Muscle wasting is a serious complication of chronic kidney disease (CKD) because it contributes to patient's morbidity and mortality. Muscle wasting mainly reflects increased breakdown of myofibrillar proteins. Compelling evidence has shown that ubiquitin-proteasome system (UPS) is responsible for skeletal muscle protein loss. We have demonstrated that in muscle of CKD mice, depressed IGF-1/PI3 kinase/Akt signaling augments UPS activity via stimulation of the E3 ubiquitin ligases (Atrogin-1, MuRf-1) that affect UPS-mediated muscle proteolysis. Defects in the IGF-1/PI3K/p- Akt pathway stimulate caspase-3 activation and contribute to muscle wasting. We also have discovered that mitochondrial dysfunction and impaired energy metabolism contribute to muscle wasting. In this proposed project, I plan to study ROCK1 as a key molecule linked both to UPS activation and mitochondrial dysfunction. The proposed project is supported by preliminary Results: 1) in muscle of CKD mice, ROCK1 activity is increased and this change is associated with low levels of p-Akt and increased mitochondrial fission; 2) muscle-specific ROCK1 activation stimulates muscle proteolysis and causes mitochondrial dysfunction; and 3) in CKD mice, knockout of ROCK1 ameliorates muscle wasting. We hypothesize that CKD activates ROCK1 leading to both PTEN activation and mitochondria fission, ultimately resulting in muscle wasting. We will accomplish this goal by studying the following three aims Aim 1: To determine how CKD activates ROCK1 leading to depressed PI3K/Akt signaling and enhanced muscle proteolysis.
Aim 2 : To determine an association between CKD and mitochondrial dysfunction and explore how ROCK1 mediates mitochondrial fission.
Aim 3 : To determine if pharmacologic inhibition of ROCK1 and PTEN will reverse the muscle wasting caused by CKD. We propose a new pathway that regulates muscle protein and energy metabolism in CKD. Since this pathway can be manipulated pharmacologically. The results of our study might be used to define cell biologic responses that are interrupted by CKD. The results also might yield the initial steps towards designing new therapies for this dreaded complication of CKD. .

Public Health Relevance

The project is directed at identifying a new mechanism for CKD -induced 'protein-energy wasting' which is frequently associated with increased morbidity and mortality. The PI has engineered transgenic mouse models specifically designed to identify the cause of protein-energy wasting. The pathway PI proposed might be manipulated pharmacologically to prevent or reverse protein-energy wasting in CKD.

Agency
National Institute of Health (NIH)
Institute
National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
Type
Research Project (R01)
Project #
4R01AR063686-04
Application #
9057956
Study Section
Skeletal Muscle Biology and Exercise Physiology Study Section (SMEP)
Program Officer
Boyce, Amanda T
Project Start
2013-07-01
Project End
2018-04-30
Budget Start
2016-05-01
Budget End
2017-04-30
Support Year
4
Fiscal Year
2016
Total Cost
Indirect Cost
Name
Baylor College of Medicine
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
051113330
City
Houston
State
TX
Country
United States
Zip Code
77030
Wang, Yuguo; Jia, Li; Hu, Zhaoyong et al. (2018) AMP-activated protein kinase/myocardin-related transcription factor-A signaling regulates fibroblast activation and renal fibrosis. Kidney Int 93:81-94
Cao, Jin; Yu, Yi; Zhang, Zhengmao et al. (2018) SCP4 Promotes Gluconeogenesis Through FoxO1/3a Dephosphorylation. Diabetes 67:46-57
Peng, Hui; Wang, Qianqian; Lou, Tanqi et al. (2017) Myokine mediated muscle-kidney crosstalk suppresses metabolic reprogramming and fibrosis in damaged kidneys. Nat Commun 8:1493
Yu, Rizhen; Chen, Ji-An; Xu, Jing et al. (2017) Suppression of muscle wasting by the plant-derived compound ursolic acid in a model of chronic kidney disease. J Cachexia Sarcopenia Muscle 8:327-341
Liu, Xinyan; Yu, Rizhen; Sun, Lijing et al. (2017) The nuclear phosphatase SCP4 regulates FoxO transcription factors during muscle wasting in chronic kidney disease. Kidney Int 92:336-348
Zhou, Xiaoshuang; Li, Rongshan; Liu, Xinyan et al. (2016) ROCK1 reduces mitochondrial content and irisin production in muscle suppressing adipocyte browning and impairing insulin sensitivity. Sci Rep 6:29669
Peng, Hui; Li, Yuanqing; Wang, Cheng et al. (2016) ROCK1 Induces Endothelial-to-Mesenchymal Transition in Glomeruli to Aggravate Albuminuria in Diabetic Nephropathy. Sci Rep 6:20304
Peng, Hui; Cao, Jin; Yu, Rizhen et al. (2016) CKD Stimulates Muscle Protein Loss Via Rho-associated Protein Kinase 1 Activation. J Am Soc Nephrol 27:509-19
Lin, Jamie; Shi, Yuanyuan; Peng, Hui et al. (2015) Loss of PTEN promotes podocyte cytoskeletal rearrangement, aggravating diabetic nephropathy. J Pathol 236:30-40
Hu, Zhaoyong; Klein, Janet D; Mitch, William E et al. (2014) MicroRNA-29 induces cellular senescence in aging muscle through multiple signaling pathways. Aging (Albany NY) 6:160-75

Showing the most recent 10 out of 11 publications