Heme oxygenase (HO) converts heme to biliverdin during which iron is released and carbon monoxide (CO) is emitted;biliverdin reductase subsequently converts biliverdin to bilirubin. In 1992, the Pi'slaboratory provided the first evidence that HO-1 was cytoprotective, a finding derived in acute renal heme-mediated injury, and which was the basis for the 1993-1997 funding cycle;the 1997-2002 cycle was based on the finding by the Pi's laboratory that HO-1 was cytoprotective in other forms of renal injury. The 2002-2007 cycle sought to identify pathogenetic pathways interrupted by HO-1, and demonstrated, along with relevant mechanistic insights, that the HO system inhibits: i) vasoconstriction/ischemia, li) inflammation, and iii) apoptosis. The proposed aims continue these investigative themes.
Aim I will delineate the role of the HO system in militating against vasoconstriction by examining the systemic and renal hemodynamic effects of Ang ll-dependent and Ang ll-independent vasoconstriction (theDOCA model) in HO-1""""""""'"""""""" and HO^"""""""" mice.
This aim will examine such mechanisms as NADPH oxidase, superoxide anion generation, BH4/BH2 levels and coupling of eNOS, and the role of endothelin-1;
this aim will also determine the extent to which products of HO can reverse the enhanced vasoconstriction when HO-1 or HO-2 is deficient.
Aim II will determine the basis for the anti-inflammatory effect of the HO system against LPS-induced_inflammation, targeting activation of NF-KB as a critical locus for the anti-inflammatory effects of HO. These studies will determine the following: the extent to which HO-1 regulates inflammatory responses by altering activation of NF-KB;the contribution of specific NF-KB-dependent cytokines (MCP-1, IL-6,IL-12(p40), and TNF) to the exaggerated inflammation due to HO-1 deficiency;the anti-inflammatory capacity of specific HO products and whether such products influence NF-KB activation;and finally, the capacity of HO-1 overexpression to inhibit LPS- driven inflammation.
Aim III will examine the basis for apoptosis, acute renal injury, and increased mortality in HO-1""""""""'"""""""" mice, subjected to renal ischemia, focusing on IL-6 and its signaling species, pSTAT3rthe .latter now identified as proapoptotic in the kidney. IL-6is markedly and uniquely induced in the kidney, Iung7 arid heart, and increased in the serum. In this model of apoptosis, the effect of inhibiting IL-6/pSTAT3 will Jbe determined.
This aim will also determine th$ source of IL-6 in the kidney and systemic circulation, arid the regulation of IL-6expression by HO-1.
This aim will thus determine the contribution of IL-6^'STAT-3^ apoptosis and other adverse effects of ischemia in the absence of HO-1, and the reciprocating effects between cellular expression and signaling of IL-6and HO-1. Lav Summary. This application seeks to understand how a protein called heme oxygenase-1 protects the kidney against different types of diseases. This understanding may facilitate the development of new therapies for kidney disease.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Method to Extend Research in Time (MERIT) Award (R37)
Project #
3R37DK047060-18S1
Application #
7903739
Study Section
Special Emphasis Panel (NSS)
Program Officer
Kimmel, Paul
Project Start
2009-09-22
Project End
2011-08-31
Budget Start
2009-09-22
Budget End
2011-08-31
Support Year
18
Fiscal Year
2009
Total Cost
$99,554
Indirect Cost
Name
Mayo Clinic, Rochester
Department
Type
DUNS #
006471700
City
Rochester
State
MN
Country
United States
Zip Code
55905
Nath, Karl A; Katusic, Zvonimir S (2017) Endothelin-A Receptor Antagonism Retards the Progression of Murine Sickle Cell Nephropathy. J Am Soc Nephrol 28:2253-2255
Kang, Lu; Grande, Joseph P; Hillestad, Matthew L et al. (2016) A new model of an arteriovenous fistula in chronic kidney disease in the mouse: beneficial effects of upregulated heme oxygenase-1. Am J Physiol Renal Physiol 310:F466-76
Nath, Karl A (2015) Models of Human AKI: Resemblance, Reproducibility, and Return on Investment. J Am Soc Nephrol 26:2891-3
Kang, Lu; Hillestad, Matthew L; Grande, Joseph P et al. (2015) Induction and functional significance of the heme oxygenase system in pathological shear stress in vivo. Am J Physiol Heart Circ Physiol 308:H1402-13
Belcher, John D; Chen, Chunsheng; Nguyen, Julia et al. (2014) Heme triggers TLR4 signaling leading to endothelial cell activation and vaso-occlusion in murine sickle cell disease. Blood 123:377-90
Nath, Karl A (2014) Heme oxygenase-1 and acute kidney injury. Curr Opin Nephrol Hypertens 23:17-24
Kang, Lu; Grande, Joseph P; Farrugia, Gianrico et al. (2013) Functioning of an arteriovenous fistula requires heme oxygenase-2. Am J Physiol Renal Physiol 305:F545-52
Nath, Karl A; Grande, Joseph P; Farrugia, Gianrico et al. (2013) Age sensitizes the kidney to heme protein-induced acute kidney injury. Am J Physiol Renal Physiol 304:F317-25
Abudiab, Muaz; Krause, Megan L; Fidler, Mary E et al. (2013) Differentiating scleroderma renal crisis from other causes of thrombotic microangiopathy in a postpartum patient. Clin Nephrol 80:293-7
Nath, Karl A (2013) The role of renal research in demonstrating the protective properties of heme oxygenase-1. Kidney Int 84:3-6

Showing the most recent 10 out of 47 publications