US20170307636A1 - Biomarker for determining mitochondrial damage in friedreich's ataxia - Google Patents

Biomarker for determining mitochondrial damage in friedreich's ataxia Download PDF

Info

Publication number
US20170307636A1
US20170307636A1 US15/646,172 US201715646172A US2017307636A1 US 20170307636 A1 US20170307636 A1 US 20170307636A1 US 201715646172 A US201715646172 A US 201715646172A US 2017307636 A1 US2017307636 A1 US 2017307636A1
Authority
US
United States
Prior art keywords
disease
frataxin
sample
mitochondrial
deficiency
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US15/646,172
Inventor
R. Mark Payne
Gregory R. Wagner
Clifford M. Babbey
P. Melanie Pride
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Indiana University
Original Assignee
Indiana University Research and Technology Corp
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 Indiana University Research and Technology Corp filed Critical Indiana University Research and Technology Corp
Priority to US15/646,172 priority Critical patent/US20170307636A1/en
Assigned to INDIANA UNIVERSITY RESEARCH AND TECHNOLOGY CORPORATION reassignment INDIANA UNIVERSITY RESEARCH AND TECHNOLOGY CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BABBEY, Clifford M., PAYNE, R. Mark, PRIDE, P. Melanie, WAGNER, Gregory R.
Publication of US20170307636A1 publication Critical patent/US20170307636A1/en
Assigned to THE TRUSTEES OF INDIANA UNIVERSITY reassignment THE TRUSTEES OF INDIANA UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: INDIANA UNIVERSITY RESEARCH AND TECHNOLOGY CORPORATION
Abandoned legal-status Critical Current

Links

Images

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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/26Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving oxidoreductase
    • C12Q1/32Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving oxidoreductase involving dehydrogenase
    • 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
    • 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
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5076Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving cell organelles, e.g. Golgi complex, endoplasmic reticulum
    • G01N33/5079Mitochondria
    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/573Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes
    • G01N33/5735Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes co-enzymes or co-factors, e.g. NAD, ATP
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2440/00Post-translational modifications [PTMs] in chemical analysis of biological material
    • G01N2440/10Post-translational modifications [PTMs] in chemical analysis of biological material acylation, e.g. acetylation, formylation, lipoylation, myristoylation, palmitoylation
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/02Screening involving studying the effect of compounds C on the interaction between interacting molecules A and B (e.g. A = enzyme and B = substrate for A, or A = receptor and B = ligand for the receptor)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/285Demyelinating diseases; Multipel sclerosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present disclosure relates generally to compositions and methods for screening for diseases or disorders associated with a deficiency in frataxin. More particularly, the present disclosure relates to methods of screening for a disease or disorder associated with a deficiency in frataxin, as well as methods of screening for therapeutic agents for use in treating a disease or disorder associated with a deficiency in frataxin, or damage from reactive oxygen species to mitochondrial proteins affecting the respiratory chain, or increased mitochondrial protein acetylation affecting mitochondrial function, as well as method of screening for therapeutic agents for use in treating a disease or disorder associated with a deficiency in frataxin.
  • FRDA Friedreich's Ataxia
  • GATTC triplet nucleotide repeat
  • Frataxin is targeted to the mitochondrial matrix, where it is known to act as an iron-binding protein and participates in the proper assembly and function of iron-sulfur cluster (ISC) dependent proteins including complexes I, II, and III of the respiratory chain and aconitase of the tricarboxylic acid (TCA) cycle.
  • ISC iron-sulfur cluster
  • lysine acetylation is a highly conserved and abundant post-translational modification within mitochondria that is responsive to nutrient availability and may contribute to the physiological adaptations of reduced caloric intake.
  • Multiple investigations have demonstrated a role for reversible mitochondrial enzyme deacetylation and, specifically, the NAD + -dependent deacetylase SIRT3, in the regulation of fatty acid oxidation, the TCA cycle, electron transport via respiratory complexes I and II, and overall oxidative metabolism.
  • SIRT3-mediated deacetylation has recently emerged as a major mechanism regulating the activity of mitochondrial oxidative and intermediary metabolism.
  • SIRT3 is also uniquely poised to respond to the flux of mitochondrial NAD + and NADH, which is determined, in large part, by the capacity of the respiratory chain to oxidize NADH. This capacity is severely decreased in FRDA, as well as in other mitochondrial defects such as cytochrome c oxidase (complex IV) deficiency, causing an accumulation of NADH and, consequently, a redox state of perceived nutrient excess.
  • Defects in cellular respiration may be inherited as mitochondrial disease, or acquired over a lifetime via somatic mutations, and are linked to many conditions including neurodegenerative disease, diabetes, heart failure, cancer and in the aging process in general.
  • the involvement of cellular respiration in numerous common human pathologies emphasizes the need for greater understanding of the pathophysiological processes that occur in response to respiratory chain compromise. Accordingly, there exists a need to develop biomarkers of diseases or disorders associated with a deficiency in frataxin.
  • the present disclosure relates generally to methods and kits for screening for diseases and disorders associated with a deficiency in frataxin. More particularly, it has been discovered that the loss of frataxin in mitochondria causes the progressive hyperacetylation of mitochondrial proteins. Accordingly, the present disclosure is directed to methods for screening for diseases and disorders associated with a deficiency in frataxin, as well as to a kit for measuring or detecting protein acetylation levels.
  • the present disclosure is directed to methods for detecting the progression of a disease or disorder associated with a deficiency in frataxin in a subject having or suspected of having a deficiency in frataxin and to methods for monitoring the effectiveness of therapy in a subject having or suspected of having a disease or disorder associated with a deficiency in frataxin.
  • the present disclosure also is directed to methods of screening for therapeutic agents for use in treating diseases and disorders associated with a deficiency in frataxin.
  • the present disclosure is directed to a method of screening for a deficiency in frataxin in a subject.
  • the method includes determining the acetylation status of a mitochondrial protein in a sample tissue and determining the acetylation status of a mitochondrial protein in a normal tissue.
  • An increase in acetyl-lysine in the sample tissue as compared to acetyl-lysine in the normal tissue is indicative of a deficiency in frataxin.
  • the present disclosure is directed to a method of screening for a deficiency in frataxin in a subject.
  • the method includes determining the mitochondrial NADH level in a sample tissue and determining the mitochondrial NADH level in a normal tissue. An increase in the NADH level in the sample tissue as compared to the NADH level in the normal tissue is indicative of a deficiency in frataxin.
  • the method further includes determining the mitochondrial NAD + level in a sample tissue and/or determining the NAD + /NADH ratio in the sample tissue and determining the mitochondrial NAD + level in a normal tissue and/or determining NAD + /NADH ratio in the normal tissue, wherein a decrease in the NAD + /NADH ratio in the sample tissue as compared to the NAD + /NADH ratio in the normal tissue is indicative of a deficiency in frataxin.
  • the present disclosure is directed to a kit for measuring levels of mitochondrial protein acetylation.
  • the kit includes an arm-acetyl-lysine-specific antibody and an antibody that specifically binds to a mitochondrial protein.
  • the kit may include reagents for processing a sample, for isolating mitochondria, and for detecting protein acetylation levels (e.g., isolation buffers, colorimetric assay buffers and reagents, and acetylation detection buffers and reagents).
  • the kit measures acetyl-lysine residues with, for example, an acetyl-lysine-specific antibody.
  • the present disclosure is directed to a method of detecting the progression of a disease or a disorder associated with a deficiency in frataxin.
  • the method includes measuring mitochondrial protein acetylation in an earlier-obtained sample (i.e., a first chronological sample) and a later-obtained chronological sample (i.e., a second chronological sample) from a subject having or suspected of having the disease or the disorder associated with a deficiency in frataxin, where an increase in mitochondrial protein acetylation in the second chronological sample indicates progression of the disease or the disorder when compared to the mitochondrial protein acetylation in the first chronological sample.
  • the present disclosure is directed to a method for monitoring the effectiveness of therapy in a subject having or suspected of having a deficiency in frataxin.
  • the method includes measuring mitochondrial protein acetylation in at least a first chronological sample, administering the therapy to the subject, and measuring mitochondrial protein acetylation in at least a second chronological sample, where a decrease in the mitochondrial protein acetylation in the second chronological sample when compared to the mitochondrial protein acetylation in the first chronological sample indicates effectiveness of therapy.
  • the present disclosure is directed to a method for screening for agents that can be used for treating a disease or a disorder associated with a deficiency in frataxin.
  • the method includes measuring mitochondrial protein acetylation in a subject having hyperacetylated mitochondrial proteins, administering an agent suspected of modulating protein acetylation to the subject, measuring mitochondrial protein acetylation after administration of the agent, where the agent is considered to be a candidate for treating the disease or the disorder associated with a deficiency in frataxin if the mitochondrial protein acetylation is decreased in the sample after administration of the agent.
  • WT wild-type
  • NSE KO 24 day-old NSE-Cre knockout mice
  • WB Western blot
  • FIG. 3 is a Western Blot probing for internal acetyl-lysine residues (“Ac-K”) of total heart homogenate (“total”), the cytoplasmic fraction (“cyto”) and the mitochondrial (“mito”) fraction, a Western blot probing for the mitochondrial marker complex H iron-sulfur subunit, and a Western blot probing for the cytoplasmic marker GAPDH as discussed in Example 2.
  • Ac-K internal acetyl-lysine residues
  • FIG. 4 is a graph showing the average relative integrated densitometry values from the acetyl-lysine Western blot M FIG. 1 (mean ⁇ SD; *P ⁇ 0.05) as discussed in Example 2.
  • FIG. 5 is a graph showing the average relative integrated densitometry values from the acetyl-lysine Western blot in FIG. 2 (mean ⁇ SD; *P ⁇ 0.05) as discussed in Example 2.
  • FIG. 6 is a Western blot (WB) demonstrating the purity of the cardiac mitochondrial preparations using Histone H3 as a nuclear marker, Gapdh as a cytoplasmic marker, VDAC as a mitochondrial outer membrane marker, and Complex H as a mitochondrial inner membrane marker as discussed in Example 3.
  • WB Western blot
  • WT wild-type
  • NSE 24 day-old NSE frataxin knockout hearts
  • VDAC mitochondrial outer membrane protein voltage-dependent anion channel
  • FIG. 8 is a graph showing the calculated densitometry for SIRT3 relative to the loading control VDAC shown in FIG. 7 (mean ⁇ SD; N.S.: not significant) as discussed in Example 3.
  • FIG. 9 is a Western blot probing for internal acetyl-lysine residues of mitochondrial protein preparations from WT or NSE-Cre frataxin knockout hearts at day 7 showing an increase in acetylation as early as post-natal day 7 concomitant with downregulation of the respiratory complex II iron-sulfur as discussed in Examples 3 and 4.
  • FIG. 10 is a Western blot probing for internal acetyl-lysine residues of mitochondrial protein preparations isolated from 2-3 WT or NSE-Cre frataxin knockout hearts at day 7, day 17, and day 24 in their post-natal development, a Western blot probing for frataxin and VDAC as a loading control as discussed in Example 4.
  • NAD + nicotinamide adenine dinucleotide
  • NADH reduced NAD +
  • FIG. 13 is a Western blot showing the carbonyl pulldown of SIRT3 in frataxin-deficient mitochondria as discussed in Example 6.
  • FIG. 14 is a Western blot showing immunoprecipitation of WT (+/+) and NSE-Cre frataxin-deficient ( ⁇ / ⁇ ) cardiac mitochondrial proteins with an acetyl-lysine antibody and subsequent Western blot probing for the respiratory complex I subunit NDUFA9 and Western blots of 2 percent of the total mitochondrial lysate input used for the immunoprecipitations showing NDUFA9, SIRT3, frataxin, and the acetylation states of both samples as discussed in Example 7.
  • FIG. 15 is a Western blot showing immunoprecipitation of WT (+/+) and NSE-Cre frataxin-deficient ( ⁇ / ⁇ ) cardiac mitochondrial proteins with an acetyl-lysine antibody and subsequent Western blot probing for acetyl-CoA synthetase 2. (AceCS2) and Western blots of 2 percent of the total mitochondrial lysate input used for the immunoprecipitations showing AceCS2, SIRT3, frataxin, and the acetylation states of both samples as discussed in Example 7.
  • FIG. 16 shows Western blots probing for internal acetyl-lysine residues of mitochondrial protein preparations from WT or NSE-Cre frataxin knockout hearts incubated with recombinant SIRT3 and NAD + showing a reduction of the acetyl-lysine signal as discussed in Example 8.
  • FIG. 17 is a Western blot probing for internal acetyl-lysine residues of whole heart lysate derived from a 13 month-old wild-type (WT) animal and a 12 month-old animal harboring a missense mutation in the mitochondrial genome-encoded protein subunit of the respiratory chain enzyme cytochrome c oxidase (COI) and the same membrane used for Western blotting stained with Ponceau S as discussed in Example 9.
  • WT wild-type
  • COI cytochrome c oxidase
  • FIG. 18 is a graph showing the calculated integrated densitometry values for acetyl-lysine signal relative to Ponceau S signal from FIG. 17 as discussed in Example 9.
  • FIG. 19 is a Western blot probing for internal acetyl-lysine residues of whole heart lysate prepared from a 18 month-old WT animal and a 20 month-old animal lacking the non-respiratory chain mitochondrial inner membrane protein adenine nucleotide translocase 1 (ANTI ⁇ /31 ) and the same membrane used for Western blotting stained with Ponceau S as discussed in Example 9.
  • FIG. 20 is a graph showing the calculated integrated densitometry values for acetyl-lysine signal relative to Ponceau S signal from FIG. 19 as discussed in Example 9.
  • FIG. 21A is a Western blot probed for acetylation of mitochondrial proteins from mice with Friedreich's Ataxia treated with TAT-Frataxin as discussed in Example 10.
  • FIG. 21B is a graph quantifying the amount of acetylation of the upper most 4 bands shown in FIG. 21A by densitometry as discussed in Example 10.
  • biomarkers useful for assessing mitochondrial damage in disease have been discovered.
  • a deficiency in frataxin can cause progressive hyperacetylation of mitochondrial proteins.
  • the present disclosure relates to assessing the acetylation status of mitochondrial proteins as biomarkers for assessing mitochondrial damage.
  • Methods for assessing acetylation status of, for example, mitochondrial proteins to determine oxidative damage to mitochondria in certain diseases have also been discovered.
  • Suitable methods for determining the acetylation status of a mitochondrial protein may be performed by Western blot analysis, immunoprecipitation, enzyme-linked immunosorbent assay (ELISA) and mass spectrometry.
  • the present disclosure is directed to a method of screening for a disease or a disorder associated with a deficiency in frataxin in a subject.
  • the method includes determining the acetylation status of a mitochondrial protein, wherein acetyl-lysine in a sample from the subject is increased as compared to acetyl-lysine in normal tissue is indicative of a disease or a disorder associated with a deficiency in frataxin.
  • “normal tissue” refers to a reference sample obtained from a subject that is known to not have a disease or a disorder associated with a deficiency in frataxin.
  • the phrase “normal tissue” is also intended to refer to a reference value assigned to a subject or a panel of subjects that are known to not have a disease or a disorder associated with a deficiency in frataxin.
  • Mitochondria may be isolated by methods for isolating mitochondria from cells known to those skilled in the art.
  • the method may further include sub-fractionation of the tissue sample.
  • Sub-fractionation of the tissue sample allows for the isolation of mitochondria from other cellular and sub-cellular components of the tissue. Any suitable methods known by those skilled in the art may he used for tissue sub-fractionation.
  • tissue may be homogenized and then subjected to a standard differential centrifugation method to initially pellet nuclei and cellular debris followed by centrifugation of the resulting supernatant to pellet mitochondria.
  • Protein acetylation may be measured by methods for detecting acetylation known to those skilled in the art.
  • a particularly suitable method for detecting protein acetylation may be, for example, by detecting lysine acetylation.
  • levels of acetyl-lysine may be measured by utilizing an antibody specific for acetyl-lysine residues. Antibodies that specifically bind to acetyl-lysine residues are commercially available (Immunechem, Cell Signaling) or may be made using methods known by those skilled in the art.
  • Suitable methods for detecting lysine acetylation status of a mitochondrial protein can be determined, for example, by Western blot analysis, immunoprecipitation, enzyme-linked immunosorbent assay (ELISA) mass spectrometry and combinations of these methods.
  • Western blot analysis immunoprecipitation, enzyme-linked immunosorbent assay (ELISA) mass spectrometry and combinations of these methods.
  • ELISA enzyme-linked immunosorbent assay
  • Suitable subjects may be mammals.
  • Particularly suitable mammals can he, for example, humans and rodents such as, for example, rats and mice.
  • Mitochondrial proteins that are particularly suitable for determining acetyl-lysine status include those known by one skilled in the art (see e.g., Hebert, A. S., et al., Mol. Cell. 49:186-199 (2013)).
  • the diseases and disorders associated with a deficiency in frataxin can be, for example, Friedereich's Ataxia, Parkinson's Disease, Alzheimer's Disease, alcoholism, ischemic heart disease, dementia, Huntington's disease, Amyotrophic lateral sclerosis, cytochrome c oxidase deficiency, autosomal dominant progressive external ophthalmoplegia (adPEO), and Leber's Hereditary Optic Neuropathy (LHON).
  • Particularly suitable samples may be a tissue having cells that possess mitochondria and may be from any part of the body that is affected by the disease.
  • Particularly suitable tissues for obtaining samples can be, for example, liver, heart, white blood cells, and n Lan tissue.
  • the present disclosure is directed to a method of detecting the progression of a disease or a disorder associated with a deficiency in frataxin in a subject.
  • the method includes measuring mitochondrial protein acetylation, especially lysine acetylation, in a first chronological sample and a second chronological sample from a subject having or suspected of having the disease or the disorder associated with a deficiency in frataxin.
  • An increase in the mitochondrial protein acetylation in the second chronological sample indicates progression of the disease when compared to mitochondrial protein acetylation in the first chronological sample.
  • mitochondrial protein acetylation can he determined from about one day to about 30 days. It is within the skill of those in the art to detect the progression of a mitochondrial respiratory chain disorder by measuring mitochondrial protein acetylation.
  • the present disclosure is directed to a method of monitoring the effectiveness of a therapy in a subject having or suspected of having a disease or a disorder associated with a deficiency in frataxin.
  • the method includes measuring mitochondrial protein acetylation in at least a first chronological sample, administering the therapy to the subject and measuring mitochondrial protein acetylation in at least a second chronological sample from the subject.
  • a decrease in the mitochondrial protein acetylation in the second chronological sample indicates effectiveness of therapy when compared to the mitochondrial protein acetylation in the first chronological sample.
  • mitochondrial protein acetylation can be determined from about one day to about one year. It is within the skill of those in the art to begin a therapy and monitor the effectiveness of over the entire course of therapy, as well as for a time following termination of therapy.
  • the present disclosure is directed to a method for screening for agents that can be used as therapeutic agents for treating a disease or a disorder associated with a deficiency in frataxin.
  • the method includes measuring mitochondrial protein acetylation in a subject having hyperacetylated mitochondrial proteins, administering an agent suspected of modulating protein acetylation to the subject, and measuring mitochondrial protein acetylation after administration of the agent.
  • the agent is considered to be a candidate for treating the disease or the disorder associated with a deficiency in frataxin if the mitochondrial protein acetylation is decreased in the sample after administration of the agent.
  • the present disclosure is directed to a method of screening for a disease or a disorder associated with a deficiency in frataxin in a subject.
  • the method includes determining the mitochondrial NADH level in a sample tissue, wherein an increase in NADH level in the sample tissue as compared to NADH level in normal tissue is indicative of a mitochondrial respiratory chain disorder.
  • the method may further include determining mitochondrial NADH level a sample tissue, determining NAD + /NADH ratio in the sample tissue, determining mitochondrial NAD + level in a normal tissue, and determining NAD + /NADH ratio in the normal tissue, wherein a decrease in the NAD 30 /NADH ratio in the sample tissue as compared to the NAD 30 /NADH ratio in normal tissue is indicative of a disease or a disorder associated with a deficiency in frataxin.
  • NAD + and NADH levels can be done using methods known by those skilled in the art using commercially available assays (Bioassay Systems). Once NAD + and NADH levels are determined, the NAD + /NADH ratio can be calculated.
  • the present disclosure is directed to a kit for Measuring or detecting mitochondrial protein acetylation.
  • the kit may include reagents for processing a sample, for isolating mitochondria, and for detecting protein acetylation levels (e.g, isolation buffers, colorimetric assay buffers and reagents, and acetylation detection buffers and reagents).
  • the kit measures acetyl-lysine residues with, for example, an anti-acetyl-lysine-specific antibody. Suitable anti-acetyl-lysine antibodies are commercially available (Immunechem; Cell Signaling).
  • the kit may further use a combination of antibodies such as, for example, an anti-acetyl-lysine-specific antibody and an anti-mitochondrial protein antibody.
  • Suitable anti-mitochondrial protein antibodies can be, for example, antibodies that specifically bind to NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 9 (NDUFA9), antibodies that specifically bind to acetyl-CoA synthetase 2 (AceCS2), antibodies that specifically bind to frataxin, antibodies that specifically bind to complex II 30 kDa subunit, antibodies that specifically bind to complex III Rieske protein, antibodies that specifically bind to NAD-dependent deacetylase sirtuin-3 (SIRT3), antibodies that specifically bind to voltage-dependent anion-selective channel (VDAC) and antibodies that specifically bind to other mitochondrial proteins known to those skilled in the art.
  • the antibodies may be, for example, monoclonal antibodies, polyclonal antibodies and combinations thereof
  • Kits may also use solid supports such as, for example, microtiter plates, membranes (e.g., nitrocellulose, polyvinyl chloride, nylon, polyvinylidene fluoride, diazotized paper) and beads (e.g., polystyrene latex, activated beads, Protein A beads, Protein G beads).
  • solid supports such as, for example, microtiter plates, membranes (e.g., nitrocellulose, polyvinyl chloride, nylon, polyvinylidene fluoride, diazotized paper) and beads (e.g., polystyrene latex, activated beads, Protein A beads, Protein G beads).
  • the kit can further include instructions for using the reagents for processing a sample, for isolating mitochondria, for measuring mitochondrial protein acetylation, for detecting protein acetylation levels and combinations thereof.
  • the instructions may be provided in the kit packaging and provided by other media such as, for example, a website.
  • mice were obtained from Helene Puccio and bred as described previously to generate the NSE and MCK-Cre conditional knockout animals. PCR was used to detect the presence of a 500 bp product of exon 4 and a 790 bp product of the Cre transgene for genotyping. Mice that were genotyped as Frda L3/ ⁇ , Cre+ represent the conditional knockout animals.
  • the whole tissue homogenates were then transferred to 1.5 ml tubes and spun at 17,000 ⁇ g for 10 min at 4° C. in a desktop centrifuge to pellet membranes and tissue debris. The supernatant was saved and immediately frozen on dry-ice and stored at ⁇ 80° C. for later analysis. Frozen, RIPA buffer solubilized ANT1 ⁇ / ⁇ and COI missense mutation cardiac lysates were provided by Dr. Douglas C. Wallace of The Children's Hospital of Philadelphia.
  • Minced heart tissue from 2-3 animals was submerged in 3 ml/g MIB containing an EDTA-free protease inhibitor cocktail (Roche) and 10 mM nicotinamide (NAM), 200 nM Trichostatin A, and 5 mM sodium butyrate as deacetylase inhibitors.
  • Heart tissue was homogenized with four passes of a motor-driven Potter-Elvehjem homogenizer with a Teflon pestle at medium speed on ice. An additional 7 ml/g MIB was added to the heart homogenate, which was then transferred to a pre-chilled 50 ml conical tube.
  • a crude mitochondrial pellet was obtained via a standard differential centrifugation method.
  • nuclei and the heavy cell fraction are removed via centrifugation for 15 minutes at 1000 ⁇ g at 4° C.
  • the resulting post-nuclear supernatant was subjected to centrifugation for 15 min at 10,000 ⁇ g to pellet mitochondria.
  • the supernatant was removed as the cytosolic fraction and sedimented mitochondria were gently resuspended in 1 ml ice-cold MIB containing deacetylase inhibitors and then diluted with MIB to a total volume of 10 ml and centrifuged again for 15 min at 10,000 ⁇ g.
  • the supernatant was then removed and sedimented mitochondria are gently washed in 0.5 M KCl to remove endoplasmic reticulum and lysosome contaminants.
  • the sedimented mitochondria were solubilized in complete RIPA buffer containing protease and deacetylase inhibitors (see Mouse tissue preparation) and frozen until later analysis.
  • the pelleted mitochondria were resuspended in ice-cold buffer containing 50 mM Tris-Cl (pH 7.4), 150 mM NaCl, and an EDTA-free protease inhibitor tablet and immediately frozen at ⁇ 80° C.
  • ice-cold buffer containing 50 mM Tris-Cl (pH 7.4), 150 mM NaCl, and an EDTA-free protease inhibitor tablet and immediately frozen at ⁇ 80° C.
  • For NAD + and NADH measurements sedimented mitochondria were immediately resuspended in 180 ⁇ l of ice-cold 1 mM Tris-Cl pH 7.4, 150 mM NaCl and extracted appropriately (see Mitochondrial NAD + and NADH measurements).
  • Antibodies and Western Blotting were anti-acetyl-lysine (Immunechem anti-acetyl-lysine (Cell Signaling) anti-complex II 30 kDa subunit, anti-NDUFA9, anti-complex III Rieske protein (Mitosciences), anti-SIRT3 (provided by E. Verdin, Gladstone Institute, UCSF), anti-frataxin (provided by G. Isaya, Mayo Clinic), anti-SIRT3 D22A3, anti-VDAC D73D12, and anti-histone H3 (Cell Signaling), anti-GAPDH and anti-tubulin (Sigma), and anti-AceCS2 (ACSS 1)(Abeam).
  • the sample was incubated at 4° C. with gentle agitation for an additional 2 hours with 7.5 ⁇ l Protein A agarose (Invitrogen) plus 7.5 ⁇ l Protein G agarose (Roche). Agarose was collected via a 1 min centrifugation at 400 ⁇ g at 4° C. with a desktop centrifuge and the unbound mitochondrial supernatant was carefully removed from above the agarose beads. The beads were washed 3 times for 5 min. in 500 ⁇ l IP buffer containing 50 mM Tris-Cl (pH 7.4), 150 mM NaCl, 1 mM EGTA, and 0.5% NP-40 at 4° C. and then incubated in 30 ⁇ l SDS sample loading buffer and boiled at 95° C. for 5 min. The immunoprecipitated sample was then resolved on a 12% SDS-PAGE and processed for Western blotting.
  • IP buffer containing 50 mM Tris-Cl (pH 7.4), 150 mM NaCl, 1 m
  • NAM Mitochondrial NAD + and NADH measurements.
  • NAM was excluded from MIB used to isolate mitochondria for NAD + measurements as the enzyme nicotinamide phosphoribosyltransferase (Nampt) catalyzes the formation of NMN, an NAD + precursor, from NAM.
  • NAD(H) measurements were performed with a cycling assay according to the manufacturer's instructions (Bioassay Systems).
  • NAD + was extracted in the mitochondrial sample via addition of strong acid and then heated at 60° C. to eliminate the presence of the reduced nucleotide (NADH).
  • NADH was extracted in mitochondrial sample homogenate via addition of strong base and then heated at 60° C. to eliminate the presence of the oxidized nucleotide (NAD + ).
  • the extractions were neutralized with the opposite extraction buffer and the dehydrogenase cycling assay was performed in which, in the presence of lactate, lactate dehydrogenase, diaphorase and MTT formazan reagent, the concentration of NAD + or NADH present in the sample is proportional to the amount of reduced TvITF formation which was colorimetrically detected with a plate reader at 562 nm. Sample concentrations were determined via an NAD + standard curve. The coenzyme concentration was normalized to the protein concentration of the sample.
  • 4-HNE modified SIRT3 Detection of 4-HNE modified SIRT3.
  • cardiac mitochondrial proteins were solubilized as described earlier and 4-HNE modified proteins were derivatized to a biotin group via a 2 hour incubation in 5 mM EZ-Link hydrazide biotin (Thermo Scientific) in the dark.
  • the samples were dialyzed 1:10000 in PBS overnight to remove excess biotin hydrazide. 100 ⁇ g of protein sample was then incubated with 50 ⁇ l of NeutrAvidin Agarose Resin (Thermo Scientific) at 4° C. with gentle rocking overnight. Beads were washed 5 times for 5 mM with 500 ⁇ l PBS-0.2% Tween 20, then boiled at 95° C. in 35 ⁇ l SDS loading buffer and processed for Western blotting as described previously.
  • NSE- and MCK-Cre conditional mouse models for Friedreich's Ataxia were analyzed for protein acetyl-lysine modifications.
  • whole heart lysates from wild-type (WT), NSE, and MCK conditional mouse models of FRDA were analyzed by Western blot analysis to assay protein acetyl-lysine modifications.
  • FIGS. 1 and 2 heart lysates from both the NSE (NSE KO) and MCK (MCK KO) mouse models of FRDA exhibited marked increases in acetyl-lysine modifications as compared to age-matched control hearts.
  • hyperacetylation was localized to cardiac mitochondria.
  • FIGS. 4 and 5 the increases in acetyl-lysine modifications as compared to age-matched control hearts were statistically significant. The differences were most dramatic in proteins with an estimated molecular weight between approximately 30 and 75 kDa ( FIG. 1 ).
  • frataxin-deficient cardiac mitochondria displayed marked hyperacetylation of numerous proteins ( FIGS. 3 and 7 ). This was accompanied by a characteristic downregulation of respiratory complex I and II (succinate dehydrogenase) that was present as early as 7 days post-natal ( FIG. 9 ).
  • NSE-Cre mouse models of FRDA begin to develop cardiac hypertrophy in the second week of life.
  • cardiac mitochondria from WT control and NSE-Cre mice at post-natal days 7, 17, and 24 were prepared and analyzed by Western blot analysis.
  • NSE-Cre cardiac mitochondrial proteins exhibited only a mildly increased acetylation state as compared to their wild-type (WT) counterparts.
  • WT wild-type
  • NSE-Cre cardiac mitochondria displayed increased acetylation of cardiac mitochondrial proteins as compared to their WT counterparts, which became more dramatic by post-natal day 24.
  • the progressive increase in cardiac mitochondrial protein acetylation over this time frame corresponded with the development of cardiac hypertrophy in the NSE-Cre models ( FIG. 11 ).
  • SIRT3 in WT heart mitochondria exhibited a small amount of 4-HNE modification.
  • SIRT3 in frataxin-deficient mitochondria exhibited a marked increase in 4-HNE modification suggesting that SIRT3 may be directly inhibited via carbonyl group adduction in the setting of frataxin deficiency.
  • Mitochondrial acetyl-lysine proteins were immunoprecipitated and analyzed by Western blot for NDUFA9. Consistent with previous findings, a minimal amount of NDUFA9 acetyl-lysine signal in the WT (+/+) mitochondrial immunoprecipitate was observed. However, the frataxin-deficient ( ⁇ / ⁇ ) mitochondrial immunoprecipitate displayed a greater acetylated NDUFA9 signal despite downregulation of NDUFA9 as seen in the input ( FIG. 14 ). The acetylation state of acetyl-CoA synthetase 2 (AceCS2) was also analyzed.
  • AceCS2 acetylation state of acetyl-CoA synthetase 2
  • this analysis revealed a greater amount of acetylated AceCS2 in the frataxin-deficient ( ⁇ / ⁇ ) condition as compared to the WT (+/+) ( FIG. 15 ).
  • the increased acedation states of two known targets of SIRT3-mediated deacetylation indirectly demonstrated that SIRT3 is inhibited in frataxin-deficient cardiac mitochondria.
  • increases in the acetylation states of NDUFA9 and AceCS2 were linked to a decrease in the activity of respiratory complex I and the synthesis of activated acetate, respectively.
  • solubilized WT and frataxin-deficient cardiac mitochondrial proteins were incubated with glutathione S-transferase tagged, processed human recombinant SIRT3 (GST-hSIRT3) in the presence or absence of NAD + to assay for changes in acetylation states.
  • GST-hSIRT3 glutathione S-transferase tagged, processed human recombinant SIRT3
  • NAD + alone caused no change in acetylation signal.
  • incubating frataxin deficient mitochondrial protein with 3 ⁇ g of GST-hSIRT3 and NAD + resulted in a marked reduction of acetyl-lysine signal from nearly every protein band, while completely eliminating the acetyl-lysine signal of multiple bands ( FIG.
  • mice were treated with TAT-Frataxin for 2 weeks (FXN ⁇ / ⁇ +TAT-FXN; FIG. 21A lanes 3 and 4) or phosphate buffered saline (FXN ⁇ / ⁇ ; FIG. 21A lanes 1 and 2). Specifically, mice were dosed according to body weight with a total volume of approximately 20 ⁇ l/gram of weight administered intraperitoneally.
  • mice TAT-Frataxin decreased protein acetylation in NSE-Cre FXN knockout mice (NSE KO+TAT-FXN). Therefore, mitochondrial protein acetylation status can be used as a biomarker to monitor treatment in a mouse knockout model of Friedreich's Ataxia, which is associated with a deficiency in frataxin.
  • a deficiency in frataxin can cause progressive hyperacetylation of mitochondrial proteins. More specifically, hyperacetylation of mitochondrial proteins may be due to the inhibition of a SIRT3 deacetylase. SIRT3 may respond to the flux of mitochondrial. NAD + and NADH, which is determined, in large part, by the capacity of the respiratory chain to oxidize NADH. This capacity is severely decreased in FRDA, as well as in other mitochondrial defects such as cytochrome c oxidase (complex IV) deficiency, causing an accumulation of NADH and, consequently, a redox state of perceived nutrient excess.
  • complex IV complex IV

Abstract

Compositions and methods for screening for a disease or a disorder associated with a deficiency in frataxin in a subject using biomarkers for diseases or disorders associated with a deficiency in frataxin are disclosed. The compositions and methods include determining the acetylation status of mito-chondrial proteins. Also disclosed are methods of detecting progression of a disease or a disorder associated with a deficiency in frataxin in a subject and methods of monitoring effectiveness of a therapy for diseases or disorders associated with a deficiency in frataxin.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. Publication no. 2015/0132769, filed on Sep. 2, 2014, which claims priority to International Publication no. PCT/US2013/028609, filed on Mar. 1, 2013, which claims priority to U.S. provisional application No. 61/605,783, filed on Mar. 2, 2012 and U.S. provisional application No. 61/607,918, filed on Mar. 7, 2012, all of which are incorporated herein by reference in their entireties.
  • STATEMENT OF GOVERNMENT SUPPORT
  • This invention was made with government support under HL085098 awarded by the National Institutes of Health. The government has certain rights in the invention.
  • BACKGROUND OF THE DISCLOSURE
  • The present disclosure relates generally to compositions and methods for screening for diseases or disorders associated with a deficiency in frataxin. More particularly, the present disclosure relates to methods of screening for a disease or disorder associated with a deficiency in frataxin, as well as methods of screening for therapeutic agents for use in treating a disease or disorder associated with a deficiency in frataxin, or damage from reactive oxygen species to mitochondrial proteins affecting the respiratory chain, or increased mitochondrial protein acetylation affecting mitochondrial function, as well as method of screening for therapeutic agents for use in treating a disease or disorder associated with a deficiency in frataxin.
  • Friedreich's Ataxia (FRDA) is an autosomal recessive mitochondrial disorder caused by a homozygous triplet nucleotide repeat (GAATTC) expansion in intron 1 of the FXN gene located on chromosome 9q21.11. This intronic expansion causes impaired transcription of the FXN gene and, consequently, a pathological deficiency of the FXN gene product, frataxin. Frataxin is targeted to the mitochondrial matrix, where it is known to act as an iron-binding protein and participates in the proper assembly and function of iron-sulfur cluster (ISC) dependent proteins including complexes I, II, and III of the respiratory chain and aconitase of the tricarboxylic acid (TCA) cycle. Thus, frataxin deficiency severely compromises both cellular respiration and overall mitochondrial function leading to energetic stress and ATP deficiency. Although patients develop multisystem disease including early spinocerebellar degeneration, ataxia, and diabetes, the primary cause of death is heart failure for nearly 85% of those afflicted. Similarly, although the phenotypes of the neuron-specific enolase (NSE) and muscle creatine kinase (MCK) Cre conditional mouse models of FRDA differ, both models develop a fatal cardiomyopathy and impaired activity of iron-sulfur cluster-dependent respiratory complexes consistent with the human disease.
  • Recent work has demonstrated that lysine acetylation is a highly conserved and abundant post-translational modification within mitochondria that is responsive to nutrient availability and may contribute to the physiological adaptations of reduced caloric intake. Multiple investigations have demonstrated a role for reversible mitochondrial enzyme deacetylation and, specifically, the NAD+-dependent deacetylase SIRT3, in the regulation of fatty acid oxidation, the TCA cycle, electron transport via respiratory complexes I and II, and overall oxidative metabolism. SIRT3-mediated deacetylation has recently emerged as a major mechanism regulating the activity of mitochondrial oxidative and intermediary metabolism. SIRT3 is also uniquely poised to respond to the flux of mitochondrial NAD+ and NADH, which is determined, in large part, by the capacity of the respiratory chain to oxidize NADH. This capacity is severely decreased in FRDA, as well as in other mitochondrial defects such as cytochrome c oxidase (complex IV) deficiency, causing an accumulation of NADH and, consequently, a redox state of perceived nutrient excess.
  • Defects in cellular respiration may be inherited as mitochondrial disease, or acquired over a lifetime via somatic mutations, and are linked to many conditions including neurodegenerative disease, diabetes, heart failure, cancer and in the aging process in general. The involvement of cellular respiration in numerous common human pathologies emphasizes the need for greater understanding of the pathophysiological processes that occur in response to respiratory chain compromise. Accordingly, there exists a need to develop biomarkers of diseases or disorders associated with a deficiency in frataxin.
  • SUMMARY OF THE DISCLOSURE
  • The present disclosure relates generally to methods and kits for screening for diseases and disorders associated with a deficiency in frataxin. More particularly, it has been discovered that the loss of frataxin in mitochondria causes the progressive hyperacetylation of mitochondrial proteins. Accordingly, the present disclosure is directed to methods for screening for diseases and disorders associated with a deficiency in frataxin, as well as to a kit for measuring or detecting protein acetylation levels. Additionally, the present disclosure is directed to methods for detecting the progression of a disease or disorder associated with a deficiency in frataxin in a subject having or suspected of having a deficiency in frataxin and to methods for monitoring the effectiveness of therapy in a subject having or suspected of having a disease or disorder associated with a deficiency in frataxin. The present disclosure also is directed to methods of screening for therapeutic agents for use in treating diseases and disorders associated with a deficiency in frataxin.
  • In one aspect, the present disclosure is directed to a method of screening for a deficiency in frataxin in a subject. The method includes determining the acetylation status of a mitochondrial protein in a sample tissue and determining the acetylation status of a mitochondrial protein in a normal tissue. An increase in acetyl-lysine in the sample tissue as compared to acetyl-lysine in the normal tissue is indicative of a deficiency in frataxin.
  • In another aspect, the present disclosure is directed to a method of screening for a deficiency in frataxin in a subject. The method includes determining the mitochondrial NADH level in a sample tissue and determining the mitochondrial NADH level in a normal tissue. An increase in the NADH level in the sample tissue as compared to the NADH level in the normal tissue is indicative of a deficiency in frataxin. In some embodiments, the method further includes determining the mitochondrial NAD+ level in a sample tissue and/or determining the NAD+/NADH ratio in the sample tissue and determining the mitochondrial NAD+ level in a normal tissue and/or determining NAD+/NADH ratio in the normal tissue, wherein a decrease in the NAD+/NADH ratio in the sample tissue as compared to the NAD+/NADH ratio in the normal tissue is indicative of a deficiency in frataxin.
  • In another aspect, the present disclosure is directed to a kit for measuring levels of mitochondrial protein acetylation. The kit includes an arm-acetyl-lysine-specific antibody and an antibody that specifically binds to a mitochondrial protein. The kit may include reagents for processing a sample, for isolating mitochondria, and for detecting protein acetylation levels (e.g., isolation buffers, colorimetric assay buffers and reagents, and acetylation detection buffers and reagents). In one embodiment, the kit measures acetyl-lysine residues with, for example, an acetyl-lysine-specific antibody.
  • In another aspect, the present disclosure is directed to a method of detecting the progression of a disease or a disorder associated with a deficiency in frataxin. The method includes measuring mitochondrial protein acetylation in an earlier-obtained sample (i.e., a first chronological sample) and a later-obtained chronological sample (i.e., a second chronological sample) from a subject having or suspected of having the disease or the disorder associated with a deficiency in frataxin, where an increase in mitochondrial protein acetylation in the second chronological sample indicates progression of the disease or the disorder when compared to the mitochondrial protein acetylation in the first chronological sample.
  • In another aspect, the present disclosure is directed to a method for monitoring the effectiveness of therapy in a subject having or suspected of having a deficiency in frataxin. The method includes measuring mitochondrial protein acetylation in at least a first chronological sample, administering the therapy to the subject, and measuring mitochondrial protein acetylation in at least a second chronological sample, where a decrease in the mitochondrial protein acetylation in the second chronological sample when compared to the mitochondrial protein acetylation in the first chronological sample indicates effectiveness of therapy.
  • In another aspect, the present disclosure is directed to a method for screening for agents that can be used for treating a disease or a disorder associated with a deficiency in frataxin. The method includes measuring mitochondrial protein acetylation in a subject having hyperacetylated mitochondrial proteins, administering an agent suspected of modulating protein acetylation to the subject, measuring mitochondrial protein acetylation after administration of the agent, where the agent is considered to be a candidate for treating the disease or the disorder associated with a deficiency in frataxin if the mitochondrial protein acetylation is decreased in the sample after administration of the agent.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The disclosure will be better understood, and features, aspects and advantages other than those set forth above will become apparent when consideration is given to the following detailed description thereof. Such detailed description makes reference to the following drawings, wherein:
  • FIG. 1 is a Western blot (WB) probing for internal acetyl-lysine residues using total heart homogenates derived from 24 day-old wild-type (WT, n=2, lanes 1 and 2) and 24 day-old NSE-Cre knockout mice (“NSE KO”; n=2, lanes 3 and 4), the corresponding SDS-PAGE gel stained with Coomassie Blue and a Western blot probing for frataxin as discussed in Example 2.
  • FIG. 2 is a Western blot (WB) probing for internal acetyl-lysine residues using total heart homogenates prepared from 9 week-old WT (n=2, lanes 1 and 2) and 9 and 11 week-old MCK-Cre mouse models of FA ( lanes 3 and 4, respectively), the corresponding SDS-PAGE gel stained with Coomassie Blue and a Western blot probing for frataxin as discussed in Example 2.
  • FIG. 3 is a Western Blot probing for internal acetyl-lysine residues (“Ac-K”) of total heart homogenate (“total”), the cytoplasmic fraction (“cyto”) and the mitochondrial (“mito”) fraction, a Western blot probing for the mitochondrial marker complex H iron-sulfur subunit, and a Western blot probing for the cytoplasmic marker GAPDH as discussed in Example 2.
  • FIG. 4 is a graph showing the average relative integrated densitometry values from the acetyl-lysine Western blot M FIG. 1 (mean±SD; *P<0.05) as discussed in Example 2.
  • FIG. 5 is a graph showing the average relative integrated densitometry values from the acetyl-lysine Western blot in FIG. 2 (mean±SD; *P<0.05) as discussed in Example 2.
  • FIG. 6 is a Western blot (WB) demonstrating the purity of the cardiac mitochondrial preparations using Histone H3 as a nuclear marker, Gapdh as a cytoplasmic marker, VDAC as a mitochondrial outer membrane marker, and Complex H as a mitochondrial inner membrane marker as discussed in Example 3.
  • FIG. 7 is a Western blot probing for internal acetyl-lysine residues of isolated mitochondrial protein obtained from hearts from 24 day-old wild-type (WT, n=2, Lanes 1 and 2) hearts and 24 day-old NSE frataxin knockout hearts (“NSE”; n−2, Lanes 3 and 4), Western blots probing for the respiratory Complex 1 subunit NDUFA9, the Complex II 30 kDa iron-sulfur subunit, SIRT3 and the mitochondrial outer membrane protein voltage-dependent anion channel (VDAC) as a loading control as discussed in Example 3.
  • FIG. 8 is a graph showing the calculated densitometry for SIRT3 relative to the loading control VDAC shown in FIG. 7 (mean±SD; N.S.: not significant) as discussed in Example 3.
  • FIG. 9 is a Western blot probing for internal acetyl-lysine residues of mitochondrial protein preparations from WT or NSE-Cre frataxin knockout hearts at day 7 showing an increase in acetylation as early as post-natal day 7 concomitant with downregulation of the respiratory complex II iron-sulfur as discussed in Examples 3 and 4.
  • FIG. 10 is a Western blot probing for internal acetyl-lysine residues of mitochondrial protein preparations isolated from 2-3 WT or NSE-Cre frataxin knockout hearts at day 7, day 17, and day 24 in their post-natal development, a Western blot probing for frataxin and VDAC as a loading control as discussed in Example 4.
  • FIG. 11 is a graph assessing the post-natal development of cardiac hypertrophy in the NSE KO mice as measured by heart weight divided by body weight (mean±SD; n=3 measurements at each time point; *P<0.05) as discussed in Example 4.
  • FIG. 12 is a graph showing the determination of nicotinamide adenine dinucleotide (NAD+) and reduced NAD+ (NADH) levels in WT and frataxin deficient cardiac mitochondrial preparations (n=3-5 biological replicates per condition, mean±SD; **P<0.005) and the figure inset shows the ratio of NAD+ to NADH for WT and NSE-KO heart mitochondria as discussed in Example 5.
  • FIG. 13 is a Western blot showing the carbonyl pulldown of SIRT3 in frataxin-deficient mitochondria as discussed in Example 6.
  • FIG. 14 is a Western blot showing immunoprecipitation of WT (+/+) and NSE-Cre frataxin-deficient (−/−) cardiac mitochondrial proteins with an acetyl-lysine antibody and subsequent Western blot probing for the respiratory complex I subunit NDUFA9 and Western blots of 2 percent of the total mitochondrial lysate input used for the immunoprecipitations showing NDUFA9, SIRT3, frataxin, and the acetylation states of both samples as discussed in Example 7.
  • FIG. 15 is a Western blot showing immunoprecipitation of WT (+/+) and NSE-Cre frataxin-deficient (−/−) cardiac mitochondrial proteins with an acetyl-lysine antibody and subsequent Western blot probing for acetyl-CoA synthetase 2. (AceCS2) and Western blots of 2 percent of the total mitochondrial lysate input used for the immunoprecipitations showing AceCS2, SIRT3, frataxin, and the acetylation states of both samples as discussed in Example 7.
  • FIG. 16 shows Western blots probing for internal acetyl-lysine residues of mitochondrial protein preparations from WT or NSE-Cre frataxin knockout hearts incubated with recombinant SIRT3 and NAD+ showing a reduction of the acetyl-lysine signal as discussed in Example 8.
  • FIG. 17 is a Western blot probing for internal acetyl-lysine residues of whole heart lysate derived from a 13 month-old wild-type (WT) animal and a 12 month-old animal harboring a missense mutation in the mitochondrial genome-encoded protein subunit of the respiratory chain enzyme cytochrome c oxidase (COI) and the same membrane used for Western blotting stained with Ponceau S as discussed in Example 9.
  • FIG. 18 is a graph showing the calculated integrated densitometry values for acetyl-lysine signal relative to Ponceau S signal from FIG. 17 as discussed in Example 9.
  • FIG. 19 is a Western blot probing for internal acetyl-lysine residues of whole heart lysate prepared from a 18 month-old WT animal and a 20 month-old animal lacking the non-respiratory chain mitochondrial inner membrane protein adenine nucleotide translocase 1 (ANTI−/31) and the same membrane used for Western blotting stained with Ponceau S as discussed in Example 9.
  • FIG. 20 is a graph showing the calculated integrated densitometry values for acetyl-lysine signal relative to Ponceau S signal from FIG. 19 as discussed in Example 9.
  • FIG. 21A is a Western blot probed for acetylation of mitochondrial proteins from mice with Friedreich's Ataxia treated with TAT-Frataxin as discussed in Example 10.
  • FIG. 21B is a graph quantifying the amount of acetylation of the upper most 4 bands shown in FIG. 21A by densitometry as discussed in Example 10.
  • While the disclosure is susceptible to various modifications and alternative forms, specific embodiments thereof have been shown by way of example in the drawings and are herein described below in detail. It should be understood, however, that the description of specific embodiments is not intended to limit the disclosure to cover all modifications, equivalents and alternatives falling within the spirit and scope of the disclosure as defined by the appended claims.
  • DETAILED DESCRIPTION
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the disclosure belongs. Although any methods and materials similar to or equivalent to those described herein may be used in the practice or testing of the present disclosure, the preferred materials and methods are described below.
  • In accordance with the present disclosure, biomarkers useful for assessing mitochondrial damage in disease have been discovered. Significantly, it has been discovered that a deficiency in frataxin can cause progressive hyperacetylation of mitochondrial proteins. Accordingly, the present disclosure relates to assessing the acetylation status of mitochondrial proteins as biomarkers for assessing mitochondrial damage. Methods for assessing acetylation status of, for example, mitochondrial proteins to determine oxidative damage to mitochondria in certain diseases have also been discovered.)
  • Suitable methods for determining the acetylation status of a mitochondrial protein may be performed by Western blot analysis, immunoprecipitation, enzyme-linked immunosorbent assay (ELISA) and mass spectrometry.
  • In another aspect, the present disclosure is directed to a method of screening for a disease or a disorder associated with a deficiency in frataxin in a subject. The method includes determining the acetylation status of a mitochondrial protein, wherein acetyl-lysine in a sample from the subject is increased as compared to acetyl-lysine in normal tissue is indicative of a disease or a disorder associated with a deficiency in frataxin. As used herein, “normal tissue” refers to a reference sample obtained from a subject that is known to not have a disease or a disorder associated with a deficiency in frataxin. The phrase “normal tissue” is also intended to refer to a reference value assigned to a subject or a panel of subjects that are known to not have a disease or a disorder associated with a deficiency in frataxin.
  • Mitochondria may be isolated by methods for isolating mitochondria from cells known to those skilled in the art. The method may further include sub-fractionation of the tissue sample. Sub-fractionation of the tissue sample allows for the isolation of mitochondria from other cellular and sub-cellular components of the tissue. Any suitable methods known by those skilled in the art may he used for tissue sub-fractionation. For example, tissue may be homogenized and then subjected to a standard differential centrifugation method to initially pellet nuclei and cellular debris followed by centrifugation of the resulting supernatant to pellet mitochondria.
  • Protein acetylation may be measured by methods for detecting acetylation known to those skilled in the art. A particularly suitable method for detecting protein acetylation may be, for example, by detecting lysine acetylation. In one embodiment, levels of acetyl-lysine may be measured by utilizing an antibody specific for acetyl-lysine residues. Antibodies that specifically bind to acetyl-lysine residues are commercially available (Immunechem, Cell Signaling) or may be made using methods known by those skilled in the art. Suitable methods for detecting lysine acetylation status of a mitochondrial protein can be determined, for example, by Western blot analysis, immunoprecipitation, enzyme-linked immunosorbent assay (ELISA) mass spectrometry and combinations of these methods.
  • Suitable subjects may be mammals. Particularly suitable mammals can he, for example, humans and rodents such as, for example, rats and mice.
  • Mitochondrial proteins that are particularly suitable for determining acetyl-lysine status include those known by one skilled in the art (see e.g., Hebert, A. S., et al., Mol. Cell. 49:186-199 (2013)).
  • The diseases and disorders associated with a deficiency in frataxin can be, for example, Friedereich's Ataxia, Parkinson's Disease, Alzheimer's Disease, alcoholism, ischemic heart disease, dementia, Huntington's disease, Amyotrophic lateral sclerosis, cytochrome c oxidase deficiency, autosomal dominant progressive external ophthalmoplegia (adPEO), and Leber's Hereditary Optic Neuropathy (LHON). Particularly suitable samples may be a tissue having cells that possess mitochondria and may be from any part of the body that is affected by the disease. Particularly suitable tissues for obtaining samples can be, for example, liver, heart, white blood cells, and n Lan tissue.
  • In another aspect, the present disclosure is directed to a method of detecting the progression of a disease or a disorder associated with a deficiency in frataxin in a subject. The method includes measuring mitochondrial protein acetylation, especially lysine acetylation, in a first chronological sample and a second chronological sample from a subject having or suspected of having the disease or the disorder associated with a deficiency in frataxin. An increase in the mitochondrial protein acetylation in the second chronological sample indicates progression of the disease when compared to mitochondrial protein acetylation in the first chronological sample. For chronological measurement to detect the progression of a disease, mitochondrial protein acetylation can he determined from about one day to about 30 days. It is within the skill of those in the art to detect the progression of a mitochondrial respiratory chain disorder by measuring mitochondrial protein acetylation.
  • In another aspect, the present disclosure is directed to a method of monitoring the effectiveness of a therapy in a subject having or suspected of having a disease or a disorder associated with a deficiency in frataxin. The method includes measuring mitochondrial protein acetylation in at least a first chronological sample, administering the therapy to the subject and measuring mitochondrial protein acetylation in at least a second chronological sample from the subject. A decrease in the mitochondrial protein acetylation in the second chronological sample indicates effectiveness of therapy when compared to the mitochondrial protein acetylation in the first chronological sample. For chronological measurement to monitor the effectiveness of a mitochondrial respiratory chain disorder therapy, mitochondrial protein acetylation can be determined from about one day to about one year. It is within the skill of those in the art to begin a therapy and monitor the effectiveness of over the entire course of therapy, as well as for a time following termination of therapy.
  • In another aspect, the present disclosure is directed to a method for screening for agents that can be used as therapeutic agents for treating a disease or a disorder associated with a deficiency in frataxin. The method includes measuring mitochondrial protein acetylation in a subject having hyperacetylated mitochondrial proteins, administering an agent suspected of modulating protein acetylation to the subject, and measuring mitochondrial protein acetylation after administration of the agent. The agent is considered to be a candidate for treating the disease or the disorder associated with a deficiency in frataxin if the mitochondrial protein acetylation is decreased in the sample after administration of the agent.
  • In another aspect, the present disclosure is directed to a method of screening for a disease or a disorder associated with a deficiency in frataxin in a subject. The method includes determining the mitochondrial NADH level in a sample tissue, wherein an increase in NADH level in the sample tissue as compared to NADH level in normal tissue is indicative of a mitochondrial respiratory chain disorder.
  • The method may further include determining mitochondrial NADH level a sample tissue, determining NAD+/NADH ratio in the sample tissue, determining mitochondrial NAD+ level in a normal tissue, and determining NAD+/NADH ratio in the normal tissue, wherein a decrease in the NAD30/NADH ratio in the sample tissue as compared to the NAD30/NADH ratio in normal tissue is indicative of a disease or a disorder associated with a deficiency in frataxin.
  • NAD+ and NADH levels can be done using methods known by those skilled in the art using commercially available assays (Bioassay Systems). Once NAD+ and NADH levels are determined, the NAD+/NADH ratio can be calculated.
  • In another aspect, the present disclosure is directed to a kit for Measuring or detecting mitochondrial protein acetylation. The kit may include reagents for processing a sample, for isolating mitochondria, and for detecting protein acetylation levels (e.g, isolation buffers, colorimetric assay buffers and reagents, and acetylation detection buffers and reagents). In one embodiment, the kit measures acetyl-lysine residues with, for example, an anti-acetyl-lysine-specific antibody. Suitable anti-acetyl-lysine antibodies are commercially available (Immunechem; Cell Signaling).
  • The kit may further use a combination of antibodies such as, for example, an anti-acetyl-lysine-specific antibody and an anti-mitochondrial protein antibody. Suitable anti-mitochondrial protein antibodies can be, for example, antibodies that specifically bind to NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 9 (NDUFA9), antibodies that specifically bind to acetyl-CoA synthetase 2 (AceCS2), antibodies that specifically bind to frataxin, antibodies that specifically bind to complex II 30 kDa subunit, antibodies that specifically bind to complex III Rieske protein, antibodies that specifically bind to NAD-dependent deacetylase sirtuin-3 (SIRT3), antibodies that specifically bind to voltage-dependent anion-selective channel (VDAC) and antibodies that specifically bind to other mitochondrial proteins known to those skilled in the art. The antibodies may be, for example, monoclonal antibodies, polyclonal antibodies and combinations thereof
  • Kits may also use solid supports such as, for example, microtiter plates, membranes (e.g., nitrocellulose, polyvinyl chloride, nylon, polyvinylidene fluoride, diazotized paper) and beads (e.g., polystyrene latex, activated beads, Protein A beads, Protein G beads).
  • The kit can further include instructions for using the reagents for processing a sample, for isolating mitochondria, for measuring mitochondrial protein acetylation, for detecting protein acetylation levels and combinations thereof. The instructions may be provided in the kit packaging and provided by other media such as, for example, a website.
  • The disclosure will be more fully understood upon consideration of the following non-limiting Examples.
  • EXAMPLE Example 1 Materials and Methods
  • Frataxin conditional knockout mouse breeding and genotyping. Mice were obtained from Helene Puccio and bred as described previously to generate the NSE and MCK-Cre conditional knockout animals. PCR was used to detect the presence of a 500 bp product of exon 4 and a 790 bp product of the Cre transgene for genotyping. Mice that were genotyped as FrdaL3/Δ, Cre+ represent the conditional knockout animals.
  • Mouse tissue preparation. For the isolation of whole heart homogenate, animals were sacrificed by CO2 asphyxiation and the tissue was immediately excised, washed and minced in cold phosphate-buffered saline. The tissue was then placed in 1-2 ml of ice-cold complete MPA buffer containing an EDTA-free protease inhibitor cocktail (Roche product #05056489001), and 10 mM nicotinamide (NAM), 200 nM Trichostatin A, and 5 mM sodium butyrate as deacetylase inhibitors. The tissues were then homogenized with at least 10 passes of a motor-driven Potter-Elvehjem homogenizer at high speed. The whole tissue homogenates were then transferred to 1.5 ml tubes and spun at 17,000×g for 10 min at 4° C. in a desktop centrifuge to pellet membranes and tissue debris. The supernatant was saved and immediately frozen on dry-ice and stored at −80° C. for later analysis. Frozen, RIPA buffer solubilized ANT1−/− and COI missense mutation cardiac lysates were provided by Dr. Douglas C. Wallace of The Children's Hospital of Philadelphia.
  • Isolation of Cardiac Mitochondria. Wild-type or frataxin conditional KO mice were sacrificed by CO2 asphyxiation and their hearts were immediately removed and submerged in ice-cold mitochondrial isolation buffer (MIB) containing 220 mM mannitol, 70 rarVi. sucrose, 30 mM Tris-Cl (pH 7.4), 0.5 mM EGTA, and 0.1% BSA. Heart tissue was minced and washed in MIB to eliminate blood and serum contaminants, then quickly weighed. Minced heart tissue from 2-3 animals was submerged in 3 ml/g MIB containing an EDTA-free protease inhibitor cocktail (Roche) and 10 mM nicotinamide (NAM), 200 nM Trichostatin A, and 5 mM sodium butyrate as deacetylase inhibitors. Heart tissue was homogenized with four passes of a motor-driven Potter-Elvehjem homogenizer with a Teflon pestle at medium speed on ice. An additional 7 ml/g MIB was added to the heart homogenate, which was then transferred to a pre-chilled 50 ml conical tube. A crude mitochondrial pellet was obtained via a standard differential centrifugation method. Briefly, nuclei and the heavy cell fraction are removed via centrifugation for 15 minutes at 1000×g at 4° C. The resulting post-nuclear supernatant was subjected to centrifugation for 15 min at 10,000×g to pellet mitochondria. The supernatant was removed as the cytosolic fraction and sedimented mitochondria were gently resuspended in 1 ml ice-cold MIB containing deacetylase inhibitors and then diluted with MIB to a total volume of 10 ml and centrifuged again for 15 min at 10,000×g. The supernatant was then removed and sedimented mitochondria are gently washed in 0.5 M KCl to remove endoplasmic reticulum and lysosome contaminants. For Western blotting, the sedimented mitochondria were solubilized in complete RIPA buffer containing protease and deacetylase inhibitors (see Mouse tissue preparation) and frozen until later analysis. For immunoprecipitations, the pelleted mitochondria were resuspended in ice-cold buffer containing 50 mM Tris-Cl (pH 7.4), 150 mM NaCl, and an EDTA-free protease inhibitor tablet and immediately frozen at −80° C. For NAD+ and NADH measurements, sedimented mitochondria were immediately resuspended in 180 μl of ice-cold 1 mM Tris-Cl pH 7.4, 150 mM NaCl and extracted appropriately (see Mitochondrial NAD+ and NADH measurements).
  • Antibodies and Western Blotting. Antibodies used included anti-acetyl-lysine (Immunechem anti-acetyl-lysine (Cell Signaling) anti-complex II 30 kDa subunit, anti-NDUFA9, anti-complex III Rieske protein (Mitosciences), anti-SIRT3 (provided by E. Verdin, Gladstone Institute, UCSF), anti-frataxin (provided by G. Isaya, Mayo Clinic), anti-SIRT3 D22A3, anti-VDAC D73D12, and anti-histone H3 (Cell Signaling), anti-GAPDH and anti-tubulin (Sigma), and anti-AceCS2 (ACSS 1)(Abeam). Following transfer of the SDS-PAGE gel proteins to nitrocellulose membranes, the membrane was blocked for 45 min in PBS-0.05% Tween-20 supplemented with 5% non-fat dry milk. Signals were visualized with SuperSignal West chemiluminescent substrate (Thermo-Pierce). Western blot signal intensities were determined by density quantification using ImageJ software where appropriate. Protein concentrations were determined using the BCA method (Pierce).
  • Immunoprecipitation of acetyWysine proteins. For immunoprecipitation of acetyl-lysine proteins and detection of NDUFA9 and AceCS2 acetylation, 500 μg to 1 mg of cardiac mitochondria was solubilized for 30 min on ice in 1% lauryl maltoside (Mitosciences) and centrifuged at 16,000×g for 10 min at 4° C. to pellet debris and lipid membrane components. Anti-acetyl-lysine antibody (Cell Signaling) was loaded with the solubilized mitochondrial supernatant at a ratio of 1:25 w/w and incubated overnight at 4° C. with gentle agitation. Following incubation with the anti-acetyl-lysine antibody, the sample was incubated at 4° C. with gentle agitation for an additional 2 hours with 7.5 μl Protein A agarose (Invitrogen) plus 7.5 μl Protein G agarose (Roche). Agarose was collected via a 1 min centrifugation at 400×g at 4° C. with a desktop centrifuge and the unbound mitochondrial supernatant was carefully removed from above the agarose beads. The beads were washed 3 times for 5 min. in 500 μl IP buffer containing 50 mM Tris-Cl (pH 7.4), 150 mM NaCl, 1 mM EGTA, and 0.5% NP-40 at 4° C. and then incubated in 30 μl SDS sample loading buffer and boiled at 95° C. for 5 min. The immunoprecipitated sample was then resolved on a 12% SDS-PAGE and processed for Western blotting.
  • Mitochondrial NAD+ and NADH measurements. NAM was excluded from MIB used to isolate mitochondria for NAD+ measurements as the enzyme nicotinamide phosphoribosyltransferase (Nampt) catalyzes the formation of NMN, an NAD+ precursor, from NAM. NAD(H) measurements were performed with a cycling assay according to the manufacturer's instructions (Bioassay Systems). NAD+ was extracted in the mitochondrial sample via addition of strong acid and then heated at 60° C. to eliminate the presence of the reduced nucleotide (NADH). NADH was extracted in mitochondrial sample homogenate via addition of strong base and then heated at 60° C. to eliminate the presence of the oxidized nucleotide (NAD+). The extractions were neutralized with the opposite extraction buffer and the dehydrogenase cycling assay was performed in which, in the presence of lactate, lactate dehydrogenase, diaphorase and MTT formazan reagent, the concentration of NAD+ or NADH present in the sample is proportional to the amount of reduced TvITF formation which was colorimetrically detected with a plate reader at 562 nm. Sample concentrations were determined via an NAD+ standard curve. The coenzyme concentration was normalized to the protein concentration of the sample.
  • In vitro SIRT3 deacetylation assay 200 μl of 2 μg/μl Frataxin cardiac mitochondria frozen in 50 mM Tris-Cl (pH 7.4). 1.50 mM NaCl was thawed on ice and solubilized in 1% lauryl maltoside for 30 min on ice and centrifuged at 16,000×g for 10 min at 4° C. to pellet debris and lipid membrane components. 15 μl of the soluble mitochondrial supernatant was mixed 1:1 with 15 μl of 2× deacetylase buffer (50 mM Tris-Cl pH 8.0, 150 mM NaCl, 2 mM MgCl2, with or without 20 mM NAD+). 3 μg of recombinant GST-tagged SIRT3 corresponding to amino acids 101-399 of human SIRT3 (Sigma) in 40 mM Tris-Cl pH 8.0, 240 mM NaCl, and 20% glycerol was added to 30 μl (30 μg,) of the 1:1 mitochondrial protein:2× deacetylase buffer mix in a 1.5 ml tube. An appropriate amount of buffer containing only 40 mM Tris-Cl, 240 mM NaCl, and 20% glycerol was added to incubations lacking SIRT3 to equalize the incubation volumes and conditions. The reactions were incubated for 4 hours at 37° C. at 400 rpm in an Eppendorf Thermomixer and briefly centrifuged every 30 min of incubation time to minimize reaction condensation. Following the incubation, the reactions were stopped upon addition of SDS loading buffer, boiled and processed for Western blotting as described previously.
  • Detection of 4-HNE modified SIRT3. For the detection of 4-HNE modified SIRT3, cardiac mitochondrial proteins were solubilized as described earlier and 4-HNE modified proteins were derivatized to a biotin group via a 2 hour incubation in 5 mM EZ-Link hydrazide biotin (Thermo Scientific) in the dark. The samples were dialyzed 1:10000 in PBS overnight to remove excess biotin hydrazide. 100 μg of protein sample was then incubated with 50 μl of NeutrAvidin Agarose Resin (Thermo Scientific) at 4° C. with gentle rocking overnight. Beads were washed 5 times for 5 mM with 500 μl PBS-0.2% Tween 20, then boiled at 95° C. in 35 μl SDS loading buffer and processed for Western blotting as described previously.
  • Statistical Analysis. Statistical significance was determined using a two sample t-test assuming unequal variances. Any P value less than 0.05 was judged to be significant.
  • Example 2
  • In this Example, NSE- and MCK-Cre conditional mouse models for Friedreich's Ataxia (FRDA) were analyzed for protein acetyl-lysine modifications.
  • Specifically, whole heart lysates from wild-type (WT), NSE, and MCK conditional mouse models of FRDA were analyzed by Western blot analysis to assay protein acetyl-lysine modifications. As shown in FIGS. 1 and 2, heart lysates from both the NSE (NSE KO) and MCK (MCK KO) mouse models of FRDA exhibited marked increases in acetyl-lysine modifications as compared to age-matched control hearts. As demonstrated in FIG. 3, hyperacetylation was localized to cardiac mitochondria. As shown in FIGS. 4 and 5, the increases in acetyl-lysine modifications as compared to age-matched control hearts were statistically significant. The differences were most dramatic in proteins with an estimated molecular weight between approximately 30 and 75 kDa (FIG. 1).
  • Example 3
  • In this Example, sub-fractionation of heart samples was performed to determine the sub-cellular distribution of hyperacetylated proteins.
  • Analysis of the purity of these mitochondrial preparations using antibodies to Historic H3 (a cytoplasmic marker), Gapdh (a cytoplasmic marker), VDAC (a mitochondrial marker) and Complex II (a mitochondrial marker) showed that nuclear and cytosolic proteins were excluded and they were highly enriched for markers of both the outer and inner mitochondrial membranes (FIG. 6). Using day 24 wild-type (WT, n=2) control and NSE-Cre frataxin-deficient cardiac mitochondrial preparations (n=2), Western blot analysis was performed for acetyl-lysine modifications.
  • Control cardiac mitochondria exhibited several acetylated proteins detectable by Western blot. However, frataxin-deficient cardiac mitochondria displayed marked hyperacetylation of numerous proteins (FIGS. 3 and 7). This was accompanied by a characteristic downregulation of respiratory complex I and II (succinate dehydrogenase) that was present as early as 7 days post-natal (FIG. 9). Levels of the dominant mitochondria-localized NAD+-dependent deacetylase SIRT3 displayed a mild, though insignificant, increase in frataxin-deficient mitochondrial preparations (FIGS. 7 and 8). These results indicated that the hyperacetylation observed at the level of whole cardiac lysate was predominantly localized to mitochondria.
  • Example 4
  • In this Example, the developmental profile of mitochondrial protein acetylation was determined.
  • Specifically, the NSE-Cre mouse models of FRDA begin to develop cardiac hypertrophy in the second week of life. Thus, cardiac mitochondria from WT control and NSE-Cre mice at post-natal days 7, 17, and 24 were prepared and analyzed by Western blot analysis.
  • As shown in FIGS. 9 and 10, at post-natal day 7, NSE-Cre cardiac mitochondrial proteins exhibited only a mildly increased acetylation state as compared to their wild-type (WT) counterparts. However, at post-natal day 17, NSE-Cre cardiac mitochondria displayed increased acetylation of cardiac mitochondrial proteins as compared to their WT counterparts, which became more dramatic by post-natal day 24. The progressive increase in cardiac mitochondrial protein acetylation over this time frame corresponded with the development of cardiac hypertrophy in the NSE-Cre models (FIG. 11).
  • Example 5
  • In this Example, the redox states of WT and frataxin deficient cardiac mitochondria preparations were determined.
  • Individual measurements were performed on a pooled sample of at least 300 μg of fresh cardiac mitochondria derived from 2-3 hearts and normalized to total mitochondrial protein input. As shown in FIG. 12, WT heart mitochondria displayed robust NAD+ levels and over 100-fold less NADH by comparison, which is consistent with highly oxidative cardiac catabolism and a continuous demand for carbon fuels, Frataxin deficient mitochondria displayed a mild, though insignificant, increase in NAD+ levels when compared to WT. In contrast, frataxin deficient mitochondrial NADH levels were, on average, over 95-fold greater than in WT mitochondria (P<0.005) resulting in a corresponding NAD+/NADH ratio that was 85-fold less than in WT animals (FIG. 12 inset). The observed accumulation of NADH and consequent shift in mitochondrial redox state is consistent with the impairments of mitochondrial respiration found in both human FRDA patients, and the NSE and MCK animal models.
  • Example 6
  • In this Example, differential modification of SIRT3 by 4-HNE in frataxin deficient cardiac mitochondria was determined.
  • Specifically, all reactive aldehyde containing mitochondrial proteins following their derivatization to biotin hydrazide were pulled down, and then analyzed by Western blot for SIRT3. As shown in FIG. 13, SIRT3 in WT heart mitochondria exhibited a small amount of 4-HNE modification. In contrast, SIRT3 in frataxin-deficient mitochondria exhibited a marked increase in 4-HNE modification suggesting that SIRT3 may be directly inhibited via carbonyl group adduction in the setting of frataxin deficiency.
  • Example 7
  • In this Example, the acetylation states of known targets of SIRT3-mediated deacetylation was determined.
  • Mitochondrial acetyl-lysine proteins were immunoprecipitated and analyzed by Western blot for NDUFA9. Consistent with previous findings, a minimal amount of NDUFA9 acetyl-lysine signal in the WT (+/+) mitochondrial immunoprecipitate was observed. However, the frataxin-deficient (−/−) mitochondrial immunoprecipitate displayed a greater acetylated NDUFA9 signal despite downregulation of NDUFA9 as seen in the input (FIG. 14). The acetylation state of acetyl-CoA synthetase 2 (AceCS2) was also analyzed. Similarly, this analysis revealed a greater amount of acetylated AceCS2 in the frataxin-deficient (−/−) condition as compared to the WT (+/+) (FIG. 15). The increased acedation states of two known targets of SIRT3-mediated deacetylation indirectly demonstrated that SIRT3 is inhibited in frataxin-deficient cardiac mitochondria. Importantly, increases in the acetylation states of NDUFA9 and AceCS2 were linked to a decrease in the activity of respiratory complex I and the synthesis of activated acetate, respectively.
  • Example 8
  • In this Example, the rescue of inhibited endogenous SIRT3 by the addition of SIRT3 and NAD+ in vitro was determined.
  • Specifically, solubilized WT and frataxin-deficient cardiac mitochondrial proteins were incubated with glutathione S-transferase tagged, processed human recombinant SIRT3 (GST-hSIRT3) in the presence or absence of NAD+ to assay for changes in acetylation states. Incubating solubilized WT or frataxin cardiac mitochondrial homogenates with NAD+ alone caused no change in acetylation signal. In contrast, incubating frataxin deficient mitochondrial protein with 3 μg of GST-hSIRT3 and NAD+ resulted in a marked reduction of acetyl-lysine signal from nearly every protein band, while completely eliminating the acetyl-lysine signal of multiple bands (FIG. 16). Furthermore, the observed reduction in acetylation signal was abolished upon withdrawal of NAD+ from the incubation buffer, demonstrating that the hyperacetylated protein lysine residues in frataxin-deficient cardiac mitochondria were specifically sensitive to NAD+-dependent SIRT3-mediated deacetylation and that the observed hyperacetylation was not caused by a general increase in non-specific lysine acetylation. Taken together, these data strongly suggest that hyperacetylation in frataxin deficient mitochondria was due to both redox-state and lipid-peroxidation-mediated inhibition of endogenous SIRT3.
  • Example 9
  • In this Example, alterations in cardiac acetylation profiles of other mitochondrial disorders caused by respiratory chain defects were determined
  • Whole cardiac lysates were obtained from the cytochrome oxidase I (COI) mouse model, which harbors a mtDNA mutation causing a defect of respiratory complex IV, and the adenine nucleotide translocase knockout (ANT1−/−) mouse model, which models a non-respiratory chain defect with deficiency of ATP translocation. These mice model the human mitochondrial disorders, autosomal dominant progressive external ophthalmoplegia (adPEO) and Leber's Hereditary Optic Neuropathy (LHON), respectively, and both models develop cardiomyopathy.
  • Western blot analysis of cardiac acetylation revealed mild to moderate increases in acetylation in 12 month old COI hearts as compared to 13 month-old control hearts (FIGS. 17 and 18). Interestingly, 20 month old ANT1−/− hearts exhibited a small decrease in acetyl-lysine profiles when compared to 18 month old control hearts (FIGS. 19 and 20). Taken together with data from the frataxin deficient mice, these results suggest that increases in protein acetylation may be a common feature of respiratory chain malfunction in the mammalian heart.
  • Example 10
  • In this Example, mitochondrial acetylation status in frataxin-deficient mice treated with TAT-Frataxin fusion protein was determined.
  • The TAT-Frataxin fusion protein was prepared as described in Vyas et al. (Hum. Mol. Genet. 2012, 21(6):1230-1247). NSE-Cre Frataxin knockout mice (FXN−/−) were treated with TAT-Frataxin for 2 weeks (FXN−/−+TAT-FXN; FIG. 21A lanes 3 and 4) or phosphate buffered saline (FXN−/−; FIG. 21A lanes 1 and 2). Specifically, mice were dosed according to body weight with a total volume of approximately 20 μl/gram of weight administered intraperitoneally. Whole heart lysate from 4 week old NSE-Cre FXN knockout mice was quantified and equal loading of each lane was verified by Coomassie staining. Western blots were probed with anti-acetyl lysine antibody and pixel intensity was quantified for the top 4 bands identified in lanes 1-4 of FIG. 21A.
  • As illustrated in FIG. 21B, treatment of mice TAT-Frataxin decreased protein acetylation in NSE-Cre FXN knockout mice (NSE KO+TAT-FXN). Therefore, mitochondrial protein acetylation status can be used as a biomarker to monitor treatment in a mouse knockout model of Friedreich's Ataxia, which is associated with a deficiency in frataxin.
  • The Examples described above demonstrate that a deficiency in frataxin can cause progressive hyperacetylation of mitochondrial proteins. More specifically, hyperacetylation of mitochondrial proteins may be due to the inhibition of a SIRT3 deacetylase. SIRT3 may respond to the flux of mitochondrial. NAD+ and NADH, which is determined, in large part, by the capacity of the respiratory chain to oxidize NADH. This capacity is severely decreased in FRDA, as well as in other mitochondrial defects such as cytochrome c oxidase (complex IV) deficiency, causing an accumulation of NADH and, consequently, a redox state of perceived nutrient excess. It has now been discovered that hyperacetylation of numerous mitochondrial proteins correlates with the inhibition of the NAD+-dependent SIRT3 deacetylase. This inhibition is caused by an 85-fold decrease in mitochondrial NAD+/NADH ratio and direct carbonyl group modification of a NAD-dependent deacetylase. It has also been discovered that the inhibition may be rescued by the administration of a NAD-dependent deacetylase and NAD+. It has further been discovered that protein hyperacetylation provides a specific biomarker of a deficiency in frataxin. Thus, loss of frataxin causes hyperacetylation of proteins because the respiratory chain is defective and the Krebs cycle.
  • In view of the above, it will be seen that the several advantages of the disclosure are achieved and other advantageous results attained. As various changes could be made in the above devices and methods without departing from the scope of the disclosure, it is intended that all matter contained in the above description and shown in the accompanying drawings shall be interpreted as illustrative and not in a limiting sense.
  • Then introducing elements of the present disclosure or the various versions, embodiment(s) or aspects thereof, the articles “a”, “an”, “the” and “said” are intended to mean that there are one or more of the elements. The terms “comprising”, “including” and “having” are intended to be inclusive and mean that there may be additional elements other than the listed elements.

Claims (16)

What is claimed is:
1. A method of screening for a disease or a disorder associated with a deficiency in frataxin in a subject, the method comprising: determining the acetylation status of a mitochondrial protein in a sample tissue; and determining the acetylation status of a mitochondrial protein in a normal tissue, wherein an increase in acetyl-lysine status of the mitochondrial protein in the sample tissue as compared to acetyl-lysine status of the protein in the normal tissue is indicative of the disease or the disorder associated with the deficiency in frataxin.
2. The method of claim 1, wherein the sample tissue and the normal tissue is selected from the group consisting of liver, heart, white blood cells, and neural tissue.
3. A method of screening for a disease or a disorder associated with a deficiency in frataxin in a subject, the method comprising: determining the mitochondrial NADH level in a sample tissue; and determining the mitochondrial NADH level in a normal tissue, wherein an increase in the NADH level in the sample tissue as compared to the NADH level in the normal tissue is indicative of the disease or the disorder associated with the deficiency in frataxin.
4. The method of claim 3, further comprising determining the NAD+ level in the sample tissue and determining the NAD+ level in the normal tissue.
5. The method of claim 4, further comprising determining the NAD+/NADH ratio in the sample tissue and determining the NAD+/NADH ratio in the normal tissue, wherein a decrease in the NAD+/NADH ratio in the sample tissue as compared to the NAD+/NADH ratio in normal tissue is indicative of a disease or a disorder associated with a deficiency in frataxin.
6. A kit for measuring levels of mitochondrial protein acetylation comprising an anti-acetyl-lysine-specific antibody and an antibody that specifically binds to a mitochondrial protein.
7. The kit of claim 6, wherein the antibody that specifically binds to a mitochondrial protein is selected from the group consisting of an antibody that specifically binds to NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 9 (NDUFA9), an antibody that specifically binds to acetyl-CoA synthetase 2 (AceCS2), an antibody that specifically binds to frataxin, an antibody that specifically binds to complex II 30 kDa subunit, an antibody that specifically binds to complex HI Rieske protein, an antibody that specifically binds to NAD-dependent deacetylase sirtuin-3 (SIRT3), an antibody that specifically binds to voltage-dependent anion-selective channel (VDAC), and combinations thereof
8. The kit of claim 6 further comprising at least one of a reagent for processing a sample, a reagent for isolating mitochondria, a reagent for measuring mitochondrial protein acetylation, and a reagent for measuring mitochondrial protein acetylation.
9. A method of detecting progression of a disease or a disorder associated with a deficiency in frataxin in a subject having or suspected of having the disease or the disorder associated with the deficiency in frataxin, the method comprising: measuring mitochondrial protein acetylation in a first chronological sample from the subject; and measuring mitochondrial protein acetylation in a second chronological sample from the subject, wherein an increase in mitochondrial protein acetylation in the second chronological sample as compared to the mitochondrial protein acetylation in the first chronological sample indicates progression of the disease or the disorder.
10. The method of claim 9, wherein the disease or the disorder associated with the deficiency in frataxin is selected from the group consisting of Friedreich's Ataxia, Parkinson's Disease, Alzheimer's Disease, alcoholism, ischemic heart disease, dementia, Huntington's disease, Amyotrophic lateral sclerosis, cytochrome c oxidase deficiency, autosomal dominant progressive external ophthalmoplegia, and Leber's Hereditary Optic Neuropathy.
11. The method of claim 9, further comprising. prior to the measuring step, steps of: collecting the sample from the subject; and isolating mitochondria front the sample.
12. The method of claim 9, wherein the subject is a human.
13. The method of claim 9, wherein mitochondrial protein acetylation is measured utilizing Western blotting with an antibody that detects acetyl-lysine residues.
14. A method of monitoring effectiveness of a therapy in a subject having or suspected of having a disease or a disorder associated with a deficiency in frataxin, the method comprising: measuring mitochondrial protein acetylation in at least a first chronological sample; administering the therapy; measuring mitochondrial protein acetylation in at least a second chronological sample from the subject, analyzing the mitochondrial protein acetylation in at least the first chronological sample and the mitochondrial protein acetylation in at least the second chronological sample, wherein a decrease in mitochondrial protein acetylation in the second chronological sample as compared to mitochondrial protein acetylation in a first chronological sample indicates effectiveness of the therapy.
15. The method of claim 14, wherein the disease or the disorder associated with the deficiency in frataxin is selected front the group consisting of Friedreich's Ataxia, Parkinson's Disease, Alzheimer's Disease, alcoholism, ischemic heart disease, dementia, Huntington's disease, Amyotrophic lateral sclerosis, cytochrome c oxidase deficiency, autosomal dominant progressive external ophthalmoplegia, and Leber's Hereditary Optic Neuropathy.
16. A method of screening for agents useful for treating a disease or a disorder associated with a deficiency in frataxin, the method comprising: measuring mitochondrial protein acetylation in a subject having hyperacetylated mitochondrial proteins; administering an agent suspected of modulating protein acetylation to the subject; and measuring mitochondrial protein acetylation after administration of the agent, wherein the agent is considered to be a candidate for treating the disease or the disorder associated with the deficiency in frataxin if the mitochondrial protein acetylation is decreased in the sample after administration of the agent.
US15/646,172 2012-03-02 2017-07-11 Biomarker for determining mitochondrial damage in friedreich's ataxia Abandoned US20170307636A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/646,172 US20170307636A1 (en) 2012-03-02 2017-07-11 Biomarker for determining mitochondrial damage in friedreich's ataxia

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201261605783P 2012-03-02 2012-03-02
US201261607918P 2012-03-07 2012-03-07
PCT/US2013/028609 WO2013130964A1 (en) 2012-03-02 2013-03-01 A biomarker for determining mitochondrial damage in friedreich's ataxia
US201414382367A 2014-09-02 2014-09-02
US15/646,172 US20170307636A1 (en) 2012-03-02 2017-07-11 Biomarker for determining mitochondrial damage in friedreich's ataxia

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US14/382,367 Continuation US9733258B2 (en) 2012-03-02 2013-03-01 Biomarker for determining mitochondrial damage in friedreich's ataxia
PCT/US2013/028609 Continuation WO2013130964A1 (en) 2012-03-02 2013-03-01 A biomarker for determining mitochondrial damage in friedreich's ataxia

Publications (1)

Publication Number Publication Date
US20170307636A1 true US20170307636A1 (en) 2017-10-26

Family

ID=49083334

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/382,367 Active US9733258B2 (en) 2012-03-02 2013-03-01 Biomarker for determining mitochondrial damage in friedreich's ataxia
US15/646,172 Abandoned US20170307636A1 (en) 2012-03-02 2017-07-11 Biomarker for determining mitochondrial damage in friedreich's ataxia

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/382,367 Active US9733258B2 (en) 2012-03-02 2013-03-01 Biomarker for determining mitochondrial damage in friedreich's ataxia

Country Status (2)

Country Link
US (2) US9733258B2 (en)
WO (1) WO2013130964A1 (en)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007133689A2 (en) 2006-05-12 2007-11-22 Cell Signaling Technology, Inc. Reagents for the detection of protein acetylation signaling pathways
US20100015724A1 (en) * 2006-08-17 2010-01-21 Peter Hornbeck Lysine acetylation sites
US20100209427A1 (en) 2008-09-24 2010-08-19 Yu Li Lysine acetylation sites

Also Published As

Publication number Publication date
US20150132769A1 (en) 2015-05-14
WO2013130964A1 (en) 2013-09-06
US9733258B2 (en) 2017-08-15

Similar Documents

Publication Publication Date Title
Li et al. Mitochondrial dysfunction and mitophagy defect triggered by heterozygous GBA mutations
Cai et al. The protective effect of selenoprotein M on non-alcoholic fatty liver disease: the role of the AMPKα1–MFN2 pathway and Parkin mitophagy
Di Domenico et al. Oxidative signature of cerebrospinal fluid from mild cognitive impairment and Alzheimer disease patients
Chiang et al. Dysregulation of C/EBPα by mutant Huntingtin causes the urea cycle deficiency in Huntington's disease
Shen et al. Redox proteomics identification of specifically carbonylated proteins in the hippocampi of triple transgenic Alzheimer's disease mice at its earliest pathological stage
Di Domenico et al. Redox proteomics analysis of HNE-modified proteins in Down syndrome brain: clues for understanding the development of Alzheimer disease
Shimamoto et al. Functional characterization of FABP3, 5 and 7 gene variants identified in schizophrenia and autism spectrum disorder and mouse behavioral studies
Li et al. Mechanism of hyperinsulinism in short-chain 3-hydroxyacyl-CoA dehydrogenase deficiency involves activation of glutamate dehydrogenase
Houten et al. Mitochondrial NADP (H) deficiency due to a mutation in NADK2 causes dienoyl-CoA reductase deficiency with hyperlysinemia
Fischer-Zirnsak et al. Recurrent de novo mutations affecting residue Arg138 of pyrroline-5-carboxylate synthase cause a progeroid form of autosomal-dominant cutis laxa
Castegna et al. Oxidative stress and reduced glutamine synthetase activity in the absence of inflammation in the cortex of mice with experimental allergic encephalomyelitis
US7314720B2 (en) Method of diagnosing non-alcoholic steatohepatitis (nash) using molecular markers
Stiburkova et al. Novel homozygous insertion in SLC2A9 gene caused renal hypouricemia
Gallego-Villar et al. In vivo evidence of mitochondrial dysfunction and altered redox homeostasis in a genetic mouse model of propionic acidemia: implications for the pathophysiology of this disorder
Zellner et al. Comparative platelet proteome analysis reveals an increase of monoamine oxidase-B protein expression in Alzheimer's disease but not in non-demented Parkinson's disease patients
Tramutola et al. Proteomic identification of altered protein O-GlcNAcylation in a triple transgenic mouse model of Alzheimer's disease
Perez et al. Two hypomorphic alleles of mouse Ass1 as a new animal model of citrullinemia type I and other hyperammonemic syndromes
Subramanian et al. A pantothenate kinase-deficient mouse model reveals a gene expression program associated with brain coenzyme a reduction
Hempel et al. LYRM7-associated complex III deficiency: a clinical, molecular genetic, MR tomographic, and biochemical study
Yamamoto et al. Hippocampal level of neural specific adenylyl cyclase type I is decreased in Alzheimer’s disease
Aboudehen et al. Transcription factor hepatocyte nuclear factor–1β regulates renal cholesterol metabolism
WO2017059118A1 (en) Compositions and methods for identifying and treating dystonia disorders
US8110371B2 (en) Identifying organ damage
Freund et al. Response of the mitochondrial proteome of rat renal proximal convoluted tubules to chronic metabolic acidosis
Schuck et al. Evidence that the major metabolites accumulating in medium-chain acyl-CoA dehydrogenase deficiency disturb mitochondrial energy homeostasis in rat brain

Legal Events

Date Code Title Description
AS Assignment

Owner name: INDIANA UNIVERSITY RESEARCH AND TECHNOLOGY CORPORA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PAYNE, R. MARK;WAGNER, GREGORY R.;PRIDE, P. MELANIE;AND OTHERS;SIGNING DATES FROM 20130307 TO 20130308;REEL/FRAME:042964/0317

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: THE TRUSTEES OF INDIANA UNIVERSITY, INDIANA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:INDIANA UNIVERSITY RESEARCH AND TECHNOLOGY CORPORATION;REEL/FRAME:049801/0947

Effective date: 20190719