Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
STAR Protoc ; 4(1): 101871, 2023 03 17.
Article En | MEDLINE | ID: mdl-36856767

Evaluation of autophagy flux could be challenging for muscle fibers due to the baseline expression of mCherry-EGFP-LC3 along the Z-line. We established a protocol to overcome this difficulty. We overexpress mChery-EGFP-LC3 in the FDB muscle of an adult mouse via electroporation. Then, we enzymatically digest FDB muscle to yield individual fibers for live cell imaging. Finally, we develop an ImageJ-based program to eliminate the baseline striation pattern and semi-automatically quantify autophagosomes (APs) and autolysosomes (ALs) for autophagy flux analysis.


Autophagy , Microtubule-Associated Proteins , Mice , Animals , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Autophagy/genetics , Muscle Fibers, Skeletal/metabolism , Autophagosomes/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Coloring Agents/metabolism
2.
Cells ; 11(10)2022 05 22.
Article En | MEDLINE | ID: mdl-35626749

Identifying effective donor cells is one of obstacles that limits cell therapy for heart disease. In this study, we sorted a subpopulation of human mesenchymal progenitor cells (hMPCs) from the right atrial appendage using the low mitochondrial membrane potential. Compared to the non-sorted cells, hMPCs hold the capacity for stemness and enrich mesenchymal stem cell markers. The hMPCs display better ability for survival, faster proliferation, less production of reactive oxygen species (ROS), and greater release of cytoprotective cytokines. The hMPCs exhibit decreased expression of senescence genes and increased expression of anti-apoptotic and antioxidant genes. Intramyocardial injection of hMPCs into the infarcted heart resulted in increased left ventricular ejection fraction and reduced cardiac remodeling and infarct size in the group of animals receiving hMPCs. Both in vitro and in vivo studies indicated hMPCs have the potential to differentiate into endothelial cells and smooth muscle cells. Immunohistochemistry staining showed that cell therapy with hMPCs enhances cardiac vascular regeneration and cardiac proliferation, and decreases cardiac cell apoptosis, which is associated with the increased secretion of cytoprotective and pro-angiogenic cytokines. Overall, we discovered a subpopulation of human mesenchymal progenitor cells via their low mitochondrial membrane potential, which might provide an alternative donor cell source for cellular therapy for ischemic heart disease.


Endothelial Cells , Mesenchymal Stem Cells , Animals , Cytokines/metabolism , Endothelial Cells/metabolism , Membrane Potential, Mitochondrial , Mesenchymal Stem Cells/metabolism , Neovascularization, Pathologic/metabolism , Stroke Volume , Ventricular Function, Left
3.
J Hepatol ; 76(3): 558-567, 2022 03.
Article En | MEDLINE | ID: mdl-34736969

BACKGROUND & AIMS: Drug-induced liver injury (DILI) remains challenging to treat and is still a leading cause of acute liver failure. MG53 is a muscle-derived tissue-repair protein that circulates in the bloodstream and whose physiological role in protection against DILI has not been examined. METHODS: Recombinant MG53 protein (rhMG53) was administered exogenously, using mice with deletion of Mg53 or Ripk3. Live-cell imaging, histological, biochemical, and molecular studies were used to investigate the mechanisms that underlie the extracellular and intracellular action of rhMG53 in hepatoprotection. RESULTS: Systemic administration of rhMG53 protein, in mice, can prophylactically and therapeutically treat DILI induced through exposure to acetaminophen, tetracycline, concanavalin A, carbon tetrachloride, or thioacetamide. Circulating MG53 protects hepatocytes from injury through direct interaction with MLKL at the plasma membrane. Extracellular MG53 can enter hepatocytes and act as an E3-ligase to mitigate RIPK3-mediated MLKL phosphorylation and membrane translocation. CONCLUSIONS: Our data show that the membrane-delimited signaling and cytosolic dual action of MG53 effectively preserves hepatocyte integrity during DILI. rhMG53 may be a potential treatment option for patients with DILI. LAY SUMMARY: Interventions to treat drug-induced liver injury and halt its progression into liver failure are of great value to society. The present study reveals that muscle-liver cross talk, with MG53 as a messenger, serves an important role in liver cell protection. Thus, MG53 is a potential treatment option for patients with drug-induced liver injury.


Hepatocytes/cytology , Membrane Proteins/metabolism , Protective Agents/metabolism , Animals , Chemical and Drug Induced Liver Injury , Cytosol/metabolism , Disease Models, Animal , Hepatocytes/drug effects , Hepatocytes/physiology , Membrane Proteins/analysis , Membrane Proteins/blood , Mice , Protective Factors
4.
JCI Insight ; 6(17)2021 09 08.
Article En | MEDLINE | ID: mdl-34292883

Aging is associated with chronic oxidative stress and inflammation that affect tissue repair and regeneration capacity. MG53 is a TRIM family protein that facilitates repair of cell membrane injury in a redox-dependent manner. Here, we demonstrate that the expression of MG53 was reduced in failing human hearts and aged mouse hearts, concomitant with elevated NF-κB activation. We evaluated the safety and efficacy of longitudinal, systemic administration of recombinant human MG53 (rhMG53) protein in aged mice. Echocardiography and pressure-volume loop measurements revealed beneficial effects of rhMG53 treatment in improving heart function of aged mice. Biochemical and histological studies demonstrated that the cardioprotective effects of rhMG53 are linked to suppression of NF-κB-mediated inflammation, reducing apoptotic cell death and oxidative stress in the aged heart. Repetitive administration of rhMG53 in aged mice did not have adverse effects on major vital organ functions. These findings support the therapeutic value of rhMG53 in treating age-related decline in cardiac function.


Aging , Gene Expression Regulation , Heart Failure/genetics , Membrane Proteins/genetics , Myocardium/metabolism , NF-kappa B/genetics , Oxidative Stress , Aged , Animals , Apoptosis , Disease Models, Animal , Echocardiography , Female , Heart Failure/diagnosis , Heart Failure/metabolism , Humans , Male , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Middle Aged , Myocardium/pathology , NF-kappa B/biosynthesis , RNA/genetics , Signal Transduction
5.
Cells ; 10(1)2021 01 11.
Article En | MEDLINE | ID: mdl-33440658

Under natural conditions, injured cells can be repaired rapidly through inherent biological processes. However, in the case of diabetes, cardiovascular disease, muscular dystrophy, and other degenerative conditions, the natural repair process is impaired. Repair of injury to the cell membrane is an important aspect of physiology. Inadequate membrane repair function is implicated in the pathophysiology of many human disorders. Recent studies show that Mitsugumin 53 (MG53), a TRIM family protein, plays a key role in repairing cell membrane damage and facilitating tissue regeneration. Clarifying the role of MG53 and its molecular mechanism are important for the application of MG53 in regenerative medicine. In this review, we analyze current research dissecting MG53's function in cell membrane repair and tissue regeneration, and highlight the development of recombinant human MG53 protein as a potential therapeutic agent to repair multiple-organ injuries.


Regenerative Medicine , Tripartite Motif Proteins/metabolism , Animals , Glucose/metabolism , Humans , Phylogeny , Regeneration , Tripartite Motif Proteins/chemistry
6.
Diabetes ; 69(5): 1052-1064, 2020 05.
Article En | MEDLINE | ID: mdl-32139593

MG53 is a member of the TRIM protein family that is predominantly expressed in striated muscles and participates in cell membrane repair. Controversy exists regarding MG53's role in insulin signaling and manifestation of diabetes. We generated db/db mice with either whole-body ablation or sustained elevation of MG53 in the bloodstream in order to evaluate the physiological function of MG53 in diabetes. To quantify the amount of MG53 protein in circulation, we developed a monoclonal antibody against MG53 with high specificity. Western blot using this antibody revealed lower or no change of serum MG53 levels in db/db mice or patients with diabetes compared with control subjects. Neither whole-body ablation of MG53 nor sustained elevation of MG53 in circulation altered insulin signaling and glucose handling in db/db mice. Instead, mice with ablation of MG53 were more susceptible to streptozotocin-induced dysfunctional handling of glucose compared with the wild-type littermates. Alkaline-induced corneal injury demonstrated delayed healing in db/db mice, which was restored by topical administration of recombinant human (rh)MG53. Daily intravenous administration of rhMG53 in rats at concentrations up to 10 mg/kg did not produce adverse effects on glucose handling. These findings challenge the hypothetical function of MG53 as a causative factor for the development of diabetes. Our data suggest that rhMG53 is a potentially safe and effective biologic to treat diabetic oculopathy in rodents.


Blood Glucose , Corneal Injuries/drug therapy , Glucose/metabolism , Insulin Resistance , Membrane Proteins/metabolism , Tripartite Motif Proteins/pharmacology , Animals , Antibodies, Monoclonal , Burns, Chemical/drug therapy , Gene Expression Regulation/physiology , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Rabbits , Recombinant Proteins
...