Injury to the renal proximal tubular epithelium (PTE) represents the underlying cause for acute kidney injury (AKI) following exposure to various stresses including ischemia-reperfusion (I/R) and nephrotoxins. Kidney cells possess intrinsic mechanisms that can be activated to protect their survival under stress conditions. Facilitating repair of PTE cells will have beneficial effects in the treatment of AKI; however, the knowledge gap in understanding the mechanisms associated with repair of injury to these cells has been a setback in the development of novel therapies for AKI. Our group previously identified MG53, a novel member of the TRIM family protein, as an essential component of the cell membrane repair machinery. Although native MG53 protein is predominantly expressed in striated muscles, it is also present in the kidney particularly in PTE cells at a low, but significant concentration. PTE cells derived from the Mg53-/- mice are more vulnerable to membrane injury than normal cells, a phenotype that is also observed in striated muscles, suggesting that MG53 contributes to repair of renal epithelial cells. Compared with wild type mice, the Mg53-/- mice develop renal disorder and are more susceptible to I/R- induced AKI. We found that the recombinant human MG53 (rhMG53) protein can cross the glomeruli and target to injury sites on PTE cells to facilitate the renal protective effect against various stresses. Intravenous administration of rhMG53 could protect I/R and cisplatin-induced injury to the kidney in rodent models. These data suggest that upregulating the MG53-mediated repair mechanism in PTE cells may be beneficial in preventing AKI and treating established AKI. Since native MG53 is present in circulation, systemic administration of rhMG53 would be unlikely to induce an immune response, and potentially be a safe biologic approach for treatment of AKI. The long-term goal of this project is to test the hypothesis that MG53 contributes to maintenance of renal function under physiological conditions, and targeting MG53-mediated repair of injury to PTE cells represents an effective means for prevention and treatment of AKI.
Two specific aims are proposed: a) to elucidate the mechanism for MG53-mediated membrane repair in maintenance of renal function under physiological and AKI conditions; and b) to conduct proof-of-concept studies on rhMG53- based therapy for prevention or treatment of AKI. Fulfillment of these studies should advance our knowledge on the molecular mechanisms that underlie the protection of renal cells from stress-induced injuries, and help development of novel therapies for prevention and treatment of AKI.

Public Health Relevance

Acute kidney injury (AKI) is commonly encountered in the hospital and outpatient settings, and is associated with a high rate of mortality. Treatment and care for AKI require lengthy hospital stays thus incurring high financial costs. Because the mechanisms governing the normal homeostatic repair to renal cells have been unknown, there has been no good therapeutic strategy for treatment of AKI. This project will elucidate the role of a repair molecule in AKI, and conduct proof-of-principle studies for targeting repair of injury to renal cells for prevention and/or treatment of AKI.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK106394-04
Application #
9645630
Study Section
Pathobiology of Kidney Disease Study Section (PBKD)
Program Officer
Sadusky, Anna Burkart
Project Start
2016-05-06
Project End
2021-01-31
Budget Start
2019-02-01
Budget End
2020-01-31
Support Year
4
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Ohio State University
Department
Surgery
Type
Schools of Medicine
DUNS #
832127323
City
Columbus
State
OH
Country
United States
Zip Code
43210
Ogunbayo, Oluseye A; Duan, Jingxian; Xiong, Jian et al. (2018) mTORC1 controls lysosomal Ca2+ release through the two-pore channel TPC2. Sci Signal 11:
Wang, Sheng; Gumpper, Kristyn; Tan, Tao et al. (2018) A novel organ preservation solution with efficient clearance of red blood cells improves kidney transplantation in a canine model. Cell Biosci 8:28
Fan, Zhaobo; Xu, Zhaobin; Niu, Hong et al. (2018) An Injectable Oxygen Release System to Augment Cell Survival and Promote Cardiac Repair Following Myocardial Infarction. Sci Rep 8:1371
Choi, Sangyong; Cui, Chaochu; Luo, Yanhong et al. (2018) Selective inhibitory effects of zinc on cell proliferation in esophageal squamous cell carcinoma through Orai1. FASEB J 32:404-416
Wang, Zhen; Chen, Ken; Han, Yu et al. (2018) Irisin Protects Heart Against Ischemia-Reperfusion Injury Through a SOD2-Dependent Mitochondria Mechanism. J Cardiovasc Pharmacol 72:259-269
Zhang, Caimei; Chen, Biyi; Wang, Yihui et al. (2017) MG53 is dispensable for T-tubule maturation but critical for maintaining T-tubule integrity following cardiac stress. J Mol Cell Cardiol 112:123-130
Lin, Pei-Hui; Sermersheim, Matthew; Li, Haichang et al. (2017) Zinc in Wound Healing Modulation. Nutrients 10:
Chen, Ken; Xu, Zaicheng; Liu, Yukai et al. (2017) Irisin protects mitochondria function during pulmonary ischemia/reperfusion injury. Sci Transl Med 9:
Yue, Tao; Park, Ki Ho; Reese, Benjamin E et al. (2016) Quantifying Drug-Induced Nanomechanics and Mechanical Effects to Single Cardiomyocytes for Optimal Drug Administration To Minimize Cardiotoxicity. Langmuir 32:1909-19
Tan, Tao; Ko, Young-Gyu; Ma, Jianjie (2016) Dual function of MG53 in membrane repair and insulin signaling. BMB Rep 49:414-23

Showing the most recent 10 out of 15 publications