Approximately 22 millions Americans suffer from chronic kidney disease (CKD), and 300,000 require renal replacement therapy for end stage kidney disease (ESRD). CKD patients are at significantly greater risk of having both fatal and non-fatal cardiovascular complications Renal replacement therapy prolongs the life of patients with ESRD, however, cardiovascular morbidity and mortality remains extremely high in the dialysis population. The propensity patients with kidney disease develop cardiac disease has been linked to a number of factors including heightened sympathetic tone, increased oxidative stress, and widespread arterial calcification. To investigate the pathogenesis of cardiac disease in CKD/ESRD, we hypothesized that the kidney's endocrine function was incompletely understood, that the organ secretes proteins that regulate cardiac function, and that these proteins could be identified using a combination of molecular methods and genetic database analysis. That effort led to the discovery of renalase, a novel FAD-dependent amine oxidase that is expressed in kidney, and heart. It is secreted into blood, and degrades circulating catecholamines. Renalase administration lowers blood pressure, and also protects against acute myocardial ischemia. Blood levels correlate positively with renal function, and patients with CKD and ESRD have low renalase levels. The molecular details of the renalase pathway are beginning to emerge, and we find that catecholamines regulate renalase gene expression, secretion and enzymatic activation. We hypothesize that renalase plays a key role in regulating catechlolamines levels and sympathetic activity, and that renalase deficiency in kidney disease results in heightened sympathetic tone, hypertension and increased cardiovascular risk. In the context of these findings and of the overall hypothesis, we propose to examine how catecholamines and/or blood pressure regulate blood renalase activity in vivo. We will study the mechanisms that mediate hypertension in the renalase knockout mouse. We will use transgenic animals to determine the specific contribution of the kidney to the renalase pathway. These studies will significantly enhance our understanding of the renalase pathway, and will begin to shed light on renalase's role in the pathogenesis of hypertension and cardiovascular complications associated with kidney disease.

Public Health Relevance

Twenty millions Americans suffer from chronic kidney disease, and are at significantly greater risk for having both fatal and non-fatal cardiovascular complications. The pathogenesis of cardiac disease in this patient population is not well understood. Patients with kidney disease have a deficiency in renalase, a newly discovered hormone that is made in the kidney, is secreted in blood, and regulates blood pressure and cardiac function. Renalase itself or components of the renalase pathway may offer valuable therapeutic options for patients with kidney disease.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK081037-02
Application #
7917422
Study Section
Cellular and Molecular Biology of the Kidney Study Section (CMBK)
Program Officer
Ketchum, Christian J
Project Start
2009-08-18
Project End
2012-06-30
Budget Start
2010-07-01
Budget End
2012-06-30
Support Year
2
Fiscal Year
2010
Total Cost
$321,088
Indirect Cost
Name
Yale University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
043207562
City
New Haven
State
CT
Country
United States
Zip Code
06520
Kolodecik, Thomas R; Reed, Anamika M; Date, Kimie et al. (2017) The serum protein renalase reduces injury in experimental pancreatitis. J Biol Chem 292:21047-21059
Guo, Xiaojia; Hollander, Lindsay; MacPherson, Douglas et al. (2016) Inhibition of renalase expression and signaling has antitumor activity in pancreatic cancer. Sci Rep 6:22996
Hollander, Lindsay; Guo, Xiaojia; Velazquez, Heino et al. (2016) Renalase Expression by Melanoma and Tumor-Associated Macrophages Promotes Tumor Growth through a STAT3-Mediated Mechanism. Cancer Res 76:3884-94
Qi, Chaojun; Wang, Ling; Zhang, Minfang et al. (2015) Serum Renalase Levels Correlate with Disease Activity in Lupus Nephritis. PLoS One 10:e0139627
Quelhas-Santos, Janete; SerrĂ£o, Maria Paula; Soares-Silva, Isabel et al. (2015) Renalase regulates peripheral and central dopaminergic activities. Am J Physiol Renal Physiol 308:F84-91
Wang, Ling; Velazquez, Heino; Chang, John et al. (2015) Identification of a receptor for extracellular renalase. PLoS One 10:e0122932
Wang, Ling; Velazquez, Heino; Moeckel, Gilbert et al. (2014) Renalase prevents AKI independent of amine oxidase activity. J Am Soc Nephrol 25:1226-35
Lee, H Thomas; Kim, Joo Yun; Kim, Mihwa et al. (2013) Renalase protects against ischemic AKI. J Am Soc Nephrol 24:445-55
Sizova, Daria; Velazquez, Heino; Sampaio-Maia, Benedita et al. (2013) Renalase regulates renal dopamine and phosphate metabolism. Am J Physiol Renal Physiol 305:F839-44
Desir, Gary V; Tang, Lieqi; Wang, Peili et al. (2012) Renalase lowers ambulatory blood pressure by metabolizing circulating adrenaline. J Am Heart Assoc 1:e002634

Showing the most recent 10 out of 13 publications