WO2019085804A1 - Procédés et utilisation pour détecter et inhiber mg53 acellulaire - Google Patents

Procédés et utilisation pour détecter et inhiber mg53 acellulaire Download PDF

Info

Publication number
WO2019085804A1
WO2019085804A1 PCT/CN2018/111695 CN2018111695W WO2019085804A1 WO 2019085804 A1 WO2019085804 A1 WO 2019085804A1 CN 2018111695 W CN2018111695 W CN 2018111695W WO 2019085804 A1 WO2019085804 A1 WO 2019085804A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
antibody
certain embodiments
antigen
binding fragment
Prior art date
Application number
PCT/CN2018/111695
Other languages
English (en)
Inventor
Rui-Ping Xiao
Yan Zhang
Fengxiang LV
Xinli HU
Original Assignee
Peking University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Peking University filed Critical Peking University
Publication of WO2019085804A1 publication Critical patent/WO2019085804A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • 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
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • 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
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/04Endocrine or metabolic disorders
    • G01N2800/042Disorders of carbohydrate metabolism, e.g. diabetes, glucose metabolism
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/04Endocrine or metabolic disorders
    • G01N2800/044Hyperlipemia or hypolipemia, e.g. dyslipidaemia, obesity

Definitions

  • the present disclosure relates to the art of biomedicine, specifically, the present disclosure relates to a method for detecting cell-free MG53 or detecting a related disease or predicting a risk of an MG53 related disease, and a method for reducing or inhibiting cell-free MG53 activity in a subject, and compositions used for the methods.
  • Mitsugumin 53 (MG53) , also known as TRIM72, is a member of the Tripartite motif-containing proteins (TRIM) family. MG53 comprises a TRIM motif at the N-terminus and a SPRY motif at the C-terminus, and the TRIM motif consists of successively linked Ring, B-box and coiled-coil domains (see Chuanxi Cai et al., the Journal of Biological Chemistry, Vol. 284 (5) , 3314-3322 (2009) ) . . MG53 plays a variety of roles throughout the body, but is mainly expressed in striated muscles, and is essential for maintaining the homeostasis of skeletal muscle and the heart.
  • MG53 was previously found to have cell membrane repair function and cardioprotective function (see, e.g., Chuanxi Cai et al., Nature Cell Biology, Vol. 11, 56-64 (2009) ; CN101797375B) . In addition, further studies have found that MG53 also plays a protective role in ischemic preconditioning (IPC) and ischemic postconditioning (PostC) , by activation of the reperfusion injury salvage kinase (RISK) pathway.
  • IPC ischemic preconditioning
  • PostC ischemic postconditioning
  • the N-and C-termini of the MG53 molecule can bind to Caveolin-3 and P85-PI3K kinases respectively to form a complex, which activates the RISK pathway to elicit cardiac protection (see Chun-Mei Cao et al., Circulation 121, 2565-2574, (2010) ) .
  • MG53 has E3 ubiquitin ligase activity, which contributes tothe development of insulin resistance and metabolic syndrome.
  • the Ring domain of the TRIM motif at the N-terminus of MG53 binds to insulin receptor (IR) and insulin receptor substrate-1 (IRS1) , and mediates the ubiquitination and subsequent degradation of these proteins by the proteasome, thereby blocking the insulin signaling pathway and leading to insulin resistance and associated metabolic diseases such as obesity, diabetes, hypertension, dyslipidemia, etc.
  • IR insulin receptor
  • IRS1 insulin receptor substrate-1
  • the present disclosure relates to a method for detecting an MG53 related disease or predicting a risk of an MG53 related disease, and a method for reducing or inhibiting cell-free MG53 activity in a subject. Additionally, the present disclosure also relates to an MG53 antibody or antigen-binding fragment thereof, a nucleic acid encoding such antibody or antigen-binding fragment thereof, a clone or expression vector including such nucleic acid, a host cell including the clone or expression vector, and a pharmaceutical composition including the products above.
  • the present disclosure also relates to use of a detection agent for detecting cell-free MG53 in the manufacture of a kit, and use of the MG53 inhibitor in the manufacture of a medicament for reducing or inhibiting cell-free MG53 in a subject.
  • the present disclosure relates to a method for detecting an MG53 related disease or predicting a risk of an MG53 related disease, comprising the following steps: a) obtaining a test sample; and b) detecting cell-free MG53 in the test sample.
  • the test sample is body fluid. In certain embodiments, the test sample is selected from whole blood, plasma, serum, tissue fluid, urine and sweat. In certain embodiments, the test sample is selected from whole blood, plasma or serum. In certain embodiments, the test sample substantially does not contain any cells.
  • the step b) comprises contacting an MG53 detection agent with the test sample.
  • the MG53 detection agent is an MG53 antibody or antigen-binding fragment thereof, or an MG53 ligand or an MG53-binding fragment thereof.
  • the MG53 detection agent is a small molecule compound capable of binding MG53.
  • the MG53 detection agent has a detectable label.
  • the detectable label is luminescent, magnetic, radioactive, or enzymatically active.
  • the step b) comprises conducting radioimmunoassay, Western blot analysis, proximity ligation assay, immunofluorescence assay, enzyme immunoassay, immunoprecipitation, chemiluminescence, immunohistochemistry assay, dot blot assay or slit blot method.
  • the method of the present disclosure further comprises: c) comparing the detected value of MG53 obtained from step b) with a reference value.
  • the reference value is obtained from a reference sample.
  • the reference sample and the test sample are from the same subject or different subjects.
  • the reference sample and the test sample are from the same subject, but the time at which the reference sample was collected is earlier or later for a period of time than the test sample.
  • the subject accepts treatment or the health condition thereof has changed.
  • the reference sample is obtained from a healthy subject or a subject suffering from an MG53 related disease.
  • the subject is a human or a non-human mammal.
  • the MG53 related disease is metabolic syndrome. In certain embodiments, the MG53 related disease is a glucose metabolism or lipid metabolism related disease. In certain embodiments, the glucose metabolism or lipid metabolism related disease is selected from the group consisting of insulin resistance, diabetic cerebrovascular disease, diabetic ocular complication, diabetic neuropathy, diabetic foot, hyperinsulinemia, hypercholesterolemia, hyperglycemia, hyperlipidemia, hypertension and obesity.
  • the present disclosure relates to a method for reducing or inhibiting activity of cell-free MG53 in a subject, comprising administrating an effective amount of MG53 inhibitor to a subject in need thereof.
  • the MG53 inhibitor specifically binds to MG53.
  • the MG53 inhibitor is an MG53 antibody or antigen-binding fragment thereof, an MG53 ligand or an MG53-binding fragment thereof, or a nonfunctional protein fragment of MG53.
  • the MG53 antibody or antigen-binding fragment thereof comprises three heavy chain complementarity determining regions comprised in a sequence of SEQ ID NOs: 1, 9, 17, 25 or 33.
  • the MG53 antibody or antigen-binding fragment thereof further comprises three light chain complementary determining regions comprised in a sequence of SEQ ID NOs: 2, 10, 18, 26 or 34.
  • the MG53 antibody or antigen-binding fragment thereof comprises a heavy chain, and the heavy chain comprises a sequence of SEQ ID NOs: 1, 3, 9, 11, 17, 19, 25, 27, 33 or 35, or a sequence having at least 80%, 85%, 90%, 95%or 99%sequence homology thereof.
  • the MG53 antibody or antigen-binding fragment thereof comprises a light chain, and the light chain comprises a sequence of SEQ ID NOs: 2, 4, 10, 12, 18, 20, 26, 28, 34 or 36, or a sequence having at least 80%, 85%, 90%, 95%or 99%sequence homology thereof.
  • the MG53 inhibitor is a small molecule compound. In certain embodiments, the MG53 inhibitor is a compound having Formula Ia or Formula Ib, or a pharmaceutically acceptable salt thereof,
  • L is a chemical bond, or an optionally substituted C 1 -C 12 alkylene
  • M is a chemical bond, or an optionally substituted C 6 -C 12 arylene or a 5-12 membered heterocyclic subunit
  • Y is an optionally substituted C 6 -C 12 aryl or 5-12 membered heterocyclic group
  • Q is an optionally substituted C 1 -C 12 alkylene or C 2 -C 12 alkenylene
  • T is an optionally substituted C 6 -C 12 aryl or 5-12 membered heterocyclic group
  • each R 1 is independently hydrogen, nitro, halogen, hydroxyl, cyano, or an optional
  • L is a chemical bond or C 1 -C 3 alkylene.
  • M is a chemical bond, or an optionally substituted phenylene or furylene.
  • Y is an optionally substituted phenyl, furyl, quinolinyl or benzo dioxolanyl.
  • Q is an optionally substituted C 1 -C 3 alkylene or C 2 -C 3 alkenylene.
  • T is an optionally substituted phenyl or 1, 3, 4-thiadiazolyl.
  • the MG53 inhibitor is a compound having Formula II or a pharmaceutically acceptable salt thereof,
  • A is hydrogen, or an optionally substituted C 6 -C 12 aryl or 5-12 membered heterocyclic group
  • G is -C (O) -O-R 3 , wherein R 3 is an optionally substituted C 1 -C 12 alkyl
  • J is an optionally substituted C 6 -C 12 aryl
  • G and J together with the carbon atom linked thereto, form wherein each R 4 is independently hydrogen, nitro, halogen, hydroxyl, cyano, or an optionally substituted C 1 -C 12 alkyl or C 1 -C 12 alkoxy, n is any integral from 1 to 4
  • B is a chemical bond, or an optionally substituted C 6 -C 12 arylene or 5-12 membered heterocyclic subunit
  • D is a chemical bond, or an optionally substituted -O-R 5 -, wherein R 5 is optionally substituted C 1 -C 12 alkylene
  • E is an optionally substituted C 6 -C 12 aryl
  • A is an optionally substituted naphthyl, phenyl or thienyl.
  • G is -C (O) -O-R 3 , wherein R 3 is an optionally substituted C 1 -C 3 alkyl; J is optionally substituted phenyl; or G and J, together with the carbon atom linked thereto, form wherein each R 4 is hydrogen.
  • B is a chemical bond, or an optionally substituted phenylene or furylene.
  • D is a chemical bond, or an optionally substituted -O-CH 2 -.
  • E is an optionally substituted phenyl.
  • the MG53 inhibitor is a compound having Formula III or a pharmaceutically acceptable salt thereof,
  • A is hydrogen, or an optionally substituted C 6 -C 12 aryl or 5-12 membered heterocyclic group; each Y is independently optionally substituted C 6 -C 12 aryl or 5-12 membered heterocyclic group; R 7 is hydrogen, nitro, halogen, hydroxyl, cyano, or optionally substituted C 1 -C 12 alkyl or C 1 -C 12 alkoxy.
  • A is an optionally substituted phenyl.
  • Y is an optionally substituted phenyl.
  • R 7 is hydrogen, nitro, halogen, hydroxyl or cyano.
  • the “optionally substituted” mentioned in any of the embodiments above refers to not substituted by any substituent group or substituted by one or more substituent groups selected from the followings: nitro, halogen, hydroxyl, cyano, C 1 -C 12 alkyl, C 1 -C 12 alkoxy, benzamide group, or -C (O) -O-R 6 , wherein the C 1 -C 12 alkyl, C 1 -C 12 alkoxy or benzamide group can further be substituted by nitro, halogen, hydroxyl, cyano, C 1 -C 3 alkyl or C 1 -C 3 alkoxy, wherein R 6 is hydrogen or C 1 -C 3 alkyl.
  • the MG53 inhibitor is a compound of the followings or a pharmaceutically acceptable salt thereof.
  • the method of the present disclosure can be used to ameliorate, treat or predict an MG53 related disease or a symptom thereof.
  • the MG53 related diseased is metabolic syndrome.
  • the MG53 related diseased is a glucose metabolism or lipid metabolism related disease.
  • the glucose metabolism or lipid metabolism related disease is selected from the group consisting of insulin resistance, diabetic cerebrovascular disease, diabetic ocular complication, diabetic neuropathy, diabetic foot, hyperinsulinemia, hypercholesterolemia, hyperglycemia, hyperlipidemia, hypertension and obesity.
  • diabetic cerebrovascular disease includes cerebral arteriosclerosis, ischemic cerebrovascular diseases, cerebral hemorrhage, encephalatrophy and cerebral infarction.
  • diabetic ocular complication includes diabetic retinopathy, diabetic cataract, and uveitis and blindness associated with diabetes.
  • diabetic neuropathy includes diabetic peripheral neuropathy.
  • the present disclosure relates to an MG53 antibody or antigen-binding fragment thereof, comprising three heavy chain complementary determining regions comprised in a sequence of SEQ ID NOs: 1, 9, 17, 25 or 33.
  • the MG53 antibody or antigen-binding fragment thereof further comprises three light chain complementary determining regions comprised in a sequence of SEQ ID NOs: 2, 10, 18, 26 or 34.
  • the MG53 antibody or antigen-binding fragment thereof comprises a heavy chain, wherein the heavy chain has an amino acid sequence of SEQ ID NOs: 1, 3, 9, 11, 17, 19, 25, 27, 33 or 35, or a sequence having at least 80%, 85%, 90%, 95%or 99%sequence homology thereof.
  • the MG53 antibody or antigen-binding fragment thereof comprises further comprises a light chain, wherein the heavy chain has an amino acid sequence of SEQ ID NOs: 2, 4, 10, 12, 18, 20, 26, 28, 34 or 36, or a sequence having at least 80%, 85%, 90%, 95%or 99%sequence homology thereof.
  • the present disclosure relates to an antibody or antigen-binding fragment, competing for binding to MG53 with the antibody or antigen-binding fragment of any of the embodiments above.
  • the present disclosure relates to an isolated nucleic acid, encoding the antibody or antigen-binding fragment of any of the embodiments above.
  • the isolated nucleic acid comprises a nucleotide sequence of SEQ ID NOs: 5, 6, 7, 8, 13, 14, 15, 16, 21, 22, 23, 24, 29, 30, 31, 32, 37, 38, 39 or 40, or a sequence having at least 80%, 85%, 90%, 95%or 99%sequence homology thereof.
  • the present disclosure relates to a clone or expression vector, comprising the isolated nucleic acid of any of the embodiments above.
  • the present disclosure relates to a host cell, comprising the clone or expression vector of any of the embodiments above.
  • the present disclosure relates to a pharmaceutical composition, comprising the antibody or antigen-binding fragment of any of the embodiments above, the clone or expression vector of any of the embodiments above, or the cell of any of the embodiments above, and a pharmaceutically acceptable excipient.
  • the present disclosure relates to use of a detection agent for detecting cell-free MG53 in the manufacture of a kit, which is used for detecting an MG53 related disease, predicting a risk or development of an MG53 related disease, identifying potential MG53 inhibitor, evaluating therapeutic effect for an MG53 related disease, or detecting activity of cell-free MG53.
  • the detection agent for detecting cell-free MG53 can be the MG53 detection agent of any of the embodiments above.
  • the MG53 related disease can be any MG53 related diseases of any of the embodiments above.
  • the present disclosure relates to use of an MG53 inhibitor in the manufacture of a medicament for reducing or inhibiting cell-free MG53 in a subject.
  • the MG53 inhibitor can be the MG53 inhibitor of any of the embodiments above.
  • the method can be used to ameliorate, treat or predict an MG53 related disease or a symptom thereof.
  • the MG53 related disease can be any MG53 related diseases of any of the embodiments above.
  • Figure 1 Pattern diagram of the molecular structure of wild type MG53.
  • Figure 2 Chart showing results of SPR experiment for detecting affinity between an MG53 antibody and antigen protein.
  • K D of the antibody 17.7 nM.
  • Figure 4 Detection of MG53 content in serum of MG53 -/- mice (MG53 gene knocked out) and WT mice by Western blotting assay, using an antibody of the present disclosure as the primary antibody.
  • the upper panel is the image of Western blots, and the lower panel is bar graph chared based on values calculated from the Western blot image, wherein **represents that P ⁇ 0.01, and that the difference is significant. t-test was used for statistical tests.
  • Figure 5 Detection of MG53 content in serum of TG mice (MG53 gene overexpressed) and WT mice by Western blotting assay, using an antibody of the present disclosure as the primary antibody.
  • the upper panel is the image of Western blots, and the lower panel is bar graph chared based on values calculated from the Western blot image, wherein **represents that P ⁇ 0.01, and that the difference is significant. t-test was used for statistical tests.
  • Figure 6 Detection of recombinant mouse MG53 and recombinant human MG53 by Western blotting assay, using an antibody of the present disclosure as the primary antibody.
  • Figure 7 Detection of MG53 content in serum of Type II diabetes patients and normal people by Western blotting assay, using an antibody of the present disclosure as the primary antibody.
  • the upper panel is the image of Western blots
  • the lower panel is bar graph chared based on values calculated from the Western blot image, wherein **represents that P ⁇ 0.01, and that the difference is significant. t-test was used for statistical tests.
  • Figure 8 Detection of MG53 content in serum of mice having high-fat diet and mice having normal diet by Western blotting assay, using an antibody of the present disclosure as the primary antibody.
  • the upper panel is the image of Western blots
  • the lower panel is bar graph chared based on values calculated from the Western blot image, wherein **represents that P ⁇ 0.01, and that the difference is significant.
  • t-test was used for statistical tests.
  • Figure 9 a. Content of serum MG53 of fasted normal rats and fasted ZDF rats; b. Chart showing correlation between concentration of serum MG53 and body weight in fasted normal rats and fasted ZDF rats; c. Chart showing correlation between concentration of serum MG53 and blood glucose level in fasted normal rats and fasted ZDF rats; d. Chart showing correlation between concentration of serum MG53 and serum insulin level in fasted normal rats and fasted ZDF rats. Wherein, **represents that P ⁇ 0.01, and that the difference is significant. t-test was used for statistical tests.
  • FIG. 10 Concentration of GAPDH (control) , Akt, and Akt post insulin-induced phosphorylation, detected in different tissues (skeletal muscle, liver, visceral fat, and the heart) in mice injected or not injected with insulin, and followed by injection of BSA or rhMG53.
  • the upper panels are images of Western blots, and the lower panels are bar graphs chared based on values calculated from the Western blot images, wherein **represents that P ⁇ 0.01, and that the difference is significant.
  • One-way anova was used for statistical tests.
  • Figure 11 a. Content of serum MG53 in normal mice and diabetic model mice; b. Blood glucose levels in the control group (administered with IgG) and the treatment group (administered with MG53 antibodies) ; c. Blood glucose levels in the control group (administered with IgG) and the treatment group (administered with MG53 antibodies) in insulin tolerance test.
  • t-test was used for statistical tests.
  • FIG. 12 IRS1 plasmid (IRS1-GFP) and MG53 plasmid (MG53-myc) with Myc label were co-transfected in HEK293T cells and HEK293A cells respectively, and fluorescence intensity of GFP was detected, wherein **represents that P ⁇ 0.01, and that the difference is significant; *represents that P ⁇ 0.05, and that the difference is significant. t-test was used for statistical tests.
  • Figure 14 a. Response value for compound No. 1 in the SPR assay; b. Response value for compound No. 10 in the SPR assay; c. Response value for compound No. 26 in the SPR assay; d. Response value for compound No. 16 in the SPR assay.
  • FIG. 15 IRS1 plasmid (IRS1-GFP) and MG53 plasmid (MG53-myc) with Myc label were co-transfected in HEK293T, and compounds Nos. 1, 10, 26 and 16 were added respectively, and then fluorescence intensity of GFP was detected. ***represents that P ⁇ 0.001, and that the difference is significant. One-way anova was used for statistical tests.
  • Figure 16 This figure illustrates the amino acid sequence SEQ ID NO: 3 of heavy chain of Antibody #6 and its encoding nucleic acid sequence SEQ ID NO: 7. The sequences marked by shadow corresponds to the variable region of heavey chain of Antibody #6.
  • Figure 17 This figure illustrates the amino acid sequence SEQ ID NO: 4 of light chain of Antibody #6 and its encoding nucleic acid sequence SEQ ID NO: 8. The sequences marked by shadow corresponds to the variable region of light chain of Antibody #6.
  • Figure 18 This figure illustrates the amino acid sequence SEQ ID NO: 11 of heavy chain of Antibody #110 and its encoding nucleic acid sequence SEQ ID NO: 15. The sequences marked by shadow corresponds to the variable region of heavey chain of Antibody #110.
  • Figure 19 This figure illustrates the amino acid sequence SEQ ID NO: 12 of light chain of Antibody #110 and its encoding nucleic acid sequence SEQ ID NO: 16. The sequences marked by shadow corresponds to the variable region of light chain of Antibody #110.
  • Figure 20 This figure illustrates the amino acid sequence SEQ ID NO: 19 of heavy chain of Antibody #84 and its encoding nucleic acid sequence SEQ ID NO: 23. The sequences marked by shadow corresponds to the variable region of heavey chain of Antibody #84.
  • Figure 21 This figure illustrates the amino acid sequence SEQ ID NO: 20 of light chain of Antibody #84 and its encoding nucleic acid sequence SEQ ID NO: 24. The sequences marked by shadow corresponds to the variable region of light chain of Antibody #84.
  • Figure 22 This figure illustrates the amino acid sequence SEQ ID NO: 27 of heavy chain of Antibody #9 and its encoding nucleic acid sequence SEQ ID NO: 31. The sequences marked by shadow corresponds to the variable region of heavey chain of Antibody #9.
  • Figure 23 This figure illustrates the amino acid sequence SEQ ID NO: 28 of light chain of Antibody #9 and its encoding nucleic acid sequence SEQ ID NO: 32. The sequences marked by shadow corresponds to the variable region of light chain of Antibody #9.
  • Figure 24 This figure illustrates the amino acid sequence SEQ ID NO: 35 of heavy chain of Antibody #43 and its encoding nucleic acid sequence SEQ ID NO: 39. The sequences marked by shadow corresponds to the variable region of heavey chain of Antibody #43.
  • Figure 25 This figure illustrates the amino acid sequence SEQ ID NO: 36 of light chain of Antibody #43 and its encoding nucleic acid sequence SEQ ID NO: 40. The sequences marked by shadow corresponds to the variable region of light chain of Antibody #43.
  • Figure 26 This figure illustrates the amino acid sequence SEQ ID NO: 41 of human wild-type MG53, the amino acid sequence SEQ ID NO: 42 of monkey wild-type MG53, the amino acid sequence SEQ ID NO: 43 of rat wild-type MG53, and the amino acid sequence SEQ ID NO: 44 of mouse wild-type MG53.
  • Figure 27 This figure illustrates the amino acid sequence SEQ ID NO: 45 of swine wild-type MG53, the amino acid sequence SEQ ID NO: 46 of dog wild-type MG53, the amino acid sequence SEQ ID NO: 47 of rabbit wild-type MG53, the amino acid sequence SEQ ID NO: 47 of Marmosets wild-type MG53, and the amino acid sequence SEQ ID NO: 61 of Callithrix jacchus wild-type MG53.
  • Figure 28 This figure illustrates the sequences of MG53 mutant (SEQ ID NO: 48-50) .
  • Figure 29 This figure illustrates the sequences of MG53 mutant (SEQ ID NO: 51-53) .
  • Figure 30 This figure illustrates the sequences of MG53 mutant (SEQ ID NO: 54-57) .
  • Figure 31 This figure illustrates the sequence of insulin receptor (SEQ ID NO: 58) .
  • Figure 32 This figure illustrates high glucose-and insulin-induced MG53 release in Langendorff-perfused rodent hearts.
  • data are normalized to the corresponding non-specific bands (NS) which were obtained through Brilliant Green staining, and are presented as mean ⁇ s.e.m. (*P ⁇ 0.05, **P ⁇ 0.01) ; one-way ANOVA for A-E, G and H and t-test for F.
  • the control of each bar graph is the 1st lane of each gel.
  • Figure 33 This figure illustrates high glucose-and insulin-induced MG53 secretion in isolated skeletal muscle and in vivo.
  • Figure 34 This figure illustrates cardiac-specific overexpression of MG53 leads to systemic insulin resistance and metabolic syndrome.
  • Figure 35 This figure illustrates high glucose-and insulin-induced release of MG53 from striated muscle.
  • Figure 35A shows creatine kinase (CK, n-8) from the perfusate of mouse heart with or without high glucose (25 mM) and insulin (10 U/L) stimulation for 30 min.
  • Data are mean ⁇ s.e.m.
  • the present disclosure relates to a method for detecting an MG53 related disease or predicting a risk of an MG53 related disease, comprising the following steps: a) obtaining a test sample; and b) detecting cell-free MG53 in the test sample.
  • MG53 refers to naturally existing wild-type MG53 expressed in animals and an MG53 mutant.
  • MG53 protein exists in many animals, including but not limited to human, ape, monkey, swine, dog, rabbit, rat, mouse, and other mammals.
  • MG53 protein is a multi-functional protein with a structure shown in Figure 1.
  • the full-length MG53 proteins of different species are slightly different in length, but generally has about 477 amino acids, comprising a TRIM motif at the N-terminus and a SPRY motif at the C-terminus, with the TRIM motif consisting of successively linked Ring, B-box and coiled-coil domains (RBCC) .
  • TRIM motif consisting of successively linked Ring, B-box and coiled-coil domains (RBCC) .
  • RBCC coiled-coil domains
  • MG53 protein may also cause insulin resistance and metabolic syndrome.
  • the structure and function of MG53, as well as its interaction with other proteins have been reported in detail in the art (see, e.g., Chuanxi Cai et al., Journal of Biological Chemistry, 284 (5) , 3314-3322, (2009) ; Xianhua Wang et al., Circulation Research 107, 76-83, (2010) ; Eun Young Park et al., Proteins, 790-795 (2009) ) .
  • the amino acid sequence of the wild-type MG53 is set forth in SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47 or SEQ ID NO: 61, which corresponds to the human, Macaca mulatta /M. fascicularis, rat, mouse, swine, dog, rabbit, and Callithrix jacchus wild-type MG53, respectively.
  • the term “MG53” as used herein refers to MG53 derived from the subject to be detected or treated.
  • test sample refers to a sample obtained from a subject in vivo, or obtained from a subject in vitro, or from an in vitro sample, including any material from a subject which a person skilled in the art considers to contain, or which may contain, cell-free MG53.
  • the test sample is a blood sample.
  • the test sample is body fluid.
  • the test sample is selected from whole blood, plasma, serum, tissue fluid, urine, or sweat.
  • the test sample is selected from whole blood, plasma, or serum.
  • the test sample may further comprise other substances, such as preservatives, anticoagulants, buffers, fixatives, nutrients and antibiotics, etc.
  • the test sample substantially does not contain any cells, e.g., does not contain any cardiac muscle cell, striated muscle cell, or blood cell.
  • the term “substantially does not contain” refers to not containing a particular substance in the amount which is more than a particular amount, or more than the remaining amount after conducting common impurity removal technology of the art, or any particular substance substantially affecting functions of other substances.
  • that the sample substantially does not contain cells may refer to that the sample does not contain cells in excess of a particular amount (e.g.
  • the sample may be obtained using any method known in the art.
  • a blood sample may be obtained using vacuum or normal pressure blood collection needle.
  • the collected blood sample may be further treated, e.g., centrifuged.
  • the MG53 detection agent is an MG53 antibody or antigen-binding fragment thereof, or an MG53 ligand or an MG53-binding fragment thereof. Any MG53 antibody or antigen-binding fragment thereof, or MG53 ligand or MG53-binding fragment thereof described by the present disclosure, or a combination thereof, can be used as an MG53 detection agent.
  • the MG53 detection agent is a small molecule compound capable of binding MG53. Any small molecule compound capable of binding MG53 described by the present disclosure, or a combination thereof, can be used as an MG53 detection agent.
  • the MG53 detection agent further has a detectable label.
  • the detectable label is luminescent, magnetic, radioactive, or enzymatically active. Examples for detectable labels include fluorescence labels, enzyme labels, radioisotope labels, chemiluminescence labels, electrochemiluminescence labels, metal particle labels and hapten labels.
  • fluorescence labels examples include 5- (6) -carboxy fluorescein, 5-or 6-carboxy fluorescein, 6- (fluorescein) -5- (6) -carboxyamide hexanoic acid, isothiocyanate fluorescein, rhodamine, tetramethyl rhodamine, dye (such as Cy2, Cy3 and Cy5) , optionally substituted coumarin (e.g., AMCA, PerCP) , phycobiliprotein (e.g., R-phycoerythrin (RPE) and allophycocyanin (APC) ) , Texas Red, Princeton Red, green fluorescent protein (GFP) and analogs thereof, R-phycoerythrin and allophycocyanin conjugate and inorganic fluorescence label (such as semiconductor material based particles, e.g., particles coated with CdSe nanocrystalline) .
  • fluorescence labels include 5- (6) -carboxy fluorescein, 5-or 6-carbox
  • enzyme labels examples include horseradish peroxidase (HRP) , alkaline phosphatase (ALP or AP) , ⁇ -galactosidase (GAL) , glucose-6-phsphate dehydrogenase, ⁇ -N-acetyl glucosaminidase, ⁇ -glucuronidase, saccharase, xanthine oxidase, firefly luciferase, and glucose oxidase (GO) .
  • HRP horseradish peroxidase
  • ALP or AP alkaline phosphatase
  • GAL ⁇ -galactosidase
  • glucose-6-phsphate dehydrogenase ⁇ -N-acetyl glucosaminidase
  • ⁇ -glucuronidase ⁇ -glucuronidase
  • saccharase xanthine oxidase
  • firefly luciferase firefly
  • Examples of common substrates for horseradish peroxidase include 3, 3'-diaminobenzidine (DAB) , nickel intensified diaminobenzidine, 3-amino-9-ethyl carbazole (AEC) , benzidine dihydrochloride (BDHC) , Hanker-Yates reagent (HYR) , indophenol blue (IB) , tetramethyl benzidine (TMB) , 4-chloro-1-naphthol (CN) , ⁇ -naphthol pyronine ( ⁇ -NP) , o-dianisidine (OD) , 5-bromo-4-chloro-3-indole phosphate (BCIP) , nitroblue tetrazolium (NBT) , 2- (iodophenyl) -3-p-nitrophenyl-l-5-phenyl tetrazole chloride (INT) , tetranitroblue tetrazolium
  • Examples of common substrates for alkaline phosphatase include naphthol-AS-B 1-phosphate/fast red TR (NABP/FR) , naphthol-AS-MX-phosphate/fast red TR (NAMP/FR) , naphthol-AS-B1-phosphate/fast red TR (NABP/FR) , naphthol-AS-MX-phosphate/fast red FR (NAMP/FR) , naphthol-AS-B1-phosphat/new fuschin (NAMP/NF) , bromochloro indole phosphate/nitroblue tetrazolium (BCIP/NBT) and 5-bromo-4-chloro-3-indolyl-b-d-pyran galactosidase (BCIG) .
  • NAMP/FR naphthol-AS-B 1-phosphate/fast red TR
  • NAMP/FR naphthol-AS-MX-phosphate/fast red TR
  • NAMP/FR naphthol
  • radioisotope labels examples include isotopes of iodine, cobalt, selenium, tritium, carbon, sulfur and phosphorus.
  • chemiluminescence labels include luminol, isoluminol, acridinium ester, 1, 2-dioxolane, and pyridopyridazine.
  • electrochemiluminescence labels examples include ruthenium derivatives.
  • metal particle labels include gold particles and gold coating particles, which can be transformed by silver staining.
  • Example of hapten labels include DNP, fluorescein isothiocyanate (FITC) , biotin and digoxin.
  • FITC fluorescein isothiocyanate
  • exemplary detectable markers have one or more of the following structures.
  • detectable markers can be linked to the MG53 detection agent by means of covalent binding, affinity binding, embedding, complexing, binding, mixing or adding, etc.
  • Step b) detecting cell-free MG53 in the test sample can be conducted using conventional methods of the art.
  • these methods include but are not limited to radioimmunoassay, Western blot analysis, proximity ligation assay, immunofluorescence assay, enzyme immunoassay, immunoprecipitation, chemiluminescence, immunohistochemistry assay, dot blot assay or slit blot method.
  • a person skilled in the art is familiar with ordinary technologies and other variants of the technologies in the conduction of the various of detecting methods above, and is also capable of using a method above alone or in combination or alternatively with nuclear magnetic resonance (NMR) , matrix assisted laser desorption ionization-time of flight (MALDI-TOF) , liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) , etc.
  • NMR nuclear magnetic resonance
  • MALDI-TOF matrix assisted laser desorption ionization-time of flight
  • LC-MS/MS liquid chromatography-mass spectrometry/mass spectrometry
  • enzyme immunoassay is a sandwich enzymoimmunoassay, which detects cell-free MG53 using antibodies that specifically bind to MG53.
  • a typical sandwich enzymoimmunoassay includes the following steps: a) linking an antibodies that specifically bind to cell-free MG53 to solid phase supports, to form solid phase antibodies, and removing non-bound antibodies and impurities by washing; b) adding a test specimen, which is contacted with the solid phase antibodies for certain time to form solid phase antibody-antigen complexes, and removing other non-bound substances by washing; c) adding enzyme-labeled antibodies which can bind to cell-free MG53, which is contacted with the solid phase antibody-antigen complexes for certain time sufficient for binding to the solid phase antibody-antigen complexes, and removing non-bound enzyme-labeled antibodies by washing; d) adding the substrate for the enzyme label, which is catalyzed by the enzyme label under proper condition, and determining the amount of cell-free MG
  • the detecting method of the present disclosure may further comprise a step of data processing.
  • the method of the present disclosure further comprises: c) comparing the detected value of MG53 obtained from step b) with a reference value.
  • the reference value comes from a reference sample.
  • the reference sample and the test sample are from different subjects.
  • the reference sample may come from a healthy subject or a subject suffering from an MG53 related disease, while the test sample comes from a subject suffering from or suspected to suffer from an MG53 related disease.
  • the reference sample and the test sample are from the same subject, but the reference sample is collected at an earlier or later time period than the test sample. In certain embodiments, during the interval between the collection of the reference sample and test sample, the subject accepts treatment or the health condition thereof has changed. In some other embodiments, the reference sample and the test sample are collected from different parts of the same subject.
  • the reference value is obtained from a healthy subject or a subject confirmed to suffer from an MG53 related disease. In certain embodiments, the reference value is characterized in the form of concentration of MG53.
  • these reference values can be negative reference values, being equal to or lower than which indicates that the subject which the test sample derives from is not suffering from MG53 related diseases or is at low risk of suffering from an MG53 related disease.
  • the negative reference value can be set according to the mean value of MG53 detection values obtained from certain number of healthy subjects, e.g., setting such mean value or 80%, 90%, 110%, 120%, 150%or 200%of such mean value as the negative reference value.
  • these reference values can be positive reference values, being equal to or higher than which indicates that the subject which the test sample derives from is suffering from an MG53 related disease or is at high risk of suffering from an MG53 related disease.
  • the positive reference value can be set according to the mean value of MG53 detection values obtained from certain number of subjects confirmed to suffer from MG53 related diseases, e.g., setting such mean value or 80%, 90%, 110%, 120%, 150%or 200%of such mean value as the positive reference value.
  • subject refers to human and non-human animal.
  • Non-human animals include all vertebrates, e.g., mammals and non-mammals.
  • a “subject” may also be a livestock animal (e.g., cow, swine, goat, chicken, rabbit or horse) , or a rodent (e.g., rat or mouse) , or a primate (e.g., gorilla or monkey) , or a domestic animal (e.g., dog or cat) .
  • a “subject” may be a male or a female, and also may be at different ages.
  • a human “subject” may be Caucasian, African, Asian, Sumerian, or other races, or a hybrid of different races.
  • a human “subject” may be elderly, adult, teenager, child or infant.
  • the subject is a human or non-human mammal, e.g., mouse, rat, rabbit, goat, sheep, guinea swine or hamster.
  • MG53 related disease refers to such disease whereby the generation, onset, and development, etc., of which is related to activity of MG53.
  • a person skilled in the art can determine whether a disease is related to MG53 through ordinary technical means. For example, whether the disease is related to MG53 can be determined by overexpressing MG53 protein, knockouting MG53 or reducing the content of MG53 in a subject through e.g., antisense nucleic acid technology, CRISPR/Cas9 technology and Trim21-mediated proteasome degradation and then assessing the condition of the disease in the subject.
  • the MG53 related disease is metabolic syndrome. In certain embodiments, the MG53 related disease is a glucose metabolism or lipid metabolism related disease. In certain embodiments, the glucose metabolism or lipid metabolism related disease is selected from the group consisting of insulin resistance, diabetic cerebrovascular disease, diabetic ocular complication, diabetic neuropathy, diabetic foot, hyperinsulinemia, hypercholesterolemia, hyperglycemia, hyperlipidemia, hypertension and obesity.
  • metabolic syndrome refers to disorders characterized by aggregation of lipid and non-lipid cardiovascular risk factors caused by metabolic disturbance.
  • metabolic syndrome is identified by the presence of any three of the following risk factors: waist circumference of over 102 cm in a male or over 88 cm in a female; serum triglyceride of at least 150 mg/dL; high density lipoprotein-cholesterol of less than 40 mg/dL in a male or less than 50 mg/dL in a female; blood pressure of at least 130/85 mmHg; and fasting glucose of at least 110 mg/dL.
  • NCEP National Cholesterol Education Program
  • the method for detecting an MG53 related disease or predicting a risk of an MG53 related disease of the present disclosure can be used for many purposes.
  • the detecting method can be used to detect whether a subject suffers from an MG53 related disease, or to predict the risk at which a subject suffers from an MG53 related disease. For example, as described above, that the content of cell-free MG53 in a subject is equal to or lower than the negative reference value indicates that the subject is not suffering from MG53 related diseases or is at low risk of suffering from an MG53 related disease; that the content of cell-free MG53 in a subject is equal to or higher than the positive reference value indicates that the subject is suffering from an MG53 related disease or is at high risk of suffering from an MG53 related disease.
  • the detecting method can be used to predict the development of an MG53 related disease in a subject. For example, to predict the condition of an MG53 related disease (e.g., improvement or deterioration) in a subject by determining the change in content and/or activity of cell-free MG53 in a subject, compared with the value of the same subject a period of time ago.
  • an MG53 related disease e.g., improvement or deterioration
  • reduced content and/or activity of cell-free MG53 in a subject compared with the value of the same subject a period of time ago indicates improvement of an MG53 related disease in the subject; increased content and/or activity of cell-free MG53 in a subject compared with the value of the same subject a period of time ago, indicates deterioration of an MG53 related disease in the subject.
  • the detecting method can be used to identify potential cell-free MG53 inhibitors.
  • the use usually includes contacting a test compound or reagent with cell-free MG53, followed by detecting cell-free MG53.
  • a test compound or reagent capable of reducing the content of cell-free MG53 by at least 10% can be considered as a cell-free MG53 inhibitor.
  • the content of cell-free MG53 can be reduced by at least 20%, 40%, 50%, 80%, 90%, 95%or more.
  • Test compounds or reagents include, e.g., small molecule organic and inorganic compounds (e.g., molecules obtained from artificially synthetic chemical libraries and natural product libraries) , antibodies or antigen-binding fragments thereof, ligands or binding fragments thereof, or nonfunctional protein fragments of MG53.
  • small molecule organic and inorganic compounds e.g., molecules obtained from artificially synthetic chemical libraries and natural product libraries
  • antibodies or antigen-binding fragments thereof e.g., antibodies or antigen-binding fragments thereof, ligands or binding fragments thereof, or nonfunctional protein fragments of MG53.
  • the detecting method can be used to determine whether a subject responds to a cell-free MG53 inhibitor.
  • the presence and level of cell-free MG53 in the test sample can indicate whether the subject which the test sample derives from responds to the cell-free MG53 inhibitor.
  • the content and/or activity of cell-free MG53 in the test sample is reduced by at least 10%indicates that the subject which the test sample derives from may respond to a cell-free MG53 inhibitor.
  • the content of cell-free MG53 in the test sample can be reduced by at least 20%, 40%, 50%, 80%, 90%, 95%or more.
  • the detecting method can be used to assess therapeutic effect for an MG53 related disease.
  • the reference sample and the test sample are from the same subject, but the time at which the reference sample was collected is earlier for a period of time than the test sample, and the subject accepts treatment during the period of time.
  • that the content of cell-free MG53 in the test sample is reduced by at least 10%compared to the reference sample indicates that the subject responds to the treatment currently being accepted.
  • the content and/or activity of cell-free MG53 in the test sample can be reduced by at least 20%, 40%, 50%, 80%, 90%, 95%or more. According to the result of the assessment on response, the treatment strategy for the subject can be sustained or altered.
  • the detecting method can be used to assess the change of content of cell-free MG53 or its mutants, after the treatment using MG53 or its mutatnts.
  • the reference sample and the test sample are from the same subject, but the time at which the reference sample was collected is earlier for a period of time than the test sample, and the subject accepts the treatment using MG53 or its mutatnts during the period of time, thereby the content and/or activity of cell-free MG53 or its mutants was changed.
  • the treatment strategy for the subject can be sustained or altered.
  • the present disclosure relates to a method for reducing or inhibiting activity of cell-free MG53 in a subject, which comprises administrating an effective amount of MG53 inhibitor to a subject in need thereof.
  • MG53 inhibitors can be small molecule organic or inorganic compounds (e.g., molecules obtained from artificially synthetic chemical libraries and natural product libraries) , antibodies or antigen-binding fragments thereof, ligands or MG53 binding fragments thereof, or nonfunctional protein fragments of MG53.
  • the MG53 inhibitor can reduce the activity of cell-free MG53 by at least 5%, 10%, 20%, 40%, 50%, 80%, 90%, 95%or more.
  • activity when used together with increase or reduce, refers to the detected functional activity, which may be represented by a change in the content of cell-free MG53 or, in the absence of a change in the content of cell-free MG53, a change in the functional activity of cell-free MG53.
  • activity of the MG53 is related to metabolic syndrome.
  • activity of the MG53 is related to glucose metabolism or lipid metabolism.
  • activity of the MG53 is related to insulin resistance, hyperinsulinemia, hypercholesterolemia, hyperglycemia, hyperlipidemia, hypertension or obesity.
  • activity of the MG53 refers to insulin receptor and insulin receptor substrate binding activity.
  • the MG53 inhibitor specifically binds to MG53.
  • the term “specific binding” or “specifically binds” as used herein, with reference to MG53 inhibitor refers to that the MG53 inhibitor preferentially identifies MG53 in a complicated mixture, and the binding constant of the inhibitor to MG53 is at least 2 fold of that of the inihibitor to other non-specific binding proteins.
  • the equilibrium dissociation constant of the MG53 inhibitor to MG53 is less than or equal to 10 -6 or 10 -7 M. In certain embodiments, the equilibrium dissociation constant of the MG53 inhibitor to MG53 is less than or equal to 10 -7 M or 10 -8 M.
  • the MG53 inhibitor is an MG53 antibody or antigen-binding fragment thereof, an MG53 ligand or MG53 binding fragment thereof, or a nonfunctional protein fragment of MG53.
  • Antibodies or antigen-binding fragments thereof, ligands or MG53 binding fragments thereof, or nonfunctional protein fragments of MG53 that can be used in the present disclosure include but are not limited to any MG53 antibody or antigen-binding fragment thereof, MG53 ligand or MG53 binding fragment thereof, or nonfunctional protein fragment of MG53 described herein, or a combination thereof.
  • the MG53 inhibitor is a small molecule compound capable of binding to MG53.
  • Small molecule compounds binding to MG53 that can be used in the present disclosure include but are not limited to any small molecule compound binding to MG53 described herein, or a combination thereof.
  • the reducing or inhibiting method described herein can be used for the amelioration, treatment or prevention of an MG53 related disease or a symptom thereof.
  • the reducing or inhibiting method includes administrating a therapeutically effective amount of MG53 inhibitor to a subject. See preceding texts for definition of MG53 related disease.
  • Treatment includes preventing or alleviating a disease or discomfort, slowing the onset or rate of development of a disease or discomfort, reducing the risk of developing a disease or discomfort, preventing or delaying the development of symptoms associated with a disease or discomfort, reducing or ending symptoms associated with a disease or discomfort, generating a complete or partial regression of a disease or discomfort, curing a disease or discomfort, or some combination thereof.
  • therapeutically effective amount refers to the amount of a drug capable of ameliorating or eliminating a disease or symptom of a subject, or of preventively inhibiting or preventing the occurrence of the disease or symptom.
  • a therapeutically effective amount can be the amount of a drug that ameliorates one or more diseases or symptoms of a subject to certain extent; the amount of a drug capable of restoring one or more physiological or biochemical parameters associated with the cause of a disease or symptom, partly or completely back to normal; and/or the amount of a drug capable of reducing the possibility that a disease or symptom occurs.
  • the therapeutically effective dosage of the MG53 inhibitor provided herein is dependent on various factors known in the art, for example, body weight, age, pre-existing medical condition, therapy currently being received, health condition of the subject, and intensity, allergic, superallergic and side effect of drug interaction, and route of administration and the extent to which the disease develops.
  • a skilled artisan e.g., a physician or veterinarian may reduce or increase dosage in accordance with these or other conditions or requirement.
  • the MG53 inhibitor provided herein may be administered at a therapeutically effective dosage of about 0.01 mg/kg to about 100 mg/kg (e.g., about 0.01 mg/kg, about 0.5 mg/kg, about 1 mg/kg, about 2 mg/kg, about 5 mg/kg, about 10 mg/kg, about 15 mg/kg, about 20 mg/kg, about 25 mg/kg, about 30 mg/kg, about 35 mg/kg, about 40 mg/kg, about 45 mg/kg, about 50 mg/kg, about 55 mg/kg, about 60 mg/kg, about 65 mg/kg, about 70 mg/kg, about 75 mg/kg, about 80 mg/kg, about 85 mg/kg, about 90 mg/kg, about 95 mg/kg, or about 100 mg/kg) .
  • the MG53 inhibitor is administered at a dosage of about 50 mg/kg or less.
  • the dosage is 10 mg/kg or less, 5 mg/kg or less, 1 mg/kg or less, 0.5 mg/kg or less, or 0.1 mg/kg or less.
  • a particular dosage can be divided and administered multiple times separated by interval, e.g., once every day, twice or more every day, twice or more every month, once every week, once every two weeks, once every three weeks, once a month or once every two months or more.
  • the administered dosage may vary over the course of treatment.
  • the initially administered dosage can be higher than subsequently administered dosages.
  • the administered dosages are adjusted in the course of treatment depending on the response of the administration subject.
  • Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic response) .
  • a single dose may be administered, or several divided doses may be administered over time.
  • the MG53 inhibitor disclosed herein may be administered by any route known in the art, such as parenteral (e.g., subcutaneous, intraperitoneal, intravenous, including intravenous infusion, intramuscular, or intradermal injection) or non-parenteral (e.g., oral, intranasal, sublingual, rectal, or topical) routes.
  • parenteral e.g., subcutaneous, intraperitoneal, intravenous, including intravenous infusion, intramuscular, or intradermal injection
  • non-parenteral e.g., oral, intranasal, sublingual, rectal, or topical
  • the MG53 inhibitor disclosed herein may be administered in the form of a pharmaceutical composition of the present disclosure.
  • the MG53 inhibitor disclosed herein can be combined with other agents or therapies to treat a related disease or disorder.
  • the MG53 inhibitor is administered simultaneously or sequentially with other agents or therapies.
  • the other agents or therapies are one or more agents or therapies used for treating insulin resistance, diabetic cerebrovascular disease, diabetic ocular complication, diabetic neuropathy, diabetic foot, hyperinsulinemia, hypercholesterolemia, hyperglycemia, hyperlipidemia, hypertension and obesity.
  • the other agents are hypoglycemic agents, e.g., PPAR agonists, dipeptidyl peptidase (IV) inhibitors, GLP-1 analogs, insulins or insulin analogs, insulin sercretogogues, SGLT2 inhibitors, human dextrin analogs, biguanide, ⁇ -glocusidase inhibitors or a combination thereof.
  • hypoglycemic agents e.g., PPAR agonists, dipeptidyl peptidase (IV) inhibitors, GLP-1 analogs, insulins or insulin analogs, insulin sercretogogues, SGLT2 inhibitors, human dextrin analogs, biguanide, ⁇ -glocusidase inhibitors or a combination thereof.
  • Another aspect of the present disclosure further provides an MG53 antibody and antigen-binding fragment thereof.
  • the MG53 antibody and antigen-binding fragment thereof comprises three heavy chain complementary determining regions comprised in a sequence of SEQ ID NOs: 1, 9, 17, 25 or 33. In certain embodiments, the MG53 antibody and antigen-binding fragment thereof further comprises three light chain complementary determining regions comprised in a sequence of SEQ ID NOs: 2, 10, 18, 26 or 34. In certain embodiments, the MG53 antibody and antigen-binding fragment thereof comprises three heavy chain complementary determining regions and three light chain complementary determining regions, the six complementary determining regions comprised in a sequence of SEQ ID NOs: 1/2, 9/10, 17/18, 25/26 or 33/34.
  • the MG53 antibody and antigen-binding fragment thereof comprises heavy chain, the variable regions of the heavy chain of the MG53 antibody and antigen-binding fragment thereof have an amino acid sequence of SEQ ID NOs: 1, 9, 17, 25 or 33, or a sequence having at least 80%, 85%, 90%, 95%or 99%sequence homology thereof.
  • the heavy chain of the MG53 antibody and antigen-binding fragment thereof has an amino acid sequence of SEQ ID NOs: 3, 11, 19, 27 or 35, or a sequence having at least 80%, 85%, 90%, 95%or 99%sequence homology thereof.
  • the MG53 antibody and antigen-binding fragment thereof further comprises a light chain, the variable regions of the light chain of the MG53 antibody and antigen-binding fragment thereof have an amino acid sequence of SEQ ID NOs: 2, 10, 18, 26 or 34, or a sequence having at least 80%, 85%, 90%, 95%or 99%sequence homology thereof.
  • the light chain of the MG53 antibody and antigen-binding fragment thereof has an amino acid sequence of SEQ ID NOs: 4, 12, 20, 28 or 36, or a sequence having at least 80%, 85%, 90%, 95%or 99%sequence homology thereof.
  • a competitive antibody or antigen-binding fragment thereof is also provided, which competes for binding to MG53 with the antibody or antigen-binding fragment above.
  • Compet for binding refers to the ability of an antibody or antigen-binding fragment thereof to specifically inhibit the binding interaction between the antigen it is against and another antibody molecule (e.g. MG53 and another MG53 antibody) .
  • a competitive antibody or antigen-binding fragment that blocks binding between two antigen-antibody molecules inhibits the binding interaction between the two antigen-antibody molecules by at least 50%. In certain embodiments, this inhibition may be greater than 60%, greater than 70%, greater than 80%, or greater than 90%.
  • antibody as used herein includes any immunoglobulin, monoclonal antibody, polyclonal antibody, multispecific antibody, or bispecific (bivalent) antibody that binds to a specific antigen.
  • a native intact antibody comprises two heavy chains and two light chains. Each heavy chain consists of a variable region and a first, second, and third constant region, while each light chain consists of a variable region and a constant region. Mammalian heavy chains are classified as ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , and mammalian light chains are classified as ⁇ or ⁇ .
  • the antibody has a “Y” shape, with the stem of the Y consisting of the second and third constant regions of two heavy chains bound together via disulfide bonding.
  • Each arm of the Y includes the variable region and first constant region of a single heavy chain bound to the variable and constant regions of a single light chain, wherein the first constant region of the heavy chain is linked to the second constant region via a hinge region.
  • the variable regions of the light and heavy chains are responsible for antigen binding specificity.
  • the variable regions in both chains generally contain three highly variable loops called the complementarity determining regions (CDRs) (light (L) chain CDRs including LCDR1, LCDR2, and LCDR3, heavy (H) chain CDRs including HCDR1, HCDR2, and HCDR3) .
  • CDRs complementarity determining regions
  • CDR boundaries for the antibodies and antigen-binding fragments disclosed herein may be defined or identified by the conventions of Kabat, Chothia, or Al-Lazikani (see Al-Lazikani, B., Chothia, C., Lesk, A.M., J. Mol. Biol., 273 (4) , 927 (1997) ; Chothia, C. et al., J Mol Biol. Dec 5; 186 (3) : 651-63 (1985) ; Chothia, C. and Lesk, A.M., J. Mol. Biol., 196, 901 (1987) ; Chothia, C. et al., Nature.
  • the five major classes or isotypes of antibodies are IgA, IgD, IgE, IgG, and IgM, which are characterized by the presence of ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ heavy chains, respectively.
  • IgG1 ( ⁇ 1 heavy chain) IgG2 ( ⁇ 2 heavy chain)
  • IgG3 ( ⁇ 3 heavy chain) IgG4 ( ⁇ 4 heavy chain)
  • IgA1 ( ⁇ 1 heavy chain) ⁇ 2 heavy chain
  • IgA2 ( ⁇ 2 heavy chain) Several of the major antibody classes are divided into subclasses such as IgG1 ( ⁇ 1 heavy chain) , IgG2 ( ⁇ 2 heavy chain) , IgG3 ( ⁇ 3 heavy chain) , IgG4 ( ⁇ 4 heavy chain) , IgA1 ( ⁇ 1 heavy chain) , or IgA2 ( ⁇ 2 heavy chain) .
  • antigen-binding fragment refers to an antibody fragment formed from a portion of an antibody comprising one or more CDRs, but does not comprise an intact antibody structure.
  • antigen-binding fragment include, without limitation, an Fab, an Fab', an F (ab') 2 , an Fv fragment, a single-chain antibody molecule (scFv) , an scFv dimer, a camelized single domain antibody, and a nanobody.
  • An antigen-binding fragment is capable of binding to the same antigen to which the parent antibody binds.
  • Fab with regard to an antibody refers to that portion of the antibody consisting of a single light chain (both variable and constant regions) bound to the variable region and first constant region of a single heavy chain by a disulfide bond.
  • Fab' refers to a Fab fragment that includes a portion of the hinge region.
  • F (ab') 2 refers to a dimer of Fab'.
  • An “Fv” fragment consists of the variable region of a single light chain and the variable region of a single heavy chain.
  • Single-chain Fv antibody or “scFv” refers to an engineered antibody consisting of a light chain variable region and a heavy chain variable region connected to one another directly or via a peptide linker sequence (see e.g., Huston JS et al., Proc Natl Acad Sci USA, 85: 5879 (1988) for specific introduction) .
  • scFv dimer refers to a polymer formed by two scFvs.
  • “Camelized single domain antibody” also known as “heavy chain antibody” or “HCAb” (heavy-chain-only antibody) , refers to an antibody that contains two heavy chain variable regions but no light chains (Riechmann L. and Muyldermans S., J Immunol Methods. Dec 10; 231 (1-2) : 25-38 (1999) ; Muyldermans S., J Biotechnol. Jun; 74 (4) : 277-302 (2001) ; WO94/04678; WO94/25591; U.S. Patent No. 6,005,079) . Heavy chain antibodies were originally derived from Camelidae (camels, dromedaries, and llamas) .
  • camelized antibodies Although devoid of light chains, camelized antibodies have an authentic antigen-binding repertoire (see Hamers-Casterman C. et al., Nature. 363 (6428) : 446-8 (1993) ; Nguyen VK. et al., “Heavy-chain antibodies in Camelidae; a case of evolutionary innovation, ” Immunogenetics. 54 (1) : 39-47 (2002) ; Nguyen VK. et al., Immunology. 109 (1) : 93-101 (2003) , which are incorporated herein by reference in their entirety) .
  • a “nanobody” consists of a heavy chain variable region from a heavy chain antibody and two constant regions, CH2 and CH3.
  • the antibody provided herein is a fully human antibody, a humanized antibody, a chimeric antibody, a mouse antibody or rabbit antibody.
  • the antibody provided herein is a polyclonal antibody, a monoclonal antibody or a recombinant antibody.
  • the antibody provided herein is a monospecific antibody, a bispecific antibody or a multispecific antibody.
  • the antibody provided herein may further be labeled.
  • the antibody or antigen-binding fragment thereof is a fully human antibody, which is optionally produced by a transgenic rat, e.g., a transgenic rat in which the expression of endogenous rat immunoglobin gene is inactivated, and carrying recombinant human immunoglobin locus with J loci deletions and C-kappa mutations, and which can also be expressed by an engineered cell (e.g., CHO cell) .
  • a transgenic rat e.g., a transgenic rat in which the expression of endogenous rat immunoglobin gene is inactivated, and carrying recombinant human immunoglobin locus with J loci deletions and C-kappa mutations, and which can also be expressed by an engineered cell (e.g., CHO cell) .
  • humanized refers to an antibody or the antigen-binding fragment comprising CDRs derived from non-human animals, FR regions derived from human, and when applicable, constant regions derived from human.
  • a humanized antibody or antigen-binding fragment is useful as human therapeutics in certain embodiments because it has reduced immunogenicity.
  • the non-human animal is a mammal, for example, a mouse, a rat, a rabbit, a goat, a sheep, a guinea swine, or a hamster.
  • the humanized antibody or antigen-binding fragment is composed of substantially all human sequences except for the CDR sequences which are non-human.
  • chimeric refers to an antibody or antigen-binding fragment, having a portion of heavy and/or light chain derived from one species, and the rest of the heavy and/or light chain derived from a different species.
  • a chimeric antibody may comprise a constant region derived from human and a variable region from a non-human species, such as from mouse or rabbit.
  • the MG53 antibody and antigen-binding fragment thereof provided herein is capable of specifically binding to MG53 with a binding affinity of ⁇ 10 -6 M (e.g., ⁇ 5x10 -7 M, ⁇ 2x10 -7 M, ⁇ 10 -7 M, ⁇ 5x10 -8 M, ⁇ 2x10 -8 M) .
  • the binding affinity can be represented by K D value, which is calculated as the ratio of dissociation rate to association rate (k off /k on ) when the binding between the antigen and the antigen-binding molecule reaches equilibrium.
  • the antigen-binding affinity e.g. K D
  • K D can be detected using suitable methods known in the art, including plasmon resonance binding assay using instruments such as Biacore (see, for example, Murphy, M. et al., Current protocols in protein science, Chapter 19, unit 19.14, 2006) .
  • the antibody and antigen-binding fragment thereof provided herein inhibits the binding between MG53 and the ligand thereof with an IC 50 of 0.2 nM-2000 nM (e.g., 1 nM-1500 nM, 5 nM-1000 nM, 10 nM-900 nM, 20 nM-800 nM or 50 nM-800 nM) .
  • percent (%) sequence “homology” as used herein refers to, with respect to amino acid sequence, the percentage of amino acid in a candidate amino acid sequence that are identical to the amino acid in a reference amino acid sequence, calculated after aligning the sequences and, if necessary, introducing gaps, to achieve the maximum number of identical amino acids; with respect to nucleic acid sequence, the percentage of nucleotides in a candidate nucleic acid sequence that are identical to the nucleotides in a reference nucleic acid sequence, calculated after aligning the sequences and, if necessary, introducing gaps, to achieve the maximum number of identical nucleotides.
  • Alignment for purposes of determining percentage of homology can be achieved through various methods known in the art, for example, using publicly available tools such as BLASTp (available on the website of U.S. National Center for Biotechnology Information (NCBI) : http: //blast. ncbi. nlm. nih. gov/Blast. cgi, see also, Altschul S.F. et al., J. Mol. Biol., 215: 403–410 (1990) ; Stephen F. et al., Nucleic Acids Res., 25: 3389–3402 (1997) ) , ClustalW2 (available on the website of European Bioinformatics Institute: http: //www. ebi. ac.
  • NCBI National Center for Biotechnology Information
  • the MG53 antibodies or antigen-binding fragments thereof provided herein also include those resulted from conservative substitution of the amino acid residues to the MG53 antibody or antigen-binding fragment thereof provided herein.
  • amino acid residues refers to substitution between amino acids with similar properties, such as between polar amino acids (e.g., glutamine and asparagine) , between hydrophobic amino acids (e.g., leucine, isoleucine, methionine and valine) , and between similarly charged amino acids (e.g., argine, lysine, and histidine, or glutamine and aspartic acid) .
  • polar amino acids e.g., glutamine and asparagine
  • hydrophobic amino acids e.g., leucine, isoleucine, methionine and valine
  • similarly charged amino acids e.g., argine, lysine, and histidine, or glutamine and aspartic acid
  • the antibodies or antigen-binding fragments thereof provided herein further comprise a conjugate.
  • the conjugate can be a detectable label, a pharmacokinetic modifying moiety, a purification moiety or a cytotoxic moiety.
  • a variety of conjugates may be linked to the antibodies or antigen-binding fragments provided herein (see, for example, “Conjugate Vaccines” , Contributions to Microbiology and Immunology, J.M. Cruse and R.E. Lewis, Jr. (eds. ) , Carger Press, New York, (1989) ) .
  • the MG53 antibodies or antigen-binding fragments thereof disclosed herein may be engineered to contain one or more conjugate binding sites that may be utilized for binding to one or more conjugates.
  • such site may include one or more reactive amino acid residues, such as cysteine or histidine residues, to facilitate covalent linkage to a conjugate.
  • the MG53 antibodies or antigen-binding fragments thereof may be linked to a conjugate indirectly, or through another conjugate.
  • the MG53 antibodies or antigen-binding fragments thereof may be conjugated to biotin, then indirectly conjugated to a second conjugate (e.g., avidin) that is conjugated to biotin.
  • the conjugate can be a detectable label, for example, fluorescent label (e.g.
  • enzyme-substrate label e.g. horseradish peroxidase, alkaline phosphatase, luceriferases, glucoamylase, lysozyme, saccharide oxidases, or ⁇ -D-galactosidase
  • radioisotope e.g.
  • the conjugate can be PEG which helps increase half-life of the antibody.
  • the conjugate can be a purification moiety such as a magnetic bead.
  • the conjugate can be a cytotoxic moiety, for example, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, puromycin and analogs thereof, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine) , alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan,
  • One aspect of the present disclosure provides an MG53 ligand and MG53 binding fragment thereof.
  • the MG53 ligand and MG53 binding fragment thereof can be used as MG53 detection agent or MG53 inhibitor.
  • presence or concentration of cell-free MG53 can be determined by detecting binding of an MG53 ligand or MG53 binding fragment thereof to cell-free MG53; binding of MG53 to original effector ligand/receptor can also be blocked or reduced by administering MG53 ligand or MG53 binding fragment thereof to a subject to compete for binding to MG53.
  • the MG53 ligand is an insulin receptor or a variant thereof, e.g., human insulin receptor of SEQ ID NO: 58. In some embodiments, the MG53 ligand is the extracellular region of an insulin receptor or a variant thereof.
  • Nonfunctional protein fragment of MG53 can be any protein fragment which is lack of one or more physiological functions compared with a native MG53.
  • the nonfunctional protein fragment of MG53 can compete with a native MG53 for binding to an MG53 ligand/receptor.
  • activity of the nonfunctional protein fragment of MG53 is reduced by at least 40%, 50%, 60%, 70%, 80%, 90%, 95%or more compared with a native MG53.
  • the aforementioned reduced activity is the activity relating to an MG53 related disease.
  • the aforementioned reduced activity is the activity of MG53 activating AKT phosphorylation.
  • the nonfunctional protein fragment of MG53 is an MG53 mutant.
  • the term “MG53 mutant” or “MG53 protein mutant” as used herein refers to an MG53 protein variant or fragment in which the natural amino acid sequence of a wild-type MG53 protein is modified. Such modifications include, but are not limited to, deletion and/or substitution of one or more amino acids.
  • the MG53 mutant of the present disclosure is identical to the amino acid sequence of a wide-type MG53 except for at least one serine in the coiled-coil-SPRY region of the wild-type MG53 is deleted and/or mutated into any other non-serine or non-threonine amino acids.
  • the MG53 mutant is identical to the amino acid sequence of a wide-type MG53 except for at least one serine in the coiled-coil-SPRY region of the wild-type MG53 is mutated into a non-polar amino acid.
  • the non-polar amino acid is selected from the group consisting of glycine, alanine, leucine, isoleucine, valine, proline, phenylalanine, methionine, and tryptophan.
  • the non-polar amino acid is alanine.
  • the MG53 mutant is identical to the amino acid sequence of a wide-type MG53 except for at least one serine in the coiled-coil-SPRY region of the wild-type MG53 is mutated into any non-serine or non-threonine polar amino acids.
  • the polar amino acid is selected from the group consisting of glutamine, cysteine, asparagine, tyrosine, aspartic acid, glutamic acid, lysine, arginine, and histidine.
  • the polar amino acid is cysteine.
  • the MG53 mutant has an amino acid sequence of any of SEQ ID NOs: 48-57.
  • the amino acid sequence of the MG53 mutant has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99%sequence homology to the amino acid sequence of any of SEQ ID NOs: 48-57.
  • MG53 mutant may refer to Chinese Patent Application No. 201610847346.4, which is incorporated herein by reference in its entirety.
  • the MG53 antibody or antigen-binding fragment thereof, MG53 ligand or MG53 binding fragment hereof, or nonfunctional protein fragment of MG53 of the present disclosure can be prepared by e.g., chemical synthesis or genetic engineering.
  • Solid phase synthesis includes, e.g., Merrifield solid phase synthesis, which has been described in detail in the literature, e.g. Merrifield, J. Am. Chem. Soc. 85: 2149-2154, M. Bodanszky et al., Peptide Synthesis, John Wiley & Sons, Second Edition, 1976, and J. Meienhofer, “Hormonal Proteins and Peptides” , Vol. 2, p. 46, Academic Press (New York) , 1983, which are incorporated herein by reference in their entirety.
  • Merrifield solid phase synthesis mainly comprises the following steps: first the protected carboxyl terminal amino acid is linked to resin according to the amino acid sequence of the target polypeptide; the resin is washed after the linking; the protective group (e.g., t-butyloxy carbonyl) on alpha amino group of carboxyl terminal amino acid is removed, during which it must be ensured that the linkage bond between the amino acid and the resin is not broken; and then the penultimate carboxyl terminal protected amino acid is conjugated to the resulted resin, during which an amide bond is formed between the free carboxyl group of the second amino acid and the amino group of the first amino acid linked to the resin; the preceding reaction processes are repeated successively according to the sequence of amino acid of the target polypeptide, until all amino acids are linked to the resin; finally, the protected peptide is cut off from the resin, and the target polypeptide is obtained after the protective group is removed.
  • the protective group e.g., t-butyloxy carbonyl
  • the polypeptides of the present disclosure can also be prepared by liquid phase synthesis, e.g., by standard solution peptide synthesis, which has been described in detail in the literature E. Schroder and K. Kubke, The Peptides, Vol. 1, Academic Press (New York) , 1965, which is incorporated herein by reference in its entirety.
  • Liquid phase synthesis mainly comprises coupling amino acid or peptide fragment step by step, utilizing amide bond forming chemical or enzyme method.
  • the MG53 antibody or antigen-binding fragment thereof, MG53 ligand or MG53 binding fragment hereof, or nonfunctional protein fragment of MG53 can be produced by genetic engineering through cell culture and expression.
  • a clone or expression vector with a target gene encoding the target protein needed to be expressed is used to transform host cells, and the transformed host cells are cultured in a nutrient medium modified to be suitable for promoter inducement, transformed cell selection or amplification of genes encoding the target sequence (see Sambrook et al. (eds. ) , Molecular Cloning: a Laboratory Manual, (Cold Spring Harbor, 1989) , for detailed description regarding this method) .
  • the host cells in the present disclosure used to produce the antibodies or antigen-binding fragments thereof, MG53 ligand or MG53 binding fragment hereof, or nonfunctional protein fragment of MG53 may be cultured in a variety of media.
  • Commercially available media such as Ham's F10, Minimal Essential Medium (MEM) , RPMI-1640, and Dulbecco's Modified Eagle's Medium (DMEM) , which are produced by Sigma, are suitable for culturing the host cells.
  • MEM Minimal Essential Medium
  • RPMI-1640 RPMI-1640
  • DMEM Dulbecco's Modified Eagle's Medium
  • any of the media described in Ham et al., Meth. Enz. 58: 44 (1979) Barnes et al., Anal. Biochem. 102: 255 (1980) , U.S. Pat. Nos.
  • any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor) , salts (such as sodium chloride, calcium, magnesium, and phosphate) , buffers (such as HEPES) , nucleotides (such as adenosine and thymidine) , antibiotics (such as GENTAMYCIN TM drug) , trace elements (defined as inorganic compounds used to provide nutrients, usually present at final concentrations in the micromolar range, such as ferrous sulfate, cupric sulfate, zinc sulfate, or manganese chloride) , and glucose or an equivalent energy source.
  • the medium may also contain any other necessary supplements at appropriate concentrations that would be known to those skilled in the art. Selection of conditions for host cell culture, such as temperature, pH, and the like selected for expression, is well known to an ordinarily skilled artisan in the art.
  • the target protein can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. If the antibody is produced intracellularly, as a first step, the host cell is subject to lysis, and then the lysed fragments or the particulate debris are removed, for example, by centrifugation or ultrasonic. If the antibody is produced in the periplasmic space, for example, the procedure described in Carter et al., Bio/Technology 10: 163-167 (1992) can be used to isolate the target protein.
  • supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
  • a protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants.
  • the product prepared from the cells can be purified using purification methods such as hydroxylapatite chromatography, gel electrophoresis, dialysis, DEAE-cellulose ion exchange chromatography, ammonium sulfate precipitation, salting out, and affinity chromatography, with affinity chromatography being the preferred purification technique.
  • purification methods such as hydroxylapatite chromatography, gel electrophoresis, dialysis, DEAE-cellulose ion exchange chromatography, ammonium sulfate precipitation, salting out, and affinity chromatography, with affinity chromatography being the preferred purification technique.
  • affinity chromatography being the preferred purification technique.
  • the mixture comprising the product of interest and contaminants may be subjected to low pH hydrophobic interaction chromatography using an elution buffer at a pH between about 2.5 to 4.5, preferably performed at low salt concentrations (e.g., from about 0 to 0.25 M salt) .
  • the present disclosure relates to an isolated nucleic acid, encoding the antibody or antigen-binding fragment of any of the embodiments above.
  • the isolated nucleic acid comprises a nucleotide sequence of SEQ ID NOs: 5, 6, 7, 8, 13, 14, 15, 16, 21, 22, 23, 24, 29, 30, 31, 32, 37, 38, 39 or 40, or a sequence having at least 80%, 85%, 90%, 95%or 99%sequence homology thereof.
  • isolated refers to that a substance (e.g., polypeptide or nucleic acid) is isolated from the environment where it normally exists in nature, or exists in an environment different from where it normally exists in nature.
  • an “isolated” substance has been altered by the hand of man from the natural state. If an “isolated” composition or substance occurs in nature, it has been changed or removed from its original environment, or both.
  • a polynucleotide or a polypeptide naturally present in a living animal is not “isolated, ” but the same polynucleotide or polypeptide is “isolated” if it has been sufficiently separated from the coexisting materials of its natural state so as to exist in a substantially pure state.
  • the antibodies and antigen-binding fragments have a purity of at least 90%, 93%, 95%, 96%, 97%, 98%, 99%as determined by electrophoretic methods (such as SDS-PAGE, isoelectric focusing, or capillary electrophoresis) , or chromatographic methods (such as ion exchange chromatography or reverse phase HPLC) .
  • electrophoretic methods such as SDS-PAGE, isoelectric focusing, or capillary electrophoresis
  • chromatographic methods such as ion exchange chromatography or reverse phase HPLC
  • nucleic acid or “polynucleotide” as used herein refers to ribonucleic acid (RNA) , deoxyribonucleic acid (DNA) or ribonucleic acid-deoxyribonucleic acid mixture (e.g., DNA-RNA hybrid) .
  • Nucleic acid or polynucleotide can be single chain or double chain DNA or RNA, or DNA-RNA hybrid.
  • Nucleic acid or polynucleotide can be linear or cyclic.
  • encode or “encoding” as used herein refers to being able to be transcribed into mRNA and/or translated into peptide or protein.
  • nucleotide including polynucleotide can be in a modification form.
  • the modification includes base modification (e.g., bromouridine) , ribose modification (e.g., cytosine arabinoside and 2’, 3’-dideoxynucleotide) and internucleotide linkage modification (e.g., thiophosphate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phoshoraniladate and phosphoramidate) .
  • base modification e.g., bromouridine
  • ribose modification e.g., cytosine arabinoside and 2’, 3’-dideoxynucleotide
  • internucleotide linkage modification e.g., thiophosphate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phospho
  • the isolated nucleic acid provided herein also includes nucleic acid substituted according to degeneracy of genetic code.
  • degeneracy of genetic code refers to the fact that there are two or more codons for the same amino acid.
  • proline has 4 synonymous codons, i.e., CCU, CCC, CCA, and CCG. It is known in the art, that due to degeneracy of nucleic acid genetic code, nucleic acids in certain sites of a known nuclei acid sequence may be substituted without altering the encoded amino acid sequence. A person skilled in the art can readily make genetic code degeneracy substitution, e.g., through site-directed mutagenesis of bases.
  • codons preferred by the biological cell can be selected to obtain the corresponding encoding gene, and the antibody or antigen-binding fragment of the present disclosure is thus generated through recombination and expression.
  • the present disclosure relates to a clone or expression vector comprising the isolated nucleic acid of any of the embodiments above.
  • vector refers to a vehicle into which a polynucleotide encoding a protein may be operably inserted so as to bring about the expression of that protein.
  • a vector may be used to transform, transduce, or transfect a host cell so as to bring about expression of the genetic material element it carries within the host cell.
  • vectors include plasmids, phagemids, cosmids, artificial chromosomes such as yeast artificial chromosome (YAC) , bacterial artificial chromosome (BAC) , or P1-derived artificial chromosome (PAC) , bacteriophages such as lambda phage or M13 phage, and animal viruses.
  • Exemplary animal viruses used as vectors include retrovirus (including lentivirus) , adenovirus, adeno-associated virus, herpesvirus (e.g., herpes simplex virus) , poxvirus, baculovirus, papillomavirus, and papovavirus (e.g., SV40) .
  • a vector may contain a variety of elements for controlling expression, including promoter sequences, transcription initiation sequences, enhancer sequences, selectable elements, and reporter genes.
  • the vector may contain an origin of replication.
  • a vector may also include materials to aid in its entry into the cell, including but not limited to a viral particle, a liposome, or a protein coating.
  • the present disclosure relates to a host cell, which comprises the clone or expression vector of any of the embodiments above.
  • host cell refers to a cell into which nucleic acid sequence (s) encoding one or more antibodies, MG53 ligands or nonfunctional protein fragment of MG53 provided herein ( “selected genes” ) have been introduced or capable of being introduced, and which further expresses or is capable of expressing the selected genes of interest.
  • selected genes include progeny of the parent cells, as long as the selected genes are present, regardless of whether the progeny is identical to the patent cells in morphology or in genetic make-up.
  • a vector containing polynucleotides encoding selected genes can be introduced into a host cell for cloning or gene expression.
  • a host cell suitable for cloning or expressing DNA in the vector is a prokaryotic cell or eukaryotic cell.
  • Prokaryotic cells suitable for the present disclosure include eubacteria and archaeba, wherein eubacteria include gram negative bacteria, gram positive bacteria and actinomyces.
  • Exemplary eubacteria include, e.g., Colibactilus, Erwinia, Klebsiella, Proteus, Salmonella, Serratia, and Shigella, Pseudomonas, etc.
  • eukaryotic microbes such as filamentous fungi or yeast can also serve as host cells for cloning and expressing vectors of selected genes.
  • Saccharomyces cerevisiae or common baker's yeast, is the most commonly used among lower eukaryotic host microorganisms.
  • Kluyveromyces hosts e.g., K. lactis, K. fragilis (ATCC 12, 424) , K. bulgaricus (ATCC 16, 045) , K. wickeramii (ATCC 24, 178) , K.
  • waltii ATCC 56, 500
  • K. drosophilarum ATCC 36, 906
  • K. thermotolerans K. marxianus
  • yarrowia EP 402, 226)
  • Pichia pastoris EP 183, 070
  • Candida Trichoderma reesia
  • Neurospora crassa Schwanniomyces such as Schwanniomyces occidentalis
  • filamentous fungi e.g., Neurospora, Penicillium, Tolypocladium, and Aspergillus
  • the host cell provided herein can be a mammalian host cell, and culture of mammalian host cells has become a routine procedure.
  • useful mammalian host cells are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651) ; human embryonic kidney line (293 or 293 cells subcloned in suspension culture, see Graham et al., J. Gen Virol. 36: 59 (1977) ) ; baby hamster kidney cells (BHK, ATCC CCL 10) ; Chinese hamster ovary cells/-DHFR (CHO, see Urlaub et al., Proc. Natl. Acad. Sci. USA 77: 4216 (1980) ) ; mouse sertoli cells (TM4, Mather, Biol.
  • monkey kidney cells (CV1 ATCC CCL 70) ; African green monkey kidney cells (VERO-76, ATCC CRL-1587) ; human cervical carcinoma cells (HELA, ATCC CCL 2) ; canine kidney cells (MDCK, ATCC CCL 34) ; buffalo rat liver cells (BRL 3A, ATCC CRL 1442) ; human lung cells (W138, ATCC CCL 75) ; human liver cells (Hep G2, HB 8065) ; mouse mammary tumor (MMT 060562, ATCC CCL51) ; TRI cells (see Mather et al., Annals N.Y. Acad. Sci. 383: 44-68 (1982) ) ; MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2) .
  • a small molecule compound binding to MG53 can be used as an MG53 detection agent or MG53 inhibitor.
  • the small molecule compound binding to MG53 is a compound having Formula Ia or Formula Ib, or a pharmaceutically acceptable salt thereof,
  • L is a chemical bond, or an optionally substituted C 1 -C 12 alkylene
  • M is a chemical bond, or an optionally substituted C 6 -C 12 arylene or a 5-12 membered heterocyclic subunit
  • Y is an optionally substituted C 6 -C 12 aryl or 5-12 membered heterocyclic group
  • Q is an optionally substituted C 1 -C 12 alkylene or C 2 -C 12 alkenylene
  • T is an optionally substituted C 6 -C 12 aryl or 5-12 membered heterocyclic group
  • each R 1 is independently hydrogen, nitro, halogen, hydroxyl, cyano, or an optional
  • the small molecule compound binding to MG53 is a compound having Formula II, or a pharmaceutically acceptable salt thereof,
  • A is hydrogen, or an optionally substituted C 6 -C 12 aryl or 5-12 membered heterocyclic group
  • G is -C (O) -O-R 3 , wherein R 3 is an optionally substituted C 1 -C 12 alkyl
  • J is an optionally substituted C 6 -C 12 aryl
  • G and J together with the carbon atom linked thereto, form wherein each R 4 is independently hydrogen, nitro, halogen, hydroxyl, cyano, or an optionally substituted C 1 -C 12 alkyl or C 1 -C 12 alkoxy, n is any integral from 1 to 4
  • B is a chemical bond, or an optionally substituted C 6 -C 12 arylene or 5-12 membered heterocyclic subunit
  • D is a chemical bond, or an optionally substituted -O-R 5 -, wherein R 5 is optionally substituted C 1 -C 12 alkylene
  • E is an optionally substituted C 6 -C 12 aryl
  • the small molecule compound binding to MG53 is a compound having Formula III, or a pharmaceutically acceptable salt thereof,
  • A is hydrogen, or an optionally substituted C 6 -C 12 aryl or 5-12 membered heterocyclic group; each Y is independently optionally substituted C 6 -C 12 aryl or 5-12 membered heterocyclic group; R 7 is hydrogen, nitro, halogen, hydroxyl, cyano, or optionally substituted C 1 -C 12 alkyl or C 1 -C 12 alkoxy.
  • substituted when refers to a chemical group, means the chemical group has one or more hydrogen atoms that is/are removed and replaced by substituents.
  • substituted has the ordinary meaning known in the art and refers to a chemical moiety that is covalently attached to, or if appropriate, fused to a parent group.
  • the term “optionally substituted” means that the chemical group may have no substituents (i.e. unsubstituted) or may have one or more substituents (i.e. substituted) . It is to be understood that substitution at a given atom is limited by valency.
  • illustrating examples of the substituent mentioned in any of the embodiments above are: nitro, halogen, hydroxyl, cyano, C 1 -C 12 alkyl or C 1 -C 12 alkoxy, benzamide group, or -C (O) -O-R 6 , wherein the C 1 -C 12 alkyl, C 1 -C 12 alkoxy or benzamide group can further be substituted by nitro, halogen, hydroxyl, cyano, C 1 -C 3 alkyl or C 1 -C 3 alkoxy, wherein R 6 is hydrogen or C 1 -C 3 alkyl.
  • C n -C m indicates a range of the carbon atoms numbers, wherein n and m are integers and the range of the carbon atoms numbers includes the endpoints (i.e., n and m) and each integer point in between.
  • C 1 -C 6 indicates a range of one to six carbon atoms, including one carbon atom, two carbon atoms, three carbon atoms, four carbon atoms, five carbon atoms and six carbon atoms.
  • alkyl refers to a saturated hydrocarbon group that may be straight-chain or branched-chain.
  • C n-m alkyl refers to an alkyl having n to m carbon atoms. In certain embodiments, the alkyl group contains 1 to 12, 1 to 8, 1 to 6, 1 to 4, 1 to 3, or 1 to 2 carbon atoms.
  • alkyl group examples include, but are not limited to, chemical groups such as methyl, ethyl, n-propyl, isopropyl, n -butyl, tert-butyl, isobutyl, sec-butyl, 2-methyl-1-butyl, n-pentyl, 3-pentyl, n-hexyl, 1, 2, 2-trimethylpropyl, and the like.
  • alkenyl refers to an unsaturated hydrocarbon group that may be straight-chain or branched-chain having at least one carbon-carbon double bond.
  • the alkenyl group contains 2 to 12, 2 to 10, 2 to 8, 2 to 6, 2 to 5, 2 to 4, or 2 to 3 carbon atoms.
  • the alkenyl group contains 1 to 6, 1 to 5, 1 to 4, 1 to 3, 1 to 2, or 1 carbon-carbon double bond.
  • alkenyl groups include, but are not limited to, chemical groups such as ethenyl, n-propenyl, isopropenyl, n-butenyl, sec-butenyl, and the like.
  • alkylene refers to a divalent saturated hydrocarbon moiety that may be straight-chain or branched-chain, and is linked to two other moieties of a molecule.
  • C n -C m alkylene refers to an alkylene having n to m carbon atoms.
  • the alkylene group contains 1 to 12, 1 to 10, 1 to 8, 1 to 6, 1 to 5, 1 to 4, or 1 to 3 carbon atoms.
  • alkylene groups include, but are not limited to, chemical groups such as methylene, ethidene, 1-methyl-methylene, propylidene, butylidene, and the like.
  • alkenylene refers to a divalent unsaturated hydrocarbon moiety that may be straight-chain or branched-chain having at least one carbon-carbon double bond, and is linked to two other moieties of a molecule.
  • C n -C m alkenylene refers to an alkylene having n to m carbon atoms.
  • the alkenylene group contains 2 to 12, 2 to 10, 2 to 8, 2 to 6, 2 to 5, 2 to 4, or 2 to 3 carbon atoms.
  • the alkenylene group contains 1 to 6, 1 to 5, 1 to 4, 1 to 3, 1 to 2, or 1 carbon-carbon double bond.
  • aryl or “aromatic” , whether as part of another term or used independently, refers to a mono-or poly-carbocyclic ring system radicals with alternating double and single bonds between carbon atoms forming the rings.
  • C n -C m aryl refers to an aryl having n to m carbon atoms forming the ring.
  • the aryl ring systems have 5 to 10, 5 to 8, or 5 to 6 carbon atoms in one or more rings.
  • the aryl ring systems have two or more rings fused together.
  • aryl groups include, but are not limited to, chemical groups such as phenyl, naphthyl, tetrahydronaphthyl, indanyl, idenyl and the like.
  • phenyls include fused phenyls, e.g., benzo dioxolanyl.
  • arylene refers to a divalent aromatic ring or ring system that is linked to two other moieties of a molecule, i.e., the aforementioned two moieties are bonded to the ring at two different ring positions.
  • the aromatic ring of an arylene is a monocyclic ring system
  • the two moieties are bonded at two ring positions of the same ring.
  • the aromatic ring of an arylene is a polycyclic ring system
  • the two moieties may be bonded at two ring positions of the same ring or different rings.
  • C n -C m arylene refers to an arylene having n to m carbon atoms forming the ring.
  • An arylene may be substituted or unsubstituted.
  • An unsubstituted arylene has no other substituents apart from the two moieties of the molecule it is linked to.
  • a substituted arylene has other substituents apart from the two moieties of the molecule it is linked to.
  • alkoxy refers to a group of the formula “-O-alkyl” .
  • C n -C m alkoxy means that the alkyl moiety of the alkoxy group has n to m carbon atoms. In certain embodiments, the alkyl moiety has 1 to 6, 1 to 4, or 1 to 3 carbon atoms. Examples of alkoxy groups include, but are not limited to, chemical groups such as methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy) , t-butoxy, and the like.
  • n membered wherein n is an integer, typically employed in combination with a ring system to describe the number of ring-forming atoms in the ring system.
  • piperidinyl is an example of a 6 membered heterocycloalkyl ring
  • pyrazolyl is an example of a 5 membered heteroaryl ring
  • pyridyl is an example of a 6 membered heteroaryl ring
  • 1, 2, 3, 4-tetrahydro-naphthalene is an example of a 10 membered cycloalkyl group.
  • heteroaryl refers to aryl group wherein at least one ring atom in the aromatic ring is a heteroatom, and the remainder of the ring atoms are carbon atoms.
  • n-m membered heteroaryl refers to heteroaryl having n to m ring-forming members.
  • Example heteroatoms include, but are not limited to, oxygen, sulfur, nitrogen, phosphorus, and the like.
  • heteroaryl can have 5 to 10, 5 to 8, or 5 to 6 ring-forming members.
  • heteroaryl is 5 membered or 6 membered heteroaryl.
  • heteroaryl examples include, but are not limited to, furanyl, thienyl, pyridyl, quinolyl, pyrrolyl, N-lower alkyl pyrrolyl, pyridyl-N-oxide, pyrimidyl, pyrazinyl, imidazolyl, indolyl and the like.
  • a 5 membered heteroaryl is a heteroaryl with a ring having five ring-forming atoms, wherein one or more (e.g., 1, 2, or 3) ring atoms can be independently selected from N, O, P, and S.
  • Examples of 5 membered heteroaryl include, but are not limited to, thienyl, furyl, pyrrolyl, imidazolyl, thiazolyl, oxazolyl, pyrazolyl, isothiazolyl, isoxazolyl, 1, 2, 3-triazolyl, tetrazolyl, 1, 2, 3-thiadiazolyl, 1, 2, 3-oxadiazolyl, 1, 2, 4-triazolyl, 1, 2, 4-thiadiazolyl, 1, 2, 4-oxadiazolyl, 1, 3, 4-triazolyl, 1, 3, 4-thiadiazolyl, 1, 3, 4-oxadiazolyl and the like.
  • a 6 membered heteroaryl is a heteroaryl with a ring having six ring atoms, wherein one or more (e.g., 1, 2, or 3) ring atoms can be independently selected from N, O, P, and S.
  • 6 membered heteroaryl include, but are not limited to, pyridyl, pyrazinyl, pyrimidinyl, triazinyl, pyridazinyl and the like.
  • heteroarylene refers to a divalent heteroaryl that is linked to two other moieties of a molecule, i.e., the aforementioned two moieties are bonded to the ring at two different ring positions.
  • aromatic ring of a heteroarylene is a monocyclic ring system
  • the two moieties are bonded at two ring positions of the same ring.
  • aromatic ring of a heteroarylene is a polycyclic ring system
  • the two moieties may be bonded at two ring positions of the same ring or different rings.
  • a heteroarylene may be substituted or unsubstituted.
  • An unsubstituted heteroarylene has no other substituents apart from the two moieties of the molecule it is linked to.
  • a substituted heteroarylene has other substituents apart from the two moieties of the molecule it is linked to.
  • heterocyclic alkyl refers to cycloalkyl group wherein at least one ring atom in the ring systems is a heteroatom, and the remainder of the ring atoms being carbon atoms.
  • n-m membered heterocyclic alkyl refers to heterocyclic alkyl having n to m ring-forming members.
  • the ring may also have one or more double bonds, but not have a completely conjugated system.
  • the heterocyclic alkyl is saturated heterocyclic alkyl. Examples of heteroatoms include, but are not limited to, oxygen, sulfur, nitrogen, phosphorus, and the like.
  • heterocyclic alkyl has 3 to 8, 3 to 6, or 4 to 6 ring-forming carbons.
  • heterocyclic alkyl include, but are not limited to, azetidine, aziridine, pyrrolidyl, piperidyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, and the like.
  • heterocyclic alkylene refers to a divalent heterocyclic alkyl that is linked to two other moieties of a molecule, i.e., the aforementioned two moieties are bonded to the ring at two different ring positions.
  • C n-m heterocyclic alkylene refers to a heterocyclic alkylene having n to m carbon atoms.
  • the heterocyclic alkylene contains 3 to 12, 3 to 6, or 4 to 6 carbon atoms.
  • heterocyclic group includes heteroaryl and heterocyclic alkyl.
  • heterocyclic subunit includes heteroarylene and heterocyclic alkylene.
  • halo and halogen refer to an atom selected from fluorine, chlorine, bromine and iodine.
  • cyano refers to a group of the formula “-CN” .
  • hydroxyl refers to a group of the formula “-OH” .
  • nitro refers to a group of the formula “-NO 2 ” .
  • the term “compound” is meant to include all stereoisomers (e.g., enantiomers and diastereomers) , geometric iosomers, tautomers, and isotopes of the structures depicted. Compounds herein identified by name or structure as one particular tautomeric form are intended to include other tautomeric forms unless otherwise specified.
  • the compounds described herein can be asymmetric (e.g., having one or more stereocenters) . All stereoisomers, such as enantiomers and diastereomers, are intended to be included unless otherwise indicated.
  • Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically inactive starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, carbon-carbon double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present disclosure. Cis and trans geometric isomers of the compounds of the present disclosure are described and may be isolated as a mixture of isomers or as separated isomeric forms.
  • the compounds described herein have the (R) -configuration. In certain embodiments, the compounds described herein have the (S) -configuration.
  • An example method includes fractional recrystallizaion using a chiral resolving acid, which is an optically active, salt-forming organic acid.
  • Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid, or the various optically active camphorsulfonic acids.
  • resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of N-methyl benzyl amine, 2-phenylglycinol, norephedrine, ephedrine, N-methylephedrine, cyclohexylethylamine, 1, 2-diaminocyclohexane, and the like.
  • Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine) .
  • an optically active resolving agent e.g., dinitrobenzoylphenylglycine
  • Suitable elution solvent composition can be determined by those skilled in the art.
  • Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton.
  • Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge.
  • Example prototropic tautomers include ketone-enol pairs, amide-imidic acid pairs, lactam-lactim pairs, enamine-imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, 1H-and 3H-imidazole, 1H-, 2H-and 4H-1, 2, 4-triazole, 1H-and 2H-isoindole, and 1H-and 2H-pyrazole.
  • Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
  • Compounds of the present disclosure can also include all isotopes of atoms occurring in the intermediates or final compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include protium, deuterium and tritium.
  • the small molecule compounds of the present disclosure can be obtained through organic synthesis.
  • Compounds of the present disclosure including salts, esters, hydrates, or solvates thereof, can be prepared using any known organic synthesis techniques and can be synthesized according to any of numerous possible synthetic routes.
  • the reactions for preparing compounds of the present disclosure can be carried out in suitable solvents, which can be readily selected by those skilled in the art of organic synthesis.
  • suitable solvents can be substantially non-reactive with the starting materials (reactants) , the intermediates, or products at the temperatures at which the reactions are carried out, e.g., temperatures that can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected by those skilled in the art.
  • Preparation of compounds of the present disclosure can involve the protection and deprotection of various chemical groups.
  • the need for protection and deprotection, and the selection of appropriate protecting groups, can be readily determined by those skilled in the art.
  • the chemistry of protecting groups can be found, for example, in T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 3rd Ed., Wiley & Sons, Inc., New York (1999) , which is incorporated herein by reference in its entirety.
  • Reactions can be monitored according to any suitable method known in the art.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C) , infrared spectroscopy, spectrophotometry (e.g., UV-visible) , mass spectrometry, or by chromatographic methods such as high performance liquid chromatography (HPLC) , liquid chromatography-mass spectroscopy (LCMS) , or thin layer chromatography (TLC) .
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C) , infrared spectroscopy, spectrophotometry (e.g., UV-visible) , mass spectrometry, or by chromatographic methods such as high performance liquid chromatography (HPLC) , liquid chromatography-mass spectroscopy (LCMS) , or thin layer chromatography (TLC) .
  • HPLC high performance liquid
  • HPLC high performance liquid chromatography
  • Preparative LC-MS Purification: Improved Compound Specific Method Optimization Karl F. Blom, Brian Glass, Richard Sparks, Andrew P. Combs J. Combi. Chem. 2004, 6 (6) , 874-883, which is incorporated herein by reference in its entirety
  • normal phase silica chromatography Karl F. Blom, Brian Glass, Richard Sparks, Andrew P. Combs J. Combi. Chem. 2004, 6 (6) , 874-883, which is incorporated herein by reference in its entirety
  • the small molecule compound of the present disclosure can be purchased by commercial approaches.
  • the small molecule compounds of the present disclosure can be commercially available compound libraries, e.g., SPECS compound library, Netherlands.
  • the present disclosure relates to a pharmaceutical composition
  • a pharmaceutical composition comprising an active substance (e.g., the antibody, antigen-binding fragment, MG53 ligand or nonfunctional fragment of MG53 of any of the embodiments above, the clone or expression vector of any of the embodiments above, or the cell of any of the embodiments above or any of the small molecule compounds above) , and a pharmaceutically acceptable excipient.
  • an active substance e.g., the antibody, antigen-binding fragment, MG53 ligand or nonfunctional fragment of MG53 of any of the embodiments above, the clone or expression vector of any of the embodiments above, or the cell of any of the embodiments above or any of the small molecule compounds above
  • a pharmaceutically acceptable excipient e.g., the antibody, antigen-binding fragment, MG53 ligand or nonfunctional fragment of MG53 of any of the embodiments above, the clone or expression vector of any of the embodiments above, or the cell of any
  • compositions can be prepared in a manner known in the pharmaceutical art.
  • the compounds of the present disclosure may be admixed with pharmaceutically acceptable excipient for the preparation of pharmaceutical compositions.
  • the phrase “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • compounds, materials, compositions, and/or dosage forms that are pharmaceutically acceptable refer to those approved by a regulatory agency (such as U.S. Food and Drug Administration, China Food and Drug Administration or European Medicines Agency) or listed in generally recognized pharmacopoeia (such as U.S. Pharmacopoeia, China Pharmacopoeia or European Pharmacopoeia) for use in animals, and more particularly in humans.
  • the pharmaceutically acceptable carriers for use in the pharmaceutical compositions of the present invention may include, but are not limited to, for example, pharmaceutically acceptable liquids, gels, or solid carriers, aqueous vehicles (e.g., sodium chloride injection, Ringer's injection, isotonic glucose injection, sterile water injection, or Ringer's injection of glucose and lactate) , non-aqueous vehicles (e.g., fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil, or peanut oil) , antimicrobial agents, isotonic agents (such as sodium chloride or dextrose) , buffers (such as phosphate or citrate buffers) , antioxidants (such as sodium bisulfate) , anesthetics (such as procaine hydrochloride) , suspending/dispending agents (such as sodium carboxymethylcellulose, hydroxypropyl methylcellulose, or polyvinylpyrrolidone) , chelating agents (such as EDTA (ethylenediamine tetra
  • the pharmaceutical composition is an oral formulation.
  • the oral formulations include, but are not limited to, capsules, cachets, pills, tablets, troches (for taste substrates, usually sucrose and acacia or tragacanth) , powders, granules, or aqueous or non-aqueous solutions or suspensions, or water-in-oil or oil-in-water emulsions, or elixirs or syrups, or confectionery lozenges (for inert bases, such as gelatin and glycerin, or sucrose or acacia) and /or mouthwash and its analogs.
  • the oral solid formulation (e.g., capsules, tablets, pills, dragees, powders, granules, etc. ) includes the active substance and one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or the followings: (1) fillers or extenders such as starch, lactose, sucrose, glucose, mannitol and/or silicic acid; (2) binders such as, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and/or acacia; (3) humectants such as glycerol; (4) cleaving agents such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) retarder solutions such as paraffin; (6) accelerating absorbers such as quaternary ammonium compounds; (7) lubricants such as acetyl alcohol and glycerol monostearate
  • the oral liquid formulation includes pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs, etc.
  • the liquid dosage forms may also contain conventional inert diluents such as water or other solvents, solubilizers and emulsifiers such as ethanol, isopropanol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzene (meth) acrylate, propylene glycol, 1, 3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, olive, castor and sesame oils) , glycerol, tetrahydrofurfuryl alcohol, polyethylene glycol and fatty acid sorbitol esters, and mixtures thereof.
  • the oral compositions may also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening,
  • the pharmaceutical composition may be an injectable formulation, including sterile aqueous solutions or dispersions, suspensions or emulsions.
  • the injectable formulation should be sterile and should be liquid to facilitate injections. It should be stable under the conditions of manufacture and storage, and should be resistant to the infection of microorganisms (such as bacteria and fungi) .
  • the carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyols (e.g., glycerol, propylene glycol, and liquid polyethylene glycols, etc. ) and suitable mixtures and /or vegetable oils thereof.
  • the injectable formulation should maintain proper fluidity, which may be maintained in a variety of ways, for example, using a coating such as lecithin, using a surfactant, etc.
  • Antimicrobial contamination can be achieved by the addition of various antibacterial and antifungal agents (e.g., parabens, chlorobutanol, phenol, sorbic acid, thimerosal, etc. ) .
  • the pharmaceutical composition is an oral spray formulation or nasal spray formulation.
  • spray formulations include, but are not limited to, aqueous aerosols, non-aqueous suspensions, liposomal formulations, or solid particulate formulations, etc.
  • Aqueous aerosols are formulated by combining an aqueous solution or suspension of the agent with a conventional pharmaceutically acceptable carrier and stabilizer.
  • the carrier and stabilizer may vary according to the needs of specific compounds, but generally include nonionic surfactants (Tweens, or polyethylene glycol) , oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugar or sugar alcohol.
  • Aerosols are usually prepared from isotonic solutions and can be delivered by nebulizers.
  • the pharmaceutical compositions may be used in combination with one or more other drugs.
  • the composition comprises at least one other drug.
  • the other drugs are cardiovascular drugs, drugs for treating kidney diseases, drugs for cell membrane repair, etc.
  • the pharmaceutical compositions may be delivered to the subject by suitable routes including, but not limited to, the oral route, injection route (e.g., intravenous injection, intramuscular injection, subcutaneous injection, intradermal injection, intracardiac injection, intrathecal injection, intrapleural injection, intraperitoneal injection, etc. ) , mucosal route (e.g., intranasal administration, oral administration, etc. ) , sublingual route, rectal route, transdermal route, intraocular route, pulmonary route.
  • injection route e.g., intravenous injection, intramuscular injection, subcutaneous injection, intradermal injection, intracardiac injection, intrathecal injection, intrapleural injection, intraperitoneal injection, etc.
  • mucosal route e.g., intranasal administration, oral administration, etc.
  • sublingual route e.g., intranasal administration, oral administration, etc.
  • rectal route e.g., transdermal route, intraocular route, pulmonary route.
  • the present disclosure relates to a kit for detecting cell-free MG53.
  • the kit comprises an MG53 detection agent (e.g., the MG53 detection agent described in the present disclosure) .
  • the kit further comprises one or more other components, e.g., MG53 standard solution, diluent, washing solution, stop solution and chromogenic agent, etc.
  • the kit provided herein can be used in a method for detecting cell-free MG53 provided herein.
  • Reagents in the kit can be placed in any kind of container, such that each component in the reagent is preserved stably and will not be absorbed or altered by the material of the container.
  • lyophilized regulating substrates/molecules and /or buffers may be contained within a sealed glass ampoule which is packed under neutral, non-reactive gas such as nitrogen.
  • the ampoule can be composed of any suitable material, e.g., glass, organic polymer such as polycarbonate or polystyrene and the like, ceramic, metal or any other material commonly used to store reagents.
  • suitable containers include simple bottles, which may be made from materials similar to ampoules, and envelopes, which may be composed of metal foil lining (e.g., aluminium or alloy) .
  • Other containers include tubes, vials, flasks, bottles, syringes, and the like.
  • the container may have a sterile inlet, such as a bottle having a stopper that can be punctured using hypodermic needle.
  • Other containers may have two compartments separated by an easily removable a film, upon removal of which the components can be mixed.
  • the removable film can be glass, plastic, rubber, and the like.
  • the kit also comprises an enzyme-labeled plate and sealing films.
  • the kit can also be provided together with instructions.
  • the instructions can be printed on paper or other base materials, and/or provided in the form of digitally readable media, such as diskette, CD-ROM, DVD-ROM, zip disk, video tape, compact disk, tape, and the like.
  • Detailed instructions may be not physically associated to the kit, but as an alternative, the users may be alternatively directed to a website designated by the manufacturer or distributor of the kit, or the instructions may be provided in the form of emails. Methods for detecting cell-free MG53 are recorded in the instructions.
  • the present disclosure relates to use of an MG53 inhibitor in the manufacture of a medicament for reducing or inhibiting cell-free MG53 in a subject.
  • the MG53 inhibitor can be any of the MG53 inhibitor above.
  • the method can be used to ameliorate, treat or predict an MG53 related disease or a symptom thereof.
  • the MG53 related disease can be any of the MG53 related diseases above.
  • the medicament is a pharmaceutical composition of the present disclosure.
  • the present disclosure relates to use of an MG53 detection agent in the manufacture of a kit for detecting cell-free MG53.
  • the kit can be used for detecting an MG53 related disease, predicting a risk or development of an MG53 related disease, identifying potential MG53 inhibitor, evaluating therapeutic effect for an MG53 related disease, or detecting activity of cell-free MG53.
  • the MG53 detection agent is provided in the form of a pharmaceutical composition of the present disclosure.
  • Example 1 preparation and characterization of MG53 antibodies
  • RNA extracted from mouse was used for reverse transcription, to synthesize cDNA.
  • cDNA library was obtained by reverse transcription using OligodT as primer and reverse transcriptase.
  • the nucleic acid encoding mouse MG53 (NP _001073401.1) was obtained by PCR method using specific primer and the synthesized cDNA library as template. PCR was performed as follows: denaturation, 98°C for 3 min; circulation process, 98°C for 30 seconds, 65°C for 30 seconds, and 72°C for 90 seconds, 30 cycles; followed by extension of 72°C for 5 min, and then stored at 4°C.
  • upstream primer 5’-ataggtaccg ccaccatgtc ggctgcaccc ggcct-3’ (SEQ ID NO: 59) ; downstream primer: 5’-atactcgagc ggcctgttcc tgctccggcc-3’ (SEQ ID NO: 60) ; carrying with KpnI and XhoI restriction site.
  • the PCR product was subject to double enzyme digestion using KpnI and XhoI, and meanwhile, empty vector plasmid pcDNA4/TO/myc-HisB was treated by the same double enzyme digestion, the gel was extracted and the PCR product was ligated into the vector using T4 ligase, forming a plasmid pcDNA4-mMG53-myc. The sequence was verified as correct by sequencing.
  • the expression plasmids pcDNA4-mMG53-myc were transiently transfected into HEK293 cells. 2 days later, supernatant harvested from the transiently transfected cell culture was used for protein purification. The protein was purified by SDS-PAGE and quantified to be used for immunization.
  • 3 rabbits were respectively immunized with 100 ⁇ g MG53 protein by intraperitoneal injection. After the rabbits were immunized for three to five times, the blood of the rabbits were taken to collect the serum, and the titer of the antibody was measured by ELISA assay, and the rabbit with the highest titer was selected.
  • B lymphocytes were isolated from the spleen of the rabbit with the highest titer.
  • the isolated B lymphocytes and myeloma cells were subject to cell fusion (by the ratio of 1: 1) .
  • the specific experimental procedure of the fusion is as follows: the cell mixture was washed and suspended with 5-10 ml ECF solution. ECF solution was added to adjust the concentration to 2 ⁇ 10 6 cells/ml.
  • the cell suspension in the fusion chamber was immediately transferred into a sterile tube containing medium of more volume. After culture at 37°C for over 24 hours, the cell suspension was mixed and pipetted into a 96-well plate (0.5 ⁇ 10 6 cells/plate) . The cells were cultured at 37°C, under 5%CO 2 . When the clone was large enough, 100 ⁇ l supernatant was transferred from the 96-well plate for antibody screening assay.
  • Binding of the hybridoma supernatant to MG53 protein was detected using ELISA. Briefly, the plate was coated with 1 ⁇ g/ml human MG53 protein overnight at 4°C. After sealing and washing, the hybridoma supernatant was diluted by different times, and then transferred to the coated plate and cultured at room temperature for 1 hour. Subsequently, the plate was washed and then cultured for 45 min with the secondary antibody of goat-anti-rabbit IgG (H+L) HRP (Goat Anti-Rabbit HRP (IgG H&L) (ab6721)
  • the positive standard value (commercially available MG53 antibody) is 0.456; the negative standard value (irrelevant antibody) is 0.083.
  • the five hybridoma cell lines were subcloned. Briefly, for each hybridoma cell line, the cells were counted and diluted to 5 cells/well, 1 cell/well and 0.5 cell/well in the clone medium. The cells were plated into 96-well plates at 200 ⁇ l/well, among the plates, one plate of 5 cells/well, one plate of 1 cell/well and four plates of 0.5 cell/well. All plates were placed under 37°C, 5%CO 2 , and cultured until all cell lines can be tested by ELISA.
  • MG53 antibodies to bind MG53 was verified by surface plasmon resonance (SPR) .
  • SPR surface plasmon resonance
  • a reference flow cell was activated and blocked in the absence of MG53.
  • the immobilization level was fixed at 500 Biacore response units (RU) , and the concentration of MG53 Antibody #84 (amino acid heavy chain sequence as shown in SEQ ID NO: 19, light chain sequence as shown in SEQ ID NO: 20, the concentration of which ranging from 1.95 nM to 1000 nM) flowing over the chip surface was serially increased, the data results of which are shown in Figure 2. It can be inferred from the experimental results above, that the MG53 antibodies of the present disclosure are capable of binding rhMG53 with high affinity.
  • MG53 antibodies to block the binding of MG53 to insulin receptor extracellular region is verified by surface plasmon resonance (SPR) .
  • SPR surface plasmon resonance
  • IR Recombinant insulin receptor
  • Amine Coupling kit GE Healthcare
  • the immobilization level was fixed at 500 Biacore response units (RU) , and the concentration of the reactant flowing over the chip surface was serially increased (rhMG53 was used in the experiment shown in Figure 3a, the concentration of which ranged from 1.95 nM to 1000 nM; a mixture of rhMG53 and MG53 Antibody #84 was used in the experiment shown in Figure 3b, wherein the concentration of MG53 antibody was fixed at 500 nM, while the concentration of rhMG53 ranging from 0 nM to 2000 nM) .
  • RU Biacore response units
  • Example 2 study on function of cell-free MG53
  • Primary antibody incubation Primary antibody solution was prepared by adding 1 ⁇ primary antibody into TBST solution containing 5% (w/v) BSA. Upon completion of sealing, the film was washed three times with TBST, each time for 5 min, added with the corresponding primary antibody solution, and gently shaken on a shaker overnight at 4°C for incubation.
  • Secondary antibody incubation Secondary antibody solution was prepared by diluting the secondary antibody by 1: 2000 in TBST containing 5%skim milk powder. Attention need to be directed to whether the secondary antibody corresponds to the primary antibody, i.e., whether it is mouse antibody or rabbit antibody, etc. The film incubated overnight with primary antibody was taken out, washed three times with TBST, each time for 5 min, and then added with the secondary antibody solution, and gently shaken on a shaker for 1 hour at room temperature for incubation.
  • Chemiluminescent assay The PVDF film incubated with the secondary antibody was taken out, washed three times with TBST, each time for 5 min, and the film was soaked in TBST for subsequent use. Chemiluminescence reaction substrates A and B were prepared, mixed by ratio of 1: 1 (vol/vol) , carefully operated to avoid light, and then added onto the PVDF film, reacted for 5 min, and chemiluminescent assay was performed on the bands by ChemiDocXRS of BIO-RAD.
  • Re-incubating primary antibody The film was washed twice with TBST, each time for 5 min. Elution buffer was added, and the film was washed for 30 min at 50°C, 50 rpm. The film was then washed 6 times with TBST, each time for 5 min. After the antibody was eluted, the protein could be further tested by incubating other corresponding primary antibodies-secondary antibodies. Gray value of blot image of the chemiluminescent assay on the bands was analyzed using ImageJ software.
  • Serum of 37 Type II diabetes patients (T2D) and serum of 21 normal people (Control) were obtained.
  • the serum samples above were tested using the same Western blotting assay as described previously, the results of the assay were shown in Figure 7. It can be inferred from the experimental results that the increase in MG53 content in the serum of Type II diabetes patients is statistically meaningful over the content in that of normal people (using t-test assay) , and thereby whether diabetes is being suffered from and risk of having diabetes can be determined based on content of MG53 in serum.
  • Serum of 18 mice fed high-fat diet only (using Cat. #D12492 feed purchased from ResearchDiets Inc., wherein 60%of the energy is from fat, fed for 35 weeks) and serum of 15 mice fed normal diet were obtained.
  • the serum samples above were tested using the same Western blotting assay as described previously, the results of the assay were shown in Figure 8. It can be inferred from the experimental results that a high-fat diet will induce increase of serum MG53, and thereby risk of having fat metabolism diseases can be determined based on content of MG53 in serum.
  • Serum of 6 12-week-old ZDF rats (Zucker diabetic fatty rats, purchased from Vital River Laboratories (Beijing, China) , Cat# 123) and 6 normal SD rats were obtained.
  • Content of MG53 in the serum was detected using ELISA kit (Cusbio)
  • content of blood glucose was detected using blood glucose meter (Roche)
  • content of blood insulin was detected using ELISA kit (Millipore)
  • body weight was measured. The data is shown in Figure 9. It can be inferred from the experimental results that serum MG53 content is linearly correlated with body weight, and content of blood glucose and blood insulin, and thereby risk and development of diabetes, as well as response to treatment can be determined based on content of MG53 in serum.
  • mice 8-10-week-old C57 (purchased from Vital River) mice were selected, fasted overnight, and then recombinant human MG53 protein (rhMG53) or BSA was injected via tail vein in the dosage of 6 mg/kg. 10 min later, insulin was injected intraperitoneally in the dosage of 1 U/kg or not injected. After another 10 min, the mice were sacrificedand corresponding tissue materials were obtained. Phosphorylation level of akt, which is an important molecule of the insulin signaling pathway, was tested by western blotting in skeletal muscle, liver, fat, and heart tissues for each experimental group, thereby evaluating insulin sensitivity in each of these tissues.
  • rhMG53 human MG53 protein
  • BSA recombinant human MG53 protein
  • the sources of various antibodies therein are as follows: Anti-phospho Ser473 Akt (p-AktS473) , CST Cat# 4060; Anti-total Akt (t-Akt) , CST Cat# 9272; Anti-GAPDH, Bioeasy Technology, Cat# BE0023. Statistical analysis was conducted using One-way anova. **represents that P ⁇ 0.01.
  • mice Male Sprague-Dawley rats (250 to 300 g) or mice (20 to 30 g) were anesthetized with pentobarbital (70 mg/kg, i.p. ) .
  • the rat or mouse heart was excised and perfused on a Langendorff apparatus at a constant pressure of 55 mmHg with Krebs-Henseleit solution (in mM: NaCl 118, KCl 4.7, CaCl 2 2.5, MgSO 4 ⁇ 7H 2 O 1.2, KH 2 PO 4 1.2, and glucose 11.1) .
  • the buffer was continuously gassed with 95%O 2 /5%CO 2 (pH 7.4) and warmed by a heating bath/circulator.
  • the heart temperature was continuously monitored and maintained at 37 ⁇ 0.5°C.
  • the outlet perfusate was collected for different periods of time.
  • 0.75 mM EDTA-Na (without Ca 2+ in the perfusion solution) or 30M BFA were included in the perfusion solution.
  • the collected perfusate samples were centrifuged at 3,000 rpm with Amicon Ultra-15 10K Centrifugal Filter Devices (Millipore, Cat# UFC801096) for 15 min, and this was repeated 3 times. The concentrated perfusate samples were used for subsequent analysis.
  • the MG53 release triggered by high glucose plus insulin occurred in the absence of any change in myocardial lactate dehydrogenase (LDH) or creatine kinase (CK) release (indexes of loss of cell membrane integrity) in perfused rat hearts, indicating that metabolism-regulated MG53 release is not caused by myocardial damage.
  • LDH myocardial lactate dehydrogenase
  • CK creatine kinase
  • glucose 75 g was orally delivered twice at a 30-min interval.
  • a blood sample was taken before the glucose treatment, and at 30 and 90 min after the second dose.
  • the blood samples were centrifuged at 3,000 rpm, and serum was collected for Western blotting
  • Metabolism-induced MG53 release is mediated by a regulated secretory pathway
  • BFA Brefeldin A
  • Cardiac-specific overexpression of MG53 is sufficient to trigger systemic insulin resistance and metabolic syndrome
  • GTTs glucose tolerance tests
  • ITTs insulin tolerance tests
  • the young mg53 h-TG mice exhibited moderate obesity (Figure 34B) , glucose intolerance, and insulin intolerance (Figure 34C and 34D) .
  • cardiacspecific overexpression of MG53 was sufficient to trigger full-blown metabolic syndrome, as manifested by hyperglycemia, hyperinsulinemia, and dyslipidemia (Figure 34E-34G) , abdominal fat accumulation, increased white fat, brown fat, and fat-to-lean ratio (Figure 34H-34M) , together with hepatosteatosis and pancreatic islet hypertrophy (Figure 34N and 34O) .
  • the mg53 h-TG mice displayed severe obesity, glucose intolerance, and insulin intolerance at this time point ( Figure 34B-34D) . Meanwhile, the daily energy expenditure of mg53 h-TG mice was significantly lower than that of the wt counterpart ( Figure 34Q-34S) , but there was no difference in their daily food intake, core body temperature, or physical activity. At the later time point, mg53 h-TG mice also developed diabetic complications such as diabetic cardiomyopathy. Hence, a chronic elevation of circulating MG53 primarily impairs the whole-body insulin response, and secondarily leads to systemic metabolic syndrome that contributes to obesity, diabetes and various cardiovascular complications.
  • mice 20 8-10-week-old db/db mice (diabetes model mice, purchased from Jackson Laboratory (Bar Harbor, ME) Cat# 000642) were selected, wherein 10 mice were used as the control group, injected with IgG (Sigma, I5381) only; the other 10 were used as the therapeutic group, injected with MG53 Antibody #84. Meanwhile, 5 db/+ mice of the same age were ordered. db/+ mice were only used for the final material collection, and were not injected.
  • mice After the mice arrived, they were stabilized for 1 week, and then the body weight and blood glucose of the mice were measured, and the condition thereof were checked. On the day of the experiment, the animals were not fasted, and body weight and blood glucose were measured at 8 a.m. The mice were then grouped according to body weight and blood glucose meausurement, and injection was conducted according to grouping. Subsequently, injection was conducted at 9 a.m., at 1 mg/mouse/time, volume of injection: 0.24ml /mouse/time. Later the mice were fasted, and blood glucose was measured at 3 p.m. and 7 p.m. on the day of the experiment respectively, and body weight was measured at 9 a.m., and blood glucose was measured at 9 a.m. and 3 p.m. on every following day for 10 days.
  • insulin tolerance test (ITT) was performed (see, Song et al., Central role of E3 biquitin ligase MG53 in insulin resistance and metabolic disorders, Nature, 494, 375-381, 2013 for description of the experimental condition) .
  • the principle for screening was to screen by different angles and different approaches. Each screening approach was scored by strength of binding between small molecule compounds and proteins, and each time the molecules with higher scores were selected, and finally the intersection of the screening results by different approaches was adopted.
  • the specific operation is as follows: first, each group in the protein pocket was fixed, which were freely joined with small molecule compounds, and 20,000 compounds with high scores were selected; then, the small molecule compounds and groups were all allowed to freely rotate and join, and four thousand compounds with high scores were selected. Next, pymol software was used for manual screening, five standards for which are as follows: a. the molecular weight of the compound is to be greater than 300 Da, specific response is reduced if the molecular weight is too small; b.
  • the compound at least three hydrogen bonds interact between the compound and the group in the pocket, which is for increasing the interaction between the compound and the protein; c. the compound is to occupy 80%or more of the protein pocket, which is also for reducing non-specificity; d. the compound cannot be a polypeptide, which is to avoid non-specific binding of polypeptides in an organism; e. the compound does not contain any metal atom.
  • 140 small molecule compounds were obtained.
  • IRS1 plasmid IRS1-GFP
  • MG53 plasmid MG53-myc
  • Myc Myc label
  • HEK293T cells were cultured using 10 ml cell culture dish. Medium were changed once every two days, and cells in good growing and health state were selected, and MG53-Myc and IRS1-GFP were co-transfected by the time the cells covered 80%of the entire cell culture dish, and at the same time, pcDNA4 empty vector and IRS1-GFP were co-transfected for the control group. The cells were cultured for 24 hours after transfection, under fluorescence microscope, strong expression of GFP could be found in the control group, while fluorescence intensity in the experimental group was attenuated by more than 50%.
  • the cells were then detached using 0.125%pancreatin, collected by low speed centrifugation, evenly mixed and then spread onto 96-well enzyme-labeled plates respectively, and continued to be cultured. 24 hours later, the cells were subject to change of medium and addition of small molecule compounds to be tested. Culture continued for another 24 hours, and at last the cells were washed 3 times with PBS, and subject to 485nm/528nm fluorescence signal assay. The compound was selected in concentrations of 100 ⁇ M, 10 ⁇ M and 1 ⁇ M respectively. A total of 48 candidate compound molecules were selected.
  • the other 11 compound molecules were poor candidates due to insufficient molecular purity (as measured by means of small molecule mass spectra) , or overly high dissociation constant (dissociation constant being over 50 ⁇ M) , or bad kinetic characteristics exhibited (kinetic curve being linear or not S-shaped, indicating that binding between the molecule and MG53 protein has strong non-specificity) , etc.
  • HEK293T cells under good condition in culture were divided into 6 groups which were transfected with IRS1 and MG53-myc respectively, 4 groups of cells transfected with IRS1 and MG53-myc were respectively treated by adding compounds, concentration of the added compounds were all 50 ⁇ M. 24 hours after the addition of those compounds, extraction of cell total protein and SDS-PAGE gel vertical electrophoresis experiment were performed. Expression of IRS1 and MG53 protein were detected respectively upon exposure, using GAPDH as internal reference.

Abstract

La présente invention concerne un procédé de détection d'une maladie liée à MG53 ou de prédiction d'un risque d'une maladie liée à MG53, comprenant les étapes suivantes : a) obtention d'un échantillon d'essai ; et b) détection de MG53 acellulaire dans l'échantillon d'essai. La présente invention concerne en outre un procédé de réduction ou d'inhibition de l'activité de MG53 acellulaire chez un sujet, comprenant l'administration d'une quantité efficace d'un inhibiteur de MG53 à un sujet en ayant besoin. L'invention concerne en outre un anticorps contre MG53, un acide nucléique codant pour l'anticorps, un clone ou un vecteur d'expression comprenant l'acide nucléique, une cellule hôte comprenant le clone ou le vecteur d'expression, et une composition pharmaceutique comprenant ceux-ci.
PCT/CN2018/111695 2017-11-02 2018-10-24 Procédés et utilisation pour détecter et inhiber mg53 acellulaire WO2019085804A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201711063104.7 2017-11-02
CN201711063104 2017-11-02
CNPCT/CN2018/109871 2018-10-11
CN2018109871 2018-10-11

Publications (1)

Publication Number Publication Date
WO2019085804A1 true WO2019085804A1 (fr) 2019-05-09

Family

ID=66331347

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/111695 WO2019085804A1 (fr) 2017-11-02 2018-10-24 Procédés et utilisation pour détecter et inhiber mg53 acellulaire

Country Status (3)

Country Link
CN (1) CN109932509B (fr)
TW (1) TW201923346A (fr)
WO (1) WO2019085804A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113223104A (zh) * 2021-04-16 2021-08-06 山东师范大学 一种基于因果关系的心脏mr图像插补方法及系统
WO2022056278A1 (fr) * 2020-09-11 2022-03-17 Alexion Pharmaceuticals, Inc. Anticorps anti-céruloplasmine et leurs utilisations

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116003594B (zh) * 2022-12-15 2023-10-13 无锡兴元富澄诊断技术有限公司 靶向cxcl4的抗体及其在抑郁障碍诊断中的用途
CN117402251B (zh) * 2023-12-15 2024-02-23 中国医学科学院基础医学研究所 一种抗小g蛋白rbj的抗体及其应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101912617A (zh) * 2010-07-29 2010-12-15 北京大学 Mg53基因在治疗胰岛素抵抗和ⅱ型糖尿病及其相关病症中的应用
WO2012061793A1 (fr) * 2010-11-05 2012-05-10 Trimedicine, Inc. Mg53 sérique convenant comme marqueur de diagnostic de lésion tissulaire

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105523969A (zh) * 2015-12-30 2016-04-27 北京大学 抑制心肌细胞程序性坏死的CaMKII的抑制剂及用途

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101912617A (zh) * 2010-07-29 2010-12-15 北京大学 Mg53基因在治疗胰岛素抵抗和ⅱ型糖尿病及其相关病症中的应用
WO2012061793A1 (fr) * 2010-11-05 2012-05-10 Trimedicine, Inc. Mg53 sérique convenant comme marqueur de diagnostic de lésion tissulaire

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022056278A1 (fr) * 2020-09-11 2022-03-17 Alexion Pharmaceuticals, Inc. Anticorps anti-céruloplasmine et leurs utilisations
CN113223104A (zh) * 2021-04-16 2021-08-06 山东师范大学 一种基于因果关系的心脏mr图像插补方法及系统

Also Published As

Publication number Publication date
TW201923346A (zh) 2019-06-16
CN109932509B (zh) 2021-08-24
CN109932509A (zh) 2019-06-25

Similar Documents

Publication Publication Date Title
WO2019085804A1 (fr) Procédés et utilisation pour détecter et inhiber mg53 acellulaire
US9599608B2 (en) Antibody against affinity complex
TW201406781A (zh) 以抗bmp9抗體爲有效成分之針對腎性貧血、癌症性貧血等貧血之治療劑
US20220089783A1 (en) Methods of using single domain antibodies directed against ebola virus
JP6900051B2 (ja) Claudin 5抗体、及びその抗体を含有する医薬
JP2022547850A (ja) 抗tigit免疫阻害剤及び応用
KR101589135B1 (ko) 인간화 항-emapii 항체 및 이의 용도
TW201302796A (zh) 抗凝血劑解毒劑
JP6486914B2 (ja) ヒト抗il−32抗体
WO2020239014A1 (fr) Anticorps anti cgrp et son utilisation
KR101159011B1 (ko) 햅텐 화합물 및 항체
WO2020230901A1 (fr) Anticorps bispécifique pouvant se lier à cd40 et gpc3
US9353188B2 (en) Modulators of Plexin B2 activity
US20200024347A1 (en) Or10h1 antigen binding proteins and uses thereof
WO2017211313A1 (fr) Protéine à liaison spécifique et durable à la pcsk9 et son application
WO2017174017A1 (fr) Protéine de liaison de la proprotéine convertase subtilisine kexine de type 9 et son application
US20220403047A1 (en) Antibody specifically recognizing itih1, and pharmaceutical composition containing same for improving insulin resistance in diseases accompanied by impaired glucose tolerance
US20230203142A1 (en) Treatment of metabolic disorders through the targeting of a novel circulating hormone complex
US20190263901A1 (en) Methods of modulating plexin b2 activity
US8497075B2 (en) Methods of identifying a modulator that inhibits the binding between Epstein-Barr virus induced receptor 2 and cholesterol derived ligands
JP2017527262A (ja) ヒト由来抗il−20抗体および抗サイトカイン抗体同定アッセイ

Legal Events

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

Ref document number: 18874761

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18874761

Country of ref document: EP

Kind code of ref document: A1