Ischemic injury is at the heart of approximately 50% of all cases of acute renal failure. One hallmark of ischemia is a severe drop in cellular ATP levels, which promotes numerous changes in cellular functions, including broad suppression of transcriptional and translational activity. To survive this consequence of energy depletion, cells must maintain a pool of mRNA transcripts that encode proteins involved in critical cellular processes. Investigation into the role of the RNA-stabilizing protein HuR in an in vitro model of renal ischemic injury demonstrated that it is required for maintaining appropriate expression of select mRNAs during cellular ATP depletion. HuR, which may bind hundreds to thousands of distinct mRNA transcripts, shuttles from the nucleus to the cytoplasm during ischemic stress, where it is capable of preventing degradation of these mRNAs. Further, ATP depletion triggers stress-induced changes in translation of HuR, while recovery of ATP levels triggers new HuR mRNA transcription. These data suggest that HuR may play a large role in promoting kidney cell survival during ischemic injury. Examination of HuR in injured kidney epithelia demonstrates tubule segment-specific regulation of HuR expression and distribution, potentially reflecting different responses to this type of stress. The goal of this project is to discern the mechanisms behind enhanced HuR expression triggered by ischemic injury, and to more precisely define tubule segment-specific regulation of this protein. Because HuR may play a role in stabilizing thousands of critical mRNAs, this work should provide important insight into a critical aspect of cell survival during ischemic stress. Relevance: Loss of blood flow to the kidney is a common cause of renal failure. Whether induced by disease, trauma, or transplantation, this circulatory problem results in cell death and loss of kidney function. The object of the proposed work is to identify some of the mechanisms that kidney cells use to protect themselves from this type of injury, in hopes of creating the ability to intervene in this process and promote cell survival.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
3R01DK052131-14S1
Application #
7916094
Study Section
Pathobiology of Kidney Disease Study Section (PBKD)
Program Officer
Rasooly, Rebekah S
Project Start
2009-09-01
Project End
2011-08-31
Budget Start
2009-09-01
Budget End
2011-08-31
Support Year
14
Fiscal Year
2009
Total Cost
$149,700
Indirect Cost
Name
Ohio State University
Department
Physiology
Type
Schools of Medicine
DUNS #
832127323
City
Columbus
State
OH
Country
United States
Zip Code
43210
Govindaraju, Suman; Lee, Beth S (2014) Krüppel -like factor 8 is a stress-responsive transcription factor that regulates expression of HuR. Cell Physiol Biochem 34:519-32
Singh, Mamata; Martinez, Alaina R; Govindaraju, Suman et al. (2013) HuR inhibits apoptosis by amplifying Akt signaling through a positive feedback loop. J Cell Physiol 228:182-9
Jeyaraj, Selvi C; Singh, Mamata; Ayupova, Dina A et al. (2010) Transcriptional control of human antigen R by bone morphogenetic protein. J Biol Chem 285:4432-40
McMichael, Brooke K; Cheney, Richard E; Lee, Beth S (2010) Myosin X regulates sealing zone patterning in osteoclasts through linkage of podosomes and microtubules. J Biol Chem 285:9506-15
McMichael, Brooke K; Wysolmerski, Robert B; Lee, Beth S (2009) Regulated proteolysis of nonmuscle myosin IIA stimulates osteoclast fusion. J Biol Chem 284:12266-75
McMichael, Brooke K; Lee, Beth S (2008) Tropomyosin 4 regulates adhesion structures and resorptive capacity in osteoclasts. Exp Cell Res 314:564-73
Kotadiya, Preeyal; McMichael, Brooke K; Lee, Beth S (2008) High molecular weight tropomyosins regulate osteoclast cytoskeletal morphology. Bone 43:951-60
McMichael, Brooke K; Kotadiya, Preeyal; Singh, Tejdeep et al. (2006) Tropomyosin isoforms localize to distinct microfilament populations in osteoclasts. Bone 39:694-705
Jeyaraj, Selvi; Dakhlallah, Duaa; Hill, Stephanie R et al. (2005) HuR stabilizes vacuolar H+-translocating ATPase mRNA during cellular energy depletion. J Biol Chem 280:37957-64
Holliday, L S; Lu, M; Lee, B S et al. (2000) The amino-terminal domain of the B subunit of vacuolar H+-ATPase contains a filamentous actin binding site. J Biol Chem 275:32331-7

Showing the most recent 10 out of 11 publications