACGTCAACATCACCTTCGG GGGGCGGCCCATCCCAGGGAGCCCGTTCCGCGTGCCAGTGAAGGATGTGGTGGACCCTGGGAAGGTGAAG TGCTCAGGGCCAGGGCTGGGGGCTGGTGTCAGGGCCCGGGTTCCTCAGACCTTCACAGTGGACTGCAGTC AAGCTGGCCGGGCGCCCCTGCAGGTGGCTGTGCTGGGCCCCACAGGTGTGGCCGAGCCTGTGGAGGTGCG GGACAATGGAGATGGCACCCACACTGTCCACTACACCCCAGCCACTGACGGGCCCTACACGGTAGCCGTC AAGTATGCTGACCAGGAGGTGCCACGCAGCCCCTTCAAGATCAAGGTCCTCCCAGCTCATGATGCCAGCA AGGTGCGGGCCAGCGGCCCAGGCCTCAACGCCTCTGGCATCCCTGCCAGCCTGCCTGTGGAGTTCACCAT CGACGCACGGGACGCGGGCGAGGGGTTGCTCACTGTCCAGATCTTGGACCCCGAGGGTAAGCCCAAGAAG GCCAACATCCGGGACAATGGGGATGGCACGTACACTGTGTCCTACCTGCCGGACATGAGTGGCCGGTACA CCATCACCATCAAGTATGGCGGTGATGAGATCCCCTACTCGCCCTTCCGCATCCATGCTCTGCCCACTGG GGATGCCAGCAAGTGCCTCGTCACAGTGTCCATTGGAGGCCATGGCCTGGGTGCCTGCCTGGGCCCTCGA ATCCAGATTGGGCAGGAGACGGTGATCACGGTGGATGCCAAGGCAGCCGGTGAGGGGAAGGTGACATGCA CGGTGTCCACGCCGGATGGGGCAGAGCTCGATGTGGATGTGGTTGAGAACCATGACGGTACCTTTGACAT CTACTACACAGCGCCCGAGCCGGGCAAGTACGTCATCACCATCCGCTTCGGGGGTGAGCACATCCCCAAC AGCCCCTTCCACGTGCTGGCGTGTGACCCCCTGCCGCACGAGGAGGAGCCCTCTGAAGTGCCACAGCTGC GCCAGCCCTACGCTCCTCCCCGGCCCGGCGCCCGCCCCACACACTGGGCCACAGAGGAGCCAGTGGTGCC TGTGGAGCCAATGGAGTCCATGCTGAGGCCCTTCAACCTGGTCATCCCCTTCGCGGTGCAGAAAGGGGAG CTCACAGGAGAGGTGCGGATGCCCTCGGGGAAGACGGCACGGCCCAACATCACCGACAACAAGGACGGCA CCATCACGGTGAGGTATGCACCCACTGAGAAAGGCCTGCACCAGATGGGGATCAAGTATGACGGCAACCA CATCCCTGGGAGCCCCTTACAGTTCTATGTGGATGCCATCAACAGCCGCCATGTCAGTGCCTATGGGCCA GGCCTGAGCCATGGCATGGTCAACAAGCCAGCCACCTTCACTATTGTCACCAAAGATGCTGGAGAAGGGG GTCTGTCACTGGCCGTGGAGGGCCCATCCAAGGCAGAGATCACCTGTAAGGACAACAAGGATGGCACCTG CACCGTGTCCTATCTGCCGACTGCGCCTGGAGACTACAGCATCATCGTGCGCTTCGATGACAAGCACATC CCGGGGAGCCCCTTCACAGCCAAGATCACAGGTGATGACTCCATGAGGACCTCACAGCTGAATGTGGGCA CCTCCACGGACGTGTCACTGAAGATCACCGAGAGTGATCTGAGCCAGCTGACCGCCAGCATCCGTGCCCC CTCGGGCAACGAGGAGCCCTGCCTGCTGAAGCGCCTGCCCAACCGGCACATTGGGATCTCCTTCACCCCC AAGGAGGTCGGGGAGCACGTGGTGAGCGTGCGCAAGAGTGGCAAGCATGTCACCAACAGCCCCTTCAAGA TCCTGGTGGGGCCATCTGAGATCGGGGACGCCAGCAAGGTGCGGGTCTGGGGCAAGGGGCTTTCCGAGGG ACACACATTCCAGGTGGCAGAGTTCATCGTGGACACTCGCAATGCAGGTTATGGGGGCTTGGGGCTGAGT ATTGAAGGCCCAAGCAAGGTGGACATCAACTGTGAGGACATGGAGGACGGGACATGCAAAGTCACCTACT GCCCCACCGAGCCCGGCACCTACATCATCAACATCAAGTTTGCTGACAAGCACGTGCCTGGAAGCCCCTT CACTGTGAAGGTGACCGGCGAGGGCCGCATGAAGGAGAGCATCACCCGGCGGAGACAGGCACCTTCCATC GCCACCATCGGCAGCACCTGTGACCTCAACCTCAAGATCCCAGGAAACTGGTTCCAGATGGTGTCTGCCC AGGAGCGCCTGACACGCACCTTCACACGCAGCAGCCACACCTACACCCGCACGGAGCGCACGGAGATCAG CAAGACGCGGGGCGGGGAGACAAAGCGCGAGGTGCGGGTGGAGGAGTCCACCCAGGTCGGCGGGGACCCC TTCCCTGCTGTGTTTGGGGACTTCCTGGGCCGGGAGCGCCTGGGATCCTTCGGCAGCATCACCCGGCAGC AGGAGGGTGAGGCCAGCTCTCAGGACATGACTGCACAGGTGACCAGCCCATCGGGCAAGGTGGAAGCCGC AGAGATCGTCGAGGGCGAGGACAGCGCCTACAGCGTGCGCTTTGTGCCCCAGGAAATGGGGCCCCATACG GTCGCTGTCAAGTACCGTGGCCAGCACGTGCCCGGCAGCCCCTTTCAGTTCACTGTGGGGCCGCTGGGTG AAGGTGGTGCCCACAAGGTGCGGGCCGGAGGCACAGGGCTGGAGCGAGGTGTGGCCGGCGTGCCAGCCGA GTTCAGCATCTGGACCCGGGAGGCTGGCGCTGGGGGCCTGTCCATTGCTGTGGAGGGTCCTAGCAAAGCG GAGATTGCATTTGAGGATCGCAAAGATGGCTCCTGCGGCGTCTCCTATGTCGTCCAGGAACCAGGTGACT ATGAGGTCTCCATCAAGTTCAATGATGAGCACATCCCAGACAGCCCCTTTGTGGTGCCTGTGGCCTCCCT CTCGGATGACGCTCGCCGTCTCACTGTCACCAGCCTCCAGGAGACGGGGCTCAAGGTGAACCAGCCAGCG TCCTTTGCCGTGCAGCTGAACGGTGCCCGGGGCGTGATTGATGCCCGGGTGCACACACCCTCGGGGGCTG TGGAGGAGTGCTACGTCTCTGAGCTGGACAGTGACAAGCACACCATCCGCTTCATCCCCCACGAGAATGG CGTCCACTCCATCGATGTCAAGTTCAACGGTGCCCACATCCCTGGAAGTCCCTTCAAGATCCGCGTTGGG GAGCAGAGCCAGGCTGGGGACCCAGGCTTGGTGTCAGCCTACGGTCCTGGGCTCGAGGGAGGCACTACCG GTGTGTCATCAGAGTTCATCGTGAACACCCTGAATGCCGGCTCGGGGGCCTTGTCTGTCACCATTGATGG CCCCTCCAAGGTGCAGCTGGACTGTCGGGAGTGTCCTGAGGGCCATGTGGTCACTTATACTCCCATGGCC CCTGGCAACTACCTCATTGCCATCAAGTACGGTGGCCCCCAGCACATCGTGGGCAGCCCCTTCAAGGCCA AGGTCACTGGTCCGAGGCTGTCCGGAGGCCACAGCCTTCACGAAACATCCACGGTTCTGGTGGAGACTGT GACCAAGTCCTCCTCAAGCCGGGGCTCCAGCTACAGCTCCATCCCCAAGTTCTCCTCAGATGCCAGCAAG GTGGTGACTCGGGGCCCTGGGCTGTCCCAGGCCTTCGTGGGCCAGAAGAACTCCTTCACCGTGGACTGCA GCAAAGCAGGCACCAACATGATGATGGTGGGCGTGCACGGCCCCAAGACCCCCTGTGAGGAGGTGTACGT GAAGCACATGGGGAACCGGGTGTACAATGTCACCTACACTGTCAAGGAGAAAGGGGACTACATCCTCATT GTCAAGTGGGGTGACGAAAGTGTCCCTGGAAGCCCCTTCAAAGTCAAGGTCCCTTGAATCCCAAAAGTGC CTCCCCAGCCTCAGCCCCCACCTCCAGCCACACACACATTACACACACACACACACACACACAAATGTGC CACACCCAGACACGCACAGAATCAGACACTACAAACACCTGCCTTGGGGGTGAAGTGAAGGCCCAGCCTC CCCACCCCACCGCGCCCCAGGGGTTGGAGGACCTTGTCTGTGTCAGGACAGTGTCCCTCCCTGGGAATGT GACATGAGGGCCGACTGGGGCCAGGCTCAGGGGCAGAGGCTGGGACACAAGGGGCTGGCGAGGGCTGCGA GGCCAGGGAAGCCCTGAGTTTCTGGCGGGGCTGAGCAGTGGGGGAGCATTGTGTTGTGGGTGTCTGTGTG TGAGGTCACCCTCAAACTGCACCGCCGGCCAGATACCCTCCTGACCCCGAGGACTTGGTCTGGTCTCTCT GGTGGCTACAACCCCAGAGTTTTAAGGACTTGGAAAGGAAAGCACAATCAGAGAAGAAAACAGCCCCCGA ACCAGCAGGAGTGGCCTGGCACATGGACCGGCCTGAGCGATGTGCACTCCACCCAAGCCAGGCTCCCAGG GGGCCTGATTTCTCTCTCACTGTCTCTTTTTTTAAAATGGTTGCACGGCTCTGCCCCATGGGGGGCCTTT TTTACACACTGCGAGGCCCAGCTTTCTAGGGGACTTTTGCACATGTCATGCAGCTCAGCTGGGAGCTGCT TAGGTGGAAAACTCCAAATAAAGTGCGGCTGTCGCAGAAAAAAAAAAAA Amino Acid Sequence
GenBank: BAG48314.1
SEQ ID NO: 36
MMNNSGYSDAGLGLGDETDEMPSTEKDLAEDAPWKKIQQNTFTRWCNEHLKCVGKRLTDLQRDLSDGLRL IALLEVLSQKRMYRKFHPRPNFRQMKLENVSVALEFLEREHIKLVSIDSKAIVDGNLKLILGLIWTLILH YSISMPMWEDEDDEDARKQTPKQRLLGWIQNKVPQLPITNFNRDWQDGKALGALVDNCAPGLCPDWEAWD PNQPVENAREAMQQADDWLGVPQVIAPEEIVDPNVDEHSVMTYLSQFPKAKLKPGAPVRSKQLNPKKAIA YGPGIEPQGNTVLQPAHFTVQTVDAGVGEVLVYIEDPEGHTEEAKWPNNDKDRTYAVSYVPKVAGLHKV TVLFAGQNIERSPFEVNVGMALGDANKVSARGPGLEPVGNVANKPTYFDIYTAGAGTGDVAWIVDPQGR RDTVEVALEDKGDSTFRCTYRPAMEGPHTVHVAFAGAPITRSPFPVHVSEACNPNACRASGRGLQPKGVR VKEVADFKVFTKGAGSGELKVTVKGPKGTEEPVKVREAGDGVFECEYYPWPGKYWTITWGGYAIPRSP FEVQVSPEAGVQKVRAWGPGLETGQVGKSADFWEAIGTEVGTLGFSIEGPSQAKIECDDKGDGSCDVRY WPTEPGEYAVHVICDDEDIRDSPFIAHILPAPPDCFPDKVKAFGPGLEPTGCIVDKPAEF IDARAAGKG DLKLYAQDADGCPIDIKVIPNGDGTFRCSYVPTKPIKHTI I ISWGGVNVPKSPFRVNVGEGSHPERVKVY GPGVEKTGLKANEPTYFTVDCSEAGQGDVSIGIKCAPGWGPAEADIDFDI IKNDNDTFTVKYTPPGAGR YTIMVLFANQEIPASPFHIKVDPSHDASKVKAEGPGLNRTGVEVGKPTHFTVLTKGAGKAKLDVQFAGTA KGEWRDFEI IDNHDYSYTVKYTAVQQGNMAVTVTYGGDPVPKSPFWNVAPPLDLSKIKVQGLNSKVAV GQEQAFSVNTRGAGGQGQLDVRMTSPSRRPIPCKLEPGGGAEAQAVRYMPPEEGPYKVDITYDGHPVPGS PFAVEGVLPPDPSKVCAYGPGLKGGLVGTPAPFSIDTKGAGTGGLGLTVEGPCEAKIECQDNGDGSCAVS YLPTEPGEYTINILFAEAHIPGSPFKATIRPVFDPSKVRASGPGLERGKVGEAATFTVDCSEAGEAELTI EILSDAGVKAEVLIHNNADGTYHITYSPAFPGTYTITIKYGGHPVPKFPTRVHVQPAVDTSGVKVSGPGV EPHGVLREVTTEFTVDARSLTATGGNHVTARVLNPSGAKTDTYVTDNGDGTYRVQYTAYEEGVHLVEVLY DEVAVPKSPFRVGVTEGCDPTRVRAFGPGLEGGLVNKANRFTVETRGAGTGGLGLAIEGPSEAKMSCKDN KDGSCTVEYIPFTPGDYDVNITFGGRPIPGSPFRVPVKDWDPGKVKCSGPGLGAGVRARVPQTFTVDCS QAGRAPLQVAVLGPTGVAEPVEVRDNGDGTHTVHY PATDGPYTVAVKYADQEVPRSPFKIKVLPAHDAS KVRASGPGLNASGIPASLPVEFTIDARDAGEGLLTVQILDPEGKPKKANIRDNGDGTYTVSYLPDMSGRY TITIKYGGDEIPYSPFRIHALPTGDASKCLVTVSIGGHGLGACLGPRIQIGQETVITVDAKAAGEGKVTC TVSTPDGAELDVDWENHDGTFDIYYTAPEPGKYVITIRFGGEHIPNSPFHVLACDPLPHEEEPSEVPQL RQPYAPPRPGARPTHWATEEPWPVEPMESMLRPFNLVIPFAVQKGELTGEVRMPSGKTARPNITDNKDG TITVRYAPTEKGLHQMGIKYDGNHIPGSPLQFYVDAINSRHVSAYGPGLSHGMVNKPATFTIVTKDAGEG GLSLAVEGPSKAEI CKDNKDGTCTVSYLPTAPGDYSI IVRFDDKHIPGSPFTAKI GDDSMR SQLNVG TSTDVSLKITESDLSQLTASIRAPSGNEEPCLLKRLPNRHIGISFTPKEVGEHWSVRKSGKHVTNSPFK ILVGPSEIGDASKVRVWGKGLSEGHTFQVAEFIVDTRNAGYGGLGLSIEGPSKVDINCEDMEDGTCKVTY CPTEPGTYI I IKFADKHVPGSPFTVKVTGEGRMKESI RRRQAPSIA IGS CDLNLKIPGNWFQMVSA QERLTRTFTRSSHTYTRTERTEISKTRGGETKREVRVEESTQVGGDPFPAVFGDFLGRERLGSFGSITRQ QEGEASSQDMTAQVTSPSGKVEAAEIVEGEDSAYSVRFVPQEMGPHTVAVKYRGQHVPGSPFQFTVGPLG EGGAHKVRAGGTGLERGVAGVPAEFSIWTREAGAGGLSIAVEGPSKAEIAFEDRKDGSCGVSYWQEPGD YEVSIKFNDEHIPDSPFWPVASLSDDARRLTVTSLQETGLKVNQPASFAVQLNGARGVIDARVHTPSGA VEECYVSELDSDKHTIRFIPHENGVHSIDVKFNGAHIPGSPFKIRVGEQSQAGDPGLVSAYGPGLEGGTT GVSSEFIVNTLNAGSGALSVTIDGPSKVQLDCRECPEGHWTYTPMAPGNYLIAIKYGGPQHIVGSPFKA KVTGPRLSGGHSLHETSTVLVETVTKSSSSRGSSYSSIPKFSSDASKWTRGPGLSQAFVGQKNSFTVDC SKAGTNMMMVGVHGPKTPCEEVYVKHMGNRVYNVTYTVKEKGDYILIVKWGDESVPGSPFKVKVP
Biomarker: Myotilin 1
mRNA Sequence
NCBI Reference Sequence: NM_006790.2
SEQ ID NO: 37
AAGCACAGAGCCACTAGATTAGTCTGTGAGGGAAGGAGATGCCTCTTCCTTCCCTTCAATAGTGGGTTAA ACCCAGCTGGCACCCTCTGGAACTACGGGAACAATATTCTTCAAGAGAAGGTCACTCTACCAAAGCCAGG AGCACAGTATTCTCAGGATCTCAACAAGGAAGAGCAGACCAAGGTTGCTTCTGATTCCTTACAACCTTCC GTAATTCCAGGCTTGTGGCCCCAAATTCAGGGCCCCACCCTTCCAGGAACAAATCATTATAGTAATAATT TGCCTTCATCTTCCATATACCAACTAAGCATGTTTAACTACGAACGTCCAAAACACTTCATCCAGTCCCA AAACCCATGTGGCTCCAGATTGCAGCCTCCTGGACCAGAAACCTCCAGCTTCTCTAGCCAGACCAAACAG TCTTCCATTATCATCCAGCCCCGCCAGTGTACAGAGCAAAGATTTTCTGCCTCCTCAACACTGAGCTCTC ACATCACCATGTCCTCCTCTGCTTTCCCTGCTTCTCCCCAGCAGCATGCTGGCTCCAACCCAGGCCAAAG GGTTACAACCACCTATAACCAGTCCCCAGCCAGCTTCCTCAGCTCCATATTACCATCACAGCCTGATTAC AATAGCAGTAAAATCCCTTCCGCTATGGATTCCAACTATCAACAGTCCTCAGCTGGCCAACCTATAAATG CAAAGCCATCCCAAACTGCAAATGCTAAGCCCATACCAAGAACTCCTGATCATGAAATACAAGGATCAAA AGAAGCTTTGATTCAAGATTTGGAAAGAAAGCTGAAATGCAAGGACACCCTTCTTCATAATGGAAATCAA CGTCTAACATATGAAGAGAAGATGGCTCGCAGATTGCTAGGACCACAGAATGCAGCTGCTGTGTTTCAAG CTCAGGATGACAGTGGTGCACAAGACTCGCAGCAACACAACTCAGAACATGCGCGACTGCAAGTTCCTAC ATCACAAGTAAGAAGTAGATCAACCTCAAGGGGAGATGTGAATGATCAGGATGCAATCCAGGAGAAATTT ACCCACCACGTTTCATTCAAGTGCCAGAGAACATGTCGATTGATGAAGGAAGATTCTGCAGAATGGACT TCAAAGTGAGTGGACTGCCAGCTCCTGATGTGTCATGGTATCTAAATGGAAGAACAGTTCAATCAGATGA TTTGCACAAAATGATAGTGTCTGAGAAGGGTCTTCATTCACTCATCTTTGAAGTAGTCAGAGCTTCAGAT GCAGGGGCTTATGCATGTGTTGCCAAGAATAGAGCAGGAGAAGCCACCTTCACTGTGCAGCTGGATGTCC TTGCAAAAGAACATAAAAGAGCACCAATGTTTATCTACAAACCACAGAGCAAAAAAGTTTTAGAGGGAGA TTCAGTGAAACTAGAATGCCAGATCTCGGCTATACCTCCACCAAAGCTTTTCTGGAAAAGAAATAATGAA ATGGTACAATTCAACACTGACCGAATAAGCTTATATCAAGATAACACTGGAAGAGTTACTTTACTGATAA AAGATGTAAACAAGAAAGATGCTGGGTGGTATACTGTGTCAGCAGTTAATGAAGCTGGAGTGACTACATG TAACACAAGATTAGACGTTACGGCACGTCCAAACCAAACTCTTCCAGCTCCTAAGCAGTTACGGGTTCGA CCAACATTCAGCAAATATTTAGCACTTAATGGGAAAGGTTTGAATGTAAAACAAGCTTTTAACCCAGAAG GAGAATTTCAGCGTTTGGCAGCTCAATCTGGACTCTATGAAAGTGAAGAACTTTAATAACTTTACCAACA TTGGAAAACAGCCAACTACACCATTAGTAATATATTTGATTACATTTTTTTGAAATTAATCCATAGCTGT ATTAACAGATTATGGTTTTAATTAGGTAATATAGTTAATATATATTTATAATATTATTTATCCTTTGACT CTTGCACATTCTATGTACCCCTCCGATTTGTGAAGCCTACAGGAAATCTGGGTATATGGATTTGTAACTG CAGAAGACTATCTTAAAATACAGGATTTTAACATTTAAGTCATGCACATTTAACAATTACAGGTTATAAA TTAGTATCAACTTTTTAAACACATCTAATGCTTGTAATAACGTTTACTGGTACTGCTTTCTAAATACTGT TTTACCCGTTTTCTCTTGTAGGAATACTAACATGGTATAGATTATCTGAGTGTTCCACAGTTGTATGTCA AAAGAAAATAAAATTCAAATATTTAAAACGGAAAAAAAAAAAAAAAAAAA
Amino Acid Sequence
NCBI Reference Sequence: NP_001287840.1
SEQ ID NO: 38
MDSNYQQSSAGQPINAKPSQTANAKPIPRTPDHEIQGSKEALIQDLERKLKCKDTLLHNGNQRLTYEEKM ARRLLGPQNAAAVFQAQDDSGAQDSQQHNSEHARLQVPTSQVRSRSTSRGDVNDQDAIQEKFYPPRFIQV PENMSIDEGRFCRMDFKVSGLPAPDVSWYLNGRTVQSDDLHKMIVSEKGLHSLIFEWRASDAGAYACVA KNRAGEATFTVQLDVLAKEHKRAPMFIYKPQSKKVLEGDSVKLECQISAIPPPKLFWKRNNEMVQFNTDR ISLYQDNTGRVTLLIKDVNKKDAGWYTVSAVNEAGVTTCNTRLDVTARPNQTLPAPKQLRVRPTFSKYLA LNGKGLNVKQAFNPEGEFQRLAAQSGLYESEEL
Biomarker: Leiomodin-2
mRNA Sequence
NCBI Reference Sequence: NM_207163.1
SEQ ID NO: 39
GCATTGTCACAGCCCTGTATCACCACTCTTAAAAGGCTCCCACAGCCACTCCTAGCACCAGTTGTTGACC AGCCTGCCACTTGCCTCCCTGCCTGCTTCTGGCCGCCTTGAATGCCTGGTCCTTCAAGCTCCTTCTGGGT CTGACAAAGCAGGGACCATGTCTACCTTTGGCTACCGAAGAGGACTCAGTAAATACGAATCCATCGACGA GGATGAACTCCTCGCCTCCCTGTCAGCCGAGGAGCTGAAGGAGCTAGAGAGAGAGTTGGAAGACATTGAA CCTGACCGCAACCTTCCCGTGGGGCTAAGGCAAAAGAGCCTGACAGAGAAAACCCCCACAGGGACATTCA GCAGAGAGGCACTGATGGCCTATTGGGAAAAGGAGTCCCAAAAACTCTTGGAGAAGGAGAGGCTGGGGGA ATGTGGAAAGGTTGCAGAAGACAAAGAGGAAAGTGAAGAAGAGCTTATCTTTACTGAAAGTAACAGTGAG GTTTCTGAGGAAGTGTATACAGAGGAGGAGGAGGAGGAGTCCCAGGAGGAAGAGGAGGAAGAAGACAGTG ACGAAGAGGAAAGAACAATTGAAACTGCAAAAGGGATTAATGGAACTGTAAATTATGATAGTGTCAATTC TGACAACTCTAAGCCAAAGATATTTAAAAGTCAAATAGAGAACATAAATTTGACCAATGGCAGCAATGGG AGGAACACAGAGTCCCCAGCTGCCATTCACCCTTGTGGAAATCCTACAGTGATTGAGGACGCTTTGGACA AGATTAAAAGCAATGACCCTGACACCACAGAAGTCAATTTGAACAACATTGAGAACATCACAACACAGAC CCTTACCCGCTTTGCTGAAGCCCTCAAGGACAACACTGTGGTGAAGACGTTCAGTCTGGCCAACACGCAT GCCGACGACAGTGCAGCCATGGCCATTGCAGAGATGCTCAAAGTCAATGAGCACATCACCAACGTAAACG TCGAGTCCAACTTCATAACGGGAAAGGGGATCCTGGCCATCATGAGAGCTCTCCAGCACAACACGGTGCT CACGGAGCTGCGTTTCCATAACCAGAGGCACATCATGGGCAGCCAGGTGGAAATGGAGATTGTCAAGCTG CTGAAGGAGAACACGACGCTGCTGAGGCTGGGATACCATTTTGAACTCCCAGGACCAAGAATGAGCATGA CGAGCATTTTGACAAGAAATATGGATAAACAGAGGCAAAAACGTTTGCAGGAGCAAAAACAGCAGGAGGG ATACGATGGAGGACCCAATCTTAGGACCAAAGTCTGGCAAAGAGGAACACCTAGCTCTTCACCTTATGTA TCTCCCAGGCACTCACCCTGGTCATCCCCAAAACTCCCCAAAAAAGTCCAGACTGTGAGGAGCCGTCCTC TGTCTCCTGTGGCCACACCTCCTCCTCCTCCCCCTCCTCCTCCTCCTCCCCCTCCTTCTTCCCAAAGGCT GCCACCACCTCCTCCTCCTCCCCCTCCTCCACTCCCAGAGAAAAAGCTCATTACCAGAAACATTGCAGAA GTCATCAAACAACAGGAGAGTGCCCAACGGGCATTACAAAATGGACAAAAAAAGAAAAAAGGGAAAAAGG TCAAGAAACAGCCAAACAGTATTCTAAAGGAAATAAAAAATTCTCTGAGGTCAGTGCAAGAGAAGAAAAT GGAAGACAGTTCCCGACCTTCTACCCCACAGAGATCAGCTCATGAGAATCTCATGGAAGCAATTCGGGGA AGCAGCATAAAACAGCTAAAGCGGGTGGAAGTTCCAGAAGCCCTGCGATAAAAACATGATCTTTAGAAGA GGATGCAGAACTGTTCAGTGGTATTACATGAAATGCATTGTGAGATGTTTCTAAAATACCTTCTTCAATT CAAAATGATCCCTGACTTTAAAAATAATCTCACCCATTAATTCCAAAGAGAATCTTAAGAAACAATCAGC ATGTTTCTTCTGTAAATATGAAAATAAATTTCTTTTTTATGTCGTGAGATTTGTATTGGCAAGAAGCAGT TAATTTAAAGATGCTCTTCCTATCTGTGGATGTGTTGGTAACTCCGAGTTGTAATGAGTTCATGAAATGT GCTGTTATTTTTGTAATCTCAATAAATGTGGATTGAAGTTTTTTCCCTTCCAAAAAAAAAAAAAAAAAAA AAAAAAAAAAA
Amino Acid Sequence
NCBI Reference Sequence: NP_997046.1
SEQ ID NO: 40 MSTFGYRRGLSKYESIDEDELLASLSAEELKELERELEDIEPDRNLPVGLRQKSLTEKTPTGTFSREALM AYWEKESQKLLEKERLGECGKVAEDKEESEEELIFTESNSEVSEEVYTEEEEEESQEEEEEEDSDEEERT IETAKGINGTVNYDSVNSDNSKPKIFKSQIENINLTNGSNGRNTESPAAIHPCGNPTVIEDALDKIKSND PDTTEVNLNNIENITTQTLTRFAEALKDNTWKTFSLANTHADDSAAMAIAEMLKVNEHITNVNVESNFI TGKGILAIMRALQHNTVLTELRFHNQRHIMGSQVEMEIVKLLKENTTLLRLGYHFELPGPRMSMTSILTR NMDKQRQKRLQEQKQQEGYDGGPNLRTKVWQRGTPSSSPYVSPRHSPWSSPKLPKKVQTVRSRPLSPVAT PPPPPPPPPPPPPSSQRLPPPPPPPPPPLPEKKLITRNIAEVIKQQESAQRALQNGQKKKKGKKVKKQPN SILKEIKNSLRSVQEKKMEDSSRPSTPQRSAHENLMEAIRGSSIKQLKRVEVPEALR
Incorporation by Reference
All of the U.S. patents and U.S. patent application publications cited herein are hereby incorporated by reference.
Equivalents
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

Claims:
1. A method of determining whether a subject is afflicted with a cardiac disorder, the method comprising:
a) determining the level of expression or level of activity of one or more biomarkers selected from glycogen synthase kinase 3β (GSK-3P) and the phosphorylation targets of GSK-3P in a biological sample obtained from the subject;
b) determining the level of expression or level of activity of the one or more biomarkers in a control sample; and
c) comparing the level of expression or level of activity of said one or more biomarkers detected in steps a) and b);
wherein a significant decrease in the level of expression or level of activity of the one or more biomarkers in the subject sample relative to the control level of expression or level of activity of the one or more biomarkers indicates that the subject is afflicted with the cardiac disorder.
2. A method of determining whether a subject afflicted with a cardiac disorder or at risk for developing a cardiac disorder would benefit from cardiac resynchronization therapy (CRT) or enhanced activity of GSK-3P, the method comprising:
a) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P in a biological sample obtained from the subject;
b) determining the level of expression or level of activity of the one or more biomarkers in a control sample; and
c) comparing the level of expression or level of activity of said one or more biomarkers detected in steps a) and b), optionally wherein between the first point in time and the subsequent point in time, the subject has undergone treatment for the cardiac disorder; wherein a significant decrease in the level of expression or level of activity of the one or more biomarkers in the subject sample relative to the control level of expression or level of activity of the one or more biomarkers indicates that the subject afflicted with the cardiac disorder or at risk for developing the cardiac disorder would benefit from CRT or enhanced activity of GSK-3P.
3. A method for monitoring the progression of a cardiac disorder in a subject, the method comprising:
a) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P in a biological sample obtained from the subject at a first point;
b) repeating step a) at a subsequent point in time; and
c) comparing the level of expression or level of activity of said one or more biomarkers detected in steps a) and b) to monitor the progression of the cardiac disorder.
4. A method for stratifying subjects afflicted with a cardiac disorder according to predicted clinical outcome of treatment with CRT or one or more GSK-3P modulators, optionally wherein the predicted clinical outcome is (a) restored mechanical cardiac synchrony, (b) decreased morbidity, or (c) increased survival time resulting from treatment with one or more modulators of GSK-3P, the method comprising:
a) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P, in a biological sample obtained from the subject;
b) determining the level of expression or level of activity of the one or more biomarkers in a control sample; and
c) comparing the level of expression or level of activity of said one or more biomarkers detected in steps a) and b);
wherein a significant modulation, optionally a decrease, in the level of expression or level of activity of the one or more biomarkers in the subject sample relative to the control level of expression or level of activity of the one or more biomarkers predicts the clinical outcome of the patient to treatment with CRT or one or more GSK-3P modulators.
5. The method of any one of the preceding claims, wherein:
the method comprises determining the level of expression of the one or more biomarkers in each sample; and
the level of expression is determined by:
contacting each sample, or one or more nucleic acids derived from each sample, with one or more nucleic acid arrays or one or more nucleic acid probes; or performing qPCR on each sample or by performing qPCR on one or more nucleic acids derived from each sample.
6. The method of any one of the preceding claims, wherein:
the method comprises determining the level of activity of the one or more biomarkers in each sample; and
the level of activity is determined by:
analyzing one or more biomarkers in each sample by mass spectroscopy; or contacting each sample with an antibody that specifically binds to the one or more biomarkers.
7. The method of any one of the preceding claims, further comprising treating the subject with a therapeutic agent that specifically modulates the level of expression or level of activity of the one or more biomarkers.
8. The method of claim 7, further comprising treating the subject with one or more modulators of GSK-3p.
9. The method of claim 7 or 8 wherein the modulator increases the expression level of GSK-3P or increases the phosphorylation state of GSK-3p.
10. A method of determining the efficacy of a test compound for treating a cardiac disorder in a subject, the method comprising:
a) exposing a first sample obtained from a subject to a test compound
b) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P in the first sample , optionally wherein the sample is analyzed in vivo, ex vivo, or in vitro;
c) determining the level of expression or level of activity of the one or more biomarkers in a second sample obtained from the subject, wherein the second sample is not exposed to the test compound; and
d) comparing the level of expression or level of activity of the one or more biomarkers in the first and second samples,
wherein the test compound has efficacy for treating a cardiac disorder if the level of expression or level of activity is greater in the first sample than in the second sample.
11. A method of determining the efficacy of a therapy for treating a cardiac disorder in a subject, the method comprising:
a) obtaining a first sample from a subject prior to providing at least a portion of the therapy to the subject
b) determining the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P in the first sample, optionally wherein the sample or therapy is analyzed in vivo, ex vivo, or in vitro;
c) obtaining a second sample from the subject following provision of the portion of the therapy
d) determining the level of expression or level of activity of the one or more biomarkers in the second sample; and
e) comparing the level of expression or level of activity of the one or more biomarkers in the first and second samples,
wherein the therapy has efficacy for treating a cardiac disorder if the level of expression or level of activity is greater in the second sample than in the first sample.
12. A method for identifying a compound which treats a cardiac disorder, the method comprising:
a) contacting one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P with a test compound, optionally wherein the contacting occurs in vivo, ex vivo, or in vitro; and
b) determining the effect of the test compound on the level of expression or level of activity of the one or more biomarkers to thereby identify a compound which treats the cardiac disorder.
13. The method of claim 12, wherein the one or more biomarkers is expressed on or in a cell.
14. The method of claim 13, wherein said cells are isolated from an animal model of the cardiac disorder.
15. The method of claim 13, wherein said cells are from a subject afflicted with the cardiac disorder.
16. A method for treating a cardiac disorder, the method comprising contacting a cell with an agent that modulates, optionally increases, the level of expression or level of activity of one or more biomarkers selected from GSK-3P and the phosphorylation targets of GSK-3P to thereby treat the cardiac disorder.
17. The method of claim 16, wherein the step of contacting occurs in vivo, ex vivo, or in vitro.
18. The method of claim 16 or 17, further comprising contacting the cell with an additional agent that treats the cardiac disorder.
19. The method of any one of the preceding claims, wherein the control sample is determined from a sample from a subject not afflicted with the cardiac disorder.
20. The method of any one of the preceding claims, wherein the sample consists of or comprises body fluid, cells, cell lines, histological slides, paraffin embedded tissue, fresh frozen tissue, fresh tissue, biopsies, blood, plasma, serum, buccal scrape, saliva, cerebrospinal fluid, urine, stool, mucus, or bone marrow, obtained from the subject.
21. The method of claim 20, wherein the body fluid is selected from amniotic fluid, aqueous humor, bile, blood and blood plasma, cerebrospinal fluid, cerumen and earwax, cowper's fluid or pre-ejaculatory fluid, chyle, chyme, stool, female ejaculate, interstitial fluid, intracellular fluid, lymph, menses, breast milk, mucus, pleural fluid, peritoneal fluid, pus, saliva, sebum, semen, serum, sweat, synovial fluid, tears, urine, vaginal lubrication, vitreous humor, and vomit.
22. The method of any one of the preceding claims, wherein the expression level of the one or more biomarkers is assessed by detecting the presence in the samples of a
polynucleotide molecule encoding the biomarker or a portion of said polynucleotide molecule.
23. The method of claim 22, wherein the polynucleotide molecule is a mR A, cDNA, or functional variants or fragments thereof and, optionally, wherein the step of detecting further comprises amplifying the polynucleotide molecule.
24. The method of any one of the preceding claims, wherein the expression level of the one or more biomarkers is assessed by annealing a nucleic acid probe with the sample of the polynucleotide encoding the one or more biomarkers or a portion of said polynucleotide molecule under stringent hybridization conditions.
25. The method of any one of the preceding claims, wherein the expression level of the biomarker is assessed by detecting the presence in the samples, whether phosphorylated, unphosphorylated, or both, of a protein of the biomarker, a polypeptide, or protein fragment thereof comprising said protein.
26. The method of claim 25, wherein the presence of said protein, polypeptide or protein fragment thereof is detected using a reagent which specifically binds with said protein, polypeptide or protein fragment thereof, optionally wherein the reagent is selected from an antibody, an antibody derivative, and an antibody fragment.
27. The method of any one of the preceding claims, wherein the activity level of the biomarker is assessed by determining the magnitude of modulation of the activity or expression level of downstream targets of the one or more biomarkers.
28. The method of any one of the preceding claims, wherein determining the level of expression or level of activity of the one or more biomarkers comprises determining the phosphorylation state of GSK-3P or one or more phosphorylation targets of GSK-3p.
29. The method of claim 28, wherein the phosphorylation state is determined either by mass spectroscopy or by using an antibody that specifically binds to the phosphorylated form of the protein.
30. The method of any one of the preceding claims, wherein the phosphorylation targets of GSK-3P are one or more myofilament phosphorylation targets.
31. The method of claim 30, wherein the one or more myofilament phosphorylation targets are selected from biomarkers listed in Tables 2-5.
32. The method of any one of claims 1-29, wherein the phosphorylation targets of GSK-3P are selected from Tnl, MyBPC, TnT, MLC2, cMyBP-C, cTnl, a-Tropomyosin, titin, obscurin, Actin binding LIM protein 1 (AbLIMl), Tensin-1, Thyroid hormone receptor-associated protein 3 (THRAP-3), Nestin, Sorbin and SH3 domain-containing protein 2 (SORB-2), LIM domain only protein 7 (LMO-7), LIM domain-binding protein 3 (LDB3/Cypher), Striated muscle preferentially expressed kinase (SPEG), Filamin-C, Myotilin 1, and Leiomodin-2.
33. The method of claim 32, wherein the phosphorylation targets GSK-3P are selected from titin, obscurin, AbLIMl, Tensin-1, THRAP-3, Nestin, SORB-2, LMO-7, LDB3, SPEG, Filamin-C, Myotilin 1, and Leiomodin 2.
34. The method of any one of the preceding claims, wherein the cardiac disorder is heart failure or ventricular dyssynchrony.
35. The method of any one of the preceding claims, wherein the subject is a mammal, optionally a human.
PCT/US2014/058062 2013-09-27 2014-09-29 Methods of identifying, assessing, preventing, and treating cardiac disorders using gsk-3b and phosphorylation targets thereof WO2015048653A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361883677P 2013-09-27 2013-09-27
US61/883,677 2013-09-27
US201361919100P 2013-12-20 2013-12-20
US61/919,100 2013-12-20

Publications (1)

Publication Number Publication Date
WO2015048653A1 true WO2015048653A1 (en) 2015-04-02

Family

ID=52744567

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/058062 WO2015048653A1 (en) 2013-09-27 2014-09-29 Methods of identifying, assessing, preventing, and treating cardiac disorders using gsk-3b and phosphorylation targets thereof

Country Status (1)

Country Link
WO (1) WO2015048653A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105543408A (en) * 2016-03-16 2016-05-04 宫蕊 Marker for early diagnosis of coronary heart disease
CN105907871A (en) * 2016-06-01 2016-08-31 北京泱深生物信息技术有限公司 Biomarker for diagnosis of coronary heart disease
CN109212466A (en) * 2018-09-01 2019-01-15 哈尔滨工程大学 A kind of broadband direction-finding method based on quantum dragonfly mechanism of Evolution
WO2020198233A1 (en) * 2019-03-25 2020-10-01 The Regents Of The University Of California Methods of treating tnni3-mediated cardiomyopathy

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080050314A1 (en) * 2004-02-26 2008-02-28 Bayer Healthcare Ag Diagnostics and Therapeutics for Diseases Associated With Glycogen Synthase Kinase 3 Beta (Gsk3b)
US20090041862A1 (en) * 2004-05-07 2009-02-12 Peter Robert Schofield Detecting disease association with aberrant glycogen synthase kinase 3beta expression
US20100008968A1 (en) * 2008-06-26 2010-01-14 Lampe John W Method for treating cardiovascular diseases using rho kinase inhibitor compounds
WO2013136334A2 (en) * 2012-03-14 2013-09-19 Marx Stephen Means and methods for diagnostics and therapeutics of diseases

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080050314A1 (en) * 2004-02-26 2008-02-28 Bayer Healthcare Ag Diagnostics and Therapeutics for Diseases Associated With Glycogen Synthase Kinase 3 Beta (Gsk3b)
US20090041862A1 (en) * 2004-05-07 2009-02-12 Peter Robert Schofield Detecting disease association with aberrant glycogen synthase kinase 3beta expression
US20100008968A1 (en) * 2008-06-26 2010-01-14 Lampe John W Method for treating cardiovascular diseases using rho kinase inhibitor compounds
WO2013136334A2 (en) * 2012-03-14 2013-09-19 Marx Stephen Means and methods for diagnostics and therapeutics of diseases

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LUPÓN J. ET AL.: "Combined Use of the Novel Biomarkers High-Sensitivity Troponin T and ST2 for Heart Failure Risk Stratification vs Conventional Assessment.", MAYO CLIN PROC., vol. 88, no. 3, March 2013 (2013-03-01), pages 234 - 43 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105543408A (en) * 2016-03-16 2016-05-04 宫蕊 Marker for early diagnosis of coronary heart disease
CN105543408B (en) * 2016-03-16 2019-01-22 宫蕊 coronary heart disease early diagnosis marker
CN105907871A (en) * 2016-06-01 2016-08-31 北京泱深生物信息技术有限公司 Biomarker for diagnosis of coronary heart disease
CN105907871B (en) * 2016-06-01 2019-09-06 北京泱深生物信息技术有限公司 Biomarker for diagnosis of coronary heart disease
CN109212466A (en) * 2018-09-01 2019-01-15 哈尔滨工程大学 A kind of broadband direction-finding method based on quantum dragonfly mechanism of Evolution
CN109212466B (en) * 2018-09-01 2022-09-02 哈尔滨工程大学 Quantum dragonfly evolution mechanism-based broadband direction finding method
WO2020198233A1 (en) * 2019-03-25 2020-10-01 The Regents Of The University Of California Methods of treating tnni3-mediated cardiomyopathy

Similar Documents

Publication Publication Date Title
Kim et al. Compartmentalization of a unique ADP/ATP carrier protein SFEC (Sperm Flagellar Energy Carrier, AAC4) with glycolytic enzymes in the fibrous sheath of the human sperm flagellar principal piece
US20040166530A1 (en) F-box proteins and genes
KR20110020853A (en) Methods of diagnosing rejection of a kidney allograft using genomic or proteomic expression profiling
JP6826543B2 (en) Biomolecules involved in Alzheimer&#39;s disease
KR20140142357A (en) Interrogatory cell-based assays and uses thereof
US20200206344A1 (en) Methods for modulating the interaction between ews-fli1 and baf complexes
KR20220035268A (en) Inhibitors of human ezh2, and methods of use thereof
US20200147381A1 (en) Methods for improving heart function
WO2015048653A1 (en) Methods of identifying, assessing, preventing, and treating cardiac disorders using gsk-3b and phosphorylation targets thereof
WO2021162981A2 (en) Methods and compositions for identifying castration resistant neuroendocrine prostate cancer
ES2583003T3 (en) A transcriptomic biomarker of myocarditis
US20090082303A1 (en) Drug for preventing and treating atherosclerosis
WO2007004692A1 (en) Prophylactic/therapeutic agent and diagnostic agent for non-small cell lung cancer
US11274349B2 (en) Methods for diagnosing cancer
EP1403281B1 (en) Novel ligand and dna thereof
EP1272517B1 (en) Multiprotein-complexes comprising a nmda receptor and uses thereof
EP1666606A1 (en) Method for detecting regulatory effect on controlling synoviolin activity
JP2013039111A (en) Splicing variant
US7943137B2 (en) FPRL1 ligands and use thereof
IL156973A (en) Antibodies that bind to a ubiquitination region of tsg101
EP4043484A1 (en) Somatostatin receptor
US10254283B2 (en) Biomarker for MELK activity and methods of using same
JP4445291B2 (en) Novel protein and its DNA
Stāvusis Molecular Mechanisms Underlying Rare Neuromuscular Diseases
Krupina Ubiquitin receptor protein UBASH3B: a novel regulator of mitotic progression

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14847627

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14847627

Country of ref document: EP

Kind code of ref document: A1