Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Nuclear hormone receptors in diabetic nephropathy

Abstract

Diabetes is the leading cause of end-stage renal disease in developed countries. In spite of glucose and blood pressure control, for example by use of angiotensin II receptor blockers, diabetic nephropathy still develops and progresses in affected patients and the development of additional protective therapeutic interventions is, therefore, required. Nuclear hormone receptors are transcription factors that regulate carbohydrate metabolism, lipid metabolism, the immune response, and inflammation. These receptors also modulate the development of fibrosis. As a result of their diverse biological effects, nuclear hormone receptors have become major pharmaceutical targets for the treatment of a host of diseases. The increasing prevalence of diabetic nephropathy has led intense investigation into the role that nuclear hormone receptors may have in slowing or preventing the progression of renal disease. This role of nuclear hormone receptors would be associated with improvements in metabolism, the immune response, and inflammation. Eight nuclear receptors have shown a renoprotective effect in the context of diabetic nephropathy. This Review discusses the evidence regarding the beneficial effects of the activation of these receptors in preventing the progression of diabetic nephropathy and describes how the discovery and development of compounds that modulate the activity of nuclear hormone receptors may provide potential additional therapeutic approaches in the management of diabetic nephropathy.

Key Points

  • Nuclear hormone receptor activity is abnormal in the diabetic kidney

  • Nuclear hormone receptors regulate metabolism, inflammation, oxidative stress and fibrosis

  • Modulation of the activity of the peroxisome-proliferator-associated receptor, vitamin D receptor, farnesoid X receptor and estrogen receptor holds great promise in the treatment of diabetic nephropathy

  • Further evidence on the role of nuclear hormone receptors in the diabetic kidney and the identification of additional specific ligands of these receptors will probably result in development of additional therapeutic strategies for diabetic nephropathy

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Nuclear hormone receptors in diabetic nephropathy.

Similar content being viewed by others

References

  1. Mauer, S. M. Structural–functional correlations of diabetic nephropathy. Kidney Int. 45, 612–622 (1994).

    Article  CAS  PubMed  Google Scholar 

  2. Qian, Y., Feldman, E., Pennathur, S., Kretzler, M. & Brosius, F. C. 3rd. From fibrosis to sclerosis: mechanisms of glomerulosclerosis in diabetic nephropathy. Diabetes 57, 1439–1445 (2008).

    Article  CAS  PubMed  Google Scholar 

  3. Forbes, J. M., Coughlan, M. T. & Cooper, M. E. Oxidative stress as a major culprit in kidney disease in diabetes. Diabetes 57, 1446–1454 (2008).

    Article  CAS  PubMed  Google Scholar 

  4. Zhu, Y., Usui, H. K. & Sharma, K. Regulation of transforming growth factor beta in diabetic nephropathy: implications for treatment. Semin. Nephrol. 27, 153–160 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gurley, S. B. & Coffman, T. M. The renin–angiotensin system and diabetic nephropathy. Semin. Nephrol. 27, 144–152 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Weinberg, J. M. Lipotoxicity. Kidney Int. 70, 1560–1566 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Kimmelstiel, P. & Wilson, C. Intercapillary lesions in the glomeruli of the kidney. Am. J. Pathol. 12, 83–98.7 (1936).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Wilens, S. L. & Elster, S. K. The role of lipid deposition in renal arteriolar sclerosis. Am. J. Med. Sci. 219, 183–196 (1950).

    Article  CAS  PubMed  Google Scholar 

  9. Sun, L., Halaihel, N., Zhang, W., Rogers, T. & Levi, M. Role of sterol regulatory element-binding protein 1 in regulation of renal lipid metabolism and glomerulosclerosis in diabetes mellitus. J. Biol. Chem. 277, 18919–18927 (2002).

    Article  CAS  PubMed  Google Scholar 

  10. Proctor, G. et al. Regulation of renal fatty acid and cholesterol metabolism, inflammation, and fibrosis in Akita and OVE26 mice with type 1 diabetes. Diabetes 55, 2502–2509 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Jiang, T. et al. Diet-induced obesity in C57BL/6J mice causes increased renal lipid accumulation and glomerulosclerosis via a sterol regulatory element-binding protein-1c-dependent pathway. J. Biol. Chem. 280, 32317–32325 (2005).

    Article  CAS  PubMed  Google Scholar 

  12. Wang, Z. et al. Regulation of renal lipid metabolism, lipid accumulation, and glomerulosclerosis in FVBdb/db mice with type 2 diabetes. Diabetes 54, 2328–2335 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Fried, L. F., Orchard, T. J. & Kasiske, B. L. Effect of lipid reduction on the progression of renal disease: a meta-analysis. Kidney Int. 59, 260–269 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Levi, M. Do statins have a beneficial effect on the kidney? Nat. Clin. Pract. Nephrol. 2, 666–667 (2006).

    Article  PubMed  Google Scholar 

  15. Evans, R. M. The steroid and thyroid hormone receptor superfamily. Science 240, 889–895 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Mangelsdorf, D. J. et al. The nuclear receptor superfamily: the second decade. Cell 83, 835–839 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bookout, A. L. et al. Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network. Cell 126, 789–799 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Guan, Y. & Breyer, M. D. Peroxisome proliferator-activated receptors (PPARs): novel therapeutic targets in renal disease. Kidney Int. 60, 14–30 (2001).

    Article  CAS  PubMed  Google Scholar 

  19. Jiang, T. et al. Farnesoid X receptor modulates renal lipid metabolism, fibrosis, and diabetic nephropathy. Diabetes 56, 2485–2493 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Niehof, M. & Borlak, J. HNF4 alpha and the Ca-channel TRPC1 are novel disease candidate genes in diabetic nephropathy. Diabetes 57, 1069–1077 (2008).

    Article  CAS  PubMed  Google Scholar 

  21. Chawla, A., Repa, J. J., Evans, R. M. & Mangelsdorf, D. J. Nuclear receptors and lipid physiology: opening the X-files. Science 294, 1866–1870 (2001).

    Article  CAS  PubMed  Google Scholar 

  22. Issemann, I. & Green, S. Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature 347, 645–650 (1990).

    Article  CAS  PubMed  Google Scholar 

  23. Skogsberg, J. et al. Characterization of the human peroxisome proliferator activated receptor delta gene and its expression. Int. J. Mol. Med. 6, 73–81 (2000).

    CAS  PubMed  Google Scholar 

  24. Forman, B. M., Chen, J. & Evans, R. M. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta. Proc. Natl Acad. Sci. USA 94, 4312–4317 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kliewer, S. A. et al. A prostaglandin J2 metabolite binds peroxisome proliferator-activated receptor gamma and promotes adipocyte differentiation. Cell 83, 813–819 (1995).

    Article  CAS  PubMed  Google Scholar 

  26. Willson, T. M., Lehmann, J. M. & Kliewer, S. A. Discovery of ligands for the nuclear peroxisome proliferator-activated receptors. Ann. NY Acad. Sci. 804, 276–283 (1996).

    Article  CAS  PubMed  Google Scholar 

  27. Schopfer, F. J. et al. Nitrolinoleic acid: an endogenous peroxisome proliferator-activated receptor gamma ligand. Proc. Natl Acad. Sci. USA 102, 2340–2345 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lefebvre, P., Chinetti, G., Fruchart, J. C. & Staels, B. Sorting out the roles of PPAR alpha in energy metabolism and vascular homeostasis. J. Clin. Invest. 116, 571–580 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Balakumar, P., Arora, M. K. & Singh, M. Emerging role of PPAR ligands in the management of diabetic nephropathy. Pharmacol. Res. 60, 170–173 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Delerive, P., Fruchart, J. C. & Staels, B. Peroxisome proliferator-activated receptors in inflammation control. J. Endocrinol. 169, 453–459 (2001).

    Article  CAS  PubMed  Google Scholar 

  31. Ouali, F., Djouadi, F., Merlet-Bénichou, C. & Bastin, J. Dietary lipids regulate beta-oxidation enzyme gene expression in the developing rat kidney. Am. J. Physiol. 275, F777–F784 (1998).

    CAS  PubMed  Google Scholar 

  32. Shin, S. J. et al. Peroxisome proliferator-activated receptor-alpha activator fenofibrate prevents high-fat diet-induced renal lipotoxicity in spontaneously hypertensive rats. Hypertens. Res. 32, 835–845 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Portilla, D. et al. Etomoxir-induced PPARalpha-modulated enzymes protect during acute renal failure. Am. J. Physiol. Renal Physiol. 278, F667–F675 (2000).

    Article  CAS  PubMed  Google Scholar 

  34. Li, S. et al. Transgenic expression of proximal tubule peroxisome proliferator-activated receptor-alpha in mice confers protection during acute kidney injury. Kidney Int. 76, 1049–1062 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Roman, R. J., Ma, Y. H., Frohlich, B. & Markham, B. Clofibrate prevents the development of hypertension in Dahl salt-sensitive rats. Hypertension 21, 985–988 (1993).

    Article  CAS  PubMed  Google Scholar 

  36. Guan, Y., Zhang, Y., Davis, L. & Breyer, M. D. Expression of peroxisome proliferator-activated receptors in urinary tract of rabbits and humans. Am. J. Physiol. 273, F1013–F1022 (1997).

    CAS  PubMed  Google Scholar 

  37. Kono, K. et al. PPAR{alpha} attenuates the proinflammatory response in activated mesangial cells. Am. J. Physiol. Renal Physiol. 296, F328–F336 (2009).

    Article  CAS  PubMed  Google Scholar 

  38. Kamijo, Y. et al. Peroxisome proliferator-activated receptor alpha protects against glomerulonephritis induced by long-term exposure to the plasticizer di-(2-ethylhexyl)phthalate. J. Am. Soc. Nephrol. 18, 176–188 (2007).

    Article  CAS  PubMed  Google Scholar 

  39. Park, C. W. et al. Accelerated diabetic nephropathy in mice lacking the peroxisome proliferator-activated receptor alpha. Diabetes 55, 885–893 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Park, C. W. et al. PPARalpha agonist fenofibrate improves diabetic nephropathy in db/db mice. Kidney Int. 69, 1511–1517 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Calkin, A. C. et al. PPAR-alpha and -gamma agonists attenuate diabetic kidney disease in the apolipoprotein E knockout mouse. Nephrol. Dial. Transplant. 21, 2399–2405 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Chen, Y. J. & Quilley, J. Fenofibrate treatment of diabetic rats reduces nitrosative stress, renal cyclooxygenase-2 expression, and enhanced renal prostaglandin release. J. Pharmacol. Exp. Ther. 324, 658–663 (2008).

    Article  CAS  PubMed  Google Scholar 

  43. Balakumar, P., Chakkarwar, V. A. & Singh, M. Ameliorative effect of combination of benfotiamine and fenofibrate in diabetes-induced vascular endothelial dysfunction and nephropathy in the rat. Mol. Cell. Biochem. 320, 149–162 (2009).

    Article  CAS  PubMed  Google Scholar 

  44. Nagai, T., Tomizawa, T., Nakajima, K. & Mori, M. Effect of bezafibrate or pravastatin on serum lipid levels and albuminuria in NIDDM patients. J. Atheroscler. Thromb. 7, 91–96 (2000).

    Article  CAS  PubMed  Google Scholar 

  45. Sacks, F. M. After the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) study: implications for fenofibrate. Am. J. Cardiol. 102, 34L–40L (2008).

    Article  CAS  PubMed  Google Scholar 

  46. Gonzalez, F. J. & Shah, Y. M. PPARα: mechanism of species differences and hepatocarcinogenesis of peroxisome proliferators. Toxicology 246, 2–8 (2008).

    Article  CAS  PubMed  Google Scholar 

  47. Ruan, X. Z. et al. PPAR agonists protect mesangial cells from interleukin 1beta-induced intracellular lipid accumulation by activating the ABCA1 cholesterol efflux pathway. J. Am. Soc. Nephrol. 14, 593–600 (2003).

    Article  CAS  PubMed  Google Scholar 

  48. Rigamonti, E., Chinetti-Gbaguidi, G. & Staels, B. Regulation of macrophage functions by PPAR-alpha, PPAR-gamma, and LXRs in mice and men. Arterioscler. Thromb. Vasc. Biol. 28, 1050–1059 (2008).

    Article  CAS  PubMed  Google Scholar 

  49. Ruan, X., Zheng, F. & Guan, Y. PPARs and the kidney in metabolic syndrome. Am. J. Physiol. Renal Physiol. 294, F1032–F1047 (2008).

    Article  CAS  PubMed  Google Scholar 

  50. Yoshioka, S. et al. Antihypertensive effects of CS-045 treatment in obese Zucker rats. Metabolism 42, 75–80 (1993).

    Article  CAS  PubMed  Google Scholar 

  51. Sarafidis, P. A. & Bakris, G. L. Protection of the kidney by thiazolidinediones: an assessment from bench to bedside. Kidney Int. 70, 1223–1233 (2006).

    Article  CAS  PubMed  Google Scholar 

  52. Guan, Y. et al. Thiazolidinediones expand body fluid volume through PPARgamma stimulation of ENaC-mediated renal salt absorption. Nat. Med. 11, 861–866 (2005).

    Article  CAS  PubMed  Google Scholar 

  53. Carmona, M. C. et al. Fenofibrate prevents Rosiglitazone-induced body weight gain in ob/ob mice. Int. J. Obes. (Lond.) 29, 864–871 (2005).

    Article  CAS  Google Scholar 

  54. Cha, D. R. et al. Peroxisome proliferator activated receptor alpha/gamma dual agonist tesaglitazar attenuates diabetic nephropathy in db/db mice. Diabetes 56, 2036–2045 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Wu, J. et al. Liver X receptor-alpha mediates cholesterol efflux in glomerular mesangial cells. Am. J. Physiol. Renal Physiol. 287, F886–F895 (2004).

    Article  CAS  PubMed  Google Scholar 

  56. Odegaard, J. I. et al. Alternative M2 activation of Kupffer cells by PPARdelta ameliorates obesity-induced insulin resistance. Cell. Metab. 7, 496–507 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Kang, K. et al. Adipocyte-derived Th2 cytokines and myeloid PPARdelta regulate macrophage polarization and insulin sensitivity. Cell. Metab. 7, 485–495 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Kelly, K. J., Wu, P., Patterson, C. E., Temm, C. & Dominguez, J. H. LOX-1 and inflammation: a new mechanism for renal injury in obesity and diabetes. Am. J. Physiol. Renal Physiol. 294, F1136–F1145 (2008).

    Article  CAS  PubMed  Google Scholar 

  59. Letavernier, E. et al. Peroxisome proliferator-activated receptor beta/delta exerts a strong protection from ischemic acute renal failure. J. Am. Soc. Nephrol. 16, 2395–2402 (2005).

    Article  CAS  PubMed  Google Scholar 

  60. Toft, D. & Gorski, J. A receptor molecule for estrogens: isolation from the rat uterus and preliminary characterization. Proc. Natl Acad. Sci. USA 55, 1574–1581 (1966).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Lubahn, D. B. et al. Alteration of reproductive function but not prenatal sexual development after insertional disruption of the mouse estrogen receptor gene. Proc. Natl Acad. Sci. USA 90, 11162–11166 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Barkhem, T. et al. Differential response of estrogen receptor alpha and estrogen receptor beta to partial estrogen agonists/antagonists. Mol. Pharmacol. 54, 105–112 (1998).

    Article  CAS  PubMed  Google Scholar 

  63. Matthews, J. & Gustafsson, J. A. Estrogen signaling: a subtle balance between ER alpha and ER beta. Mol. Interv. 3, 281–292 (2003).

    Article  CAS  PubMed  Google Scholar 

  64. Neugarten, J., Acharya, A., Lei, J. & Silbiger, S. Selective estrogen receptor modulators suppress mesangial cell collagen synthesis. Am. J. Physiol. Renal Physiol. 279, F309–F318 (2000).

    Article  CAS  PubMed  Google Scholar 

  65. Potier, M. et al. Estrogen-related abnormalities in glomerulosclerosis-prone mice: reduced mesangial cell estrogen receptor expression and prosclerotic response to estrogens. Am. J. Pathol. 160, 1877–1885 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Bhat, H. K., Hacker, H. J., Bannasch, P., Thompson, E. A. & Liehr, J. G. Localization of estrogen receptors in interstitial cells of hamster kidney and in estradiol-induced renal tumors as evidence of the mesenchymal origin of this neoplasm. Cancer Res. 53, 5447–5451 (1993).

    CAS  PubMed  Google Scholar 

  67. Jelinsky, S. A. et al. Global transcription profiling of estrogen activity: estrogen receptor alpha regulates gene expression in the kidney. Endocrinology 144, 701–710 (2003).

    Article  CAS  PubMed  Google Scholar 

  68. Pettersson, K., Delaunay, F. & Gustafsson, J. A. Estrogen receptor beta acts as a dominant regulator of estrogen signaling. Oncogene 19, 4970–4978 (2000).

    Article  CAS  PubMed  Google Scholar 

  69. Levin, E. R. Plasma membrane estrogen receptors. Trends Endocrinol. Metab. 20, 477–482 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Maric, C. & Sullivan, S. Estrogens and the diabetic kidney. Gend. Med. 5 (Suppl. A), S103–S113 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Seliger, S. L., Davis, C. & Stehman-Breen, C. Gender and the progression of renal disease. Curr. Opin. Nephrol. Hypertens. 10, 219–225 (2001).

    Article  CAS  PubMed  Google Scholar 

  72. Neugarten, J., Acharya, A. & Silbiger, S. R. Effect of gender on the progression of nondiabetic renal disease: a meta-analysis. J. Am. Soc. Nephrol. 11, 319–329 (2000).

    CAS  PubMed  Google Scholar 

  73. Xiao, S., Gillespie, D. G., Baylis, C., Jackson, E. K. & Dubey, R. K. Effects of estradiol and its metabolites on glomerular endothelial nitric oxide synthesis and mesangial cell growth. Hypertension 37, 645–650 (2001).

    Article  CAS  PubMed  Google Scholar 

  74. Catanuto, P. et al. 17 Beta-estradiol and tamoxifen upregulate estrogen receptor beta expression and control podocyte signaling pathways in a model of type 2 diabetes. Kidney Int. 75, 1194–1201 (2009).

    Article  CAS  PubMed  Google Scholar 

  75. Holick, M. F. Vitamin D and bone health. J. Nutr. 126 (4 Suppl.), 1159S–1164S (1996).

    Article  CAS  PubMed  Google Scholar 

  76. Haussler, M. R. et al. Vitamin D receptor: molecular signaling and actions of nutritional ligands in disease prevention. Nutr. Rev. 66 (Suppl. 2), S98–S112 (2008).

    Article  PubMed  Google Scholar 

  77. Ordóñez-Morán, P. & Muñoz, A. Nuclear receptors: genomic and non-genomic effects converge. Cell Cycle 8, 1675–1680 (2009).

    Article  PubMed  Google Scholar 

  78. Tan, X., Wen, X. & Liu, Y. Paricalcitol inhibits renal inflammation by promoting vitamin D receptor-mediated sequestration of NF-kappaB signaling. J. Am. Soc. Nephrol. 19, 1741–1752 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Kumar, R., Schaefer, J., Grande, J. P. & Roche, P. C. Immunolocalization of calcitriol receptor, 24-hydroxylase cytochrome P-450, and calbindin D28k in human kidney. Am. J. Physiol. 266, F477–F485 (1994).

    CAS  PubMed  Google Scholar 

  80. Zhang, Z. et al. 1,25-Dihydroxyvitamin D3 targeting of NF-kappaB suppresses high glucose-induced MCP-1 expression in mesangial cells. Kidney Int. 72, 193–201 (2007).

    Article  CAS  PubMed  Google Scholar 

  81. Wang, Y. et al. Altered vitamin D metabolism in type II diabetic mouse glomeruli may provide protection from diabetic nephropathy. Kidney Int. 70, 882–891 (2006).

    Article  CAS  PubMed  Google Scholar 

  82. Zipitis, C. S. & Akobeng, A. K. Vitamin D supplementation in early childhood and risk of type 1 diabetes: a systematic review and meta-analysis. Arch. Dis. Child. 93, 512–517 (2008).

    Article  CAS  PubMed  Google Scholar 

  83. Pittas, A. G., Lau, J., Hu, F. B. & Dawson-Hughes, B. The role of vitamin D and calcium in type 2 diabetes. A systematic review and meta-analysis. J. Clin. Endocrinol. Metab. 92, 2017–2029 (2007).

    Article  CAS  PubMed  Google Scholar 

  84. Levin, A. et al. Prevalence of abnormal serum vitamin D, PTH, calcium, and phosphorus in patients with chronic kidney disease: results of the study to evaluate early kidney disease. Kidney Int. 71, 31–38 (2007).

    Article  CAS  PubMed  Google Scholar 

  85. Teng, M. et al. Activated injectable vitamin D and hemodialysis survival: a historical cohort study. J. Am. Soc. Nephrol. 16, 1115–1125 (2005).

    Article  CAS  PubMed  Google Scholar 

  86. Zhang, Z. et al. Renoprotective role of the vitamin D receptor in diabetic nephropathy. Kidney Int. 73, 163–171 (2008).

    Article  CAS  PubMed  Google Scholar 

  87. Zhang, Z. et al. Combination therapy with AT1 blocker and vitamin D analog markedly ameliorates diabetic nephropathy: blockade of compensatory renin increase. Proc. Natl Acad. Sci. USA 105, 15896–15901 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Zhang, Y. et al. Long-term therapeutic effect of vitamin D analog doxercalciferol on diabetic nephropathy: strong synergism with AT1 receptor antagonist. Am. J. Physiol. Renal Physiol. 297, F791–F801 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Deb, D. K. et al. 1,25-Dihydroxyvitamin D3 suppresses high glucose-induced angiotensinogen expression in kidney cells by blocking the NF-{kappa}B pathway. Am. J. Physiol. Renal Physiol. 296, F1212–F1218 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Lambers Heerspink, H. J. et al. The selective vitamin D receptor activator for albuminuria lowering (VITAL) study: study design and baseline characteristics. Am. J. Nephrol. 30, 280–286 (2009).

    Article  CAS  PubMed  Google Scholar 

  91. Katona, B. W. et al. Synthesis, characterization, and receptor interaction profiles of enantiomeric bile acids. J. Med. Chem. 50, 6048–6058 (2007).

    Article  CAS  PubMed  Google Scholar 

  92. Staudinger, J. L. et al. The nuclear receptor PXR is a lithocholic acid sensor that protects against liver toxicity. Proc. Natl Acad. Sci. USA 98, 3369–3374 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Xie, W. et al. An essential role for nuclear receptors SXR/PXR in detoxification of cholestatic bile acids. Proc. Natl Acad. Sci. USA 98, 3375–3380 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Makishima, M. et al. Vitamin D receptor as an intestinal bile acid sensor. Science 296, 1313–1316 (2002).

    Article  CAS  PubMed  Google Scholar 

  95. Gilad, L. A. & Schwartz, B. Association of estrogen receptor beta with plasma-membrane caveola components: implication in control of vitamin D receptor. J. Mol. Endocrinol. 38, 603–618 (2007).

    Article  CAS  PubMed  Google Scholar 

  96. Sladek, F. M., Zhong, W. M., Lai, E. & Darnell, J. E. Liver-enriched transcription factor HNF-4 is a novel member of the steroid hormone receptor superfamily. Genes Dev. 4, 2353–2365 (1990).

    Article  CAS  PubMed  Google Scholar 

  97. Hertz, R., Magenheim, J., Berman, I. & Bar-Tana, J. Fatty acyl-CoA thioesters are ligands of hepatic nuclear factor-4alpha. Nature 392, 512–516 (1998).

    Article  CAS  PubMed  Google Scholar 

  98. Soutoglou, E., Katrakili, N. & Talianidis, I. Acetylation regulates transcription factor activity at multiple levels. Mol. Cell 5, 745–751 (2000).

    Article  CAS  PubMed  Google Scholar 

  99. Jiang, G., Nepomuceno, L., Yang, Q. & Sladek, F. M. Serine/threonine phosphorylation of orphan receptor hepatocyte nuclear factor 4. Arch. Biochem. Biophys. 340, 1–9 (1997).

    Article  CAS  PubMed  Google Scholar 

  100. Viollet, B., Kahn, A. & Raymondjean, M. Protein kinase A-dependent phosphorylation modulates DNA-binding activity of hepatocyte nuclear factor 4. Mol. Cell Biol. 17, 4208–4219 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Chou, W. C. et al. Mechanism of a transcriptional cross talk between transforming growth factor-beta-regulated Smad3 and Smad4 proteins and orphan nuclear receptor hepatocyte nuclear factor-4. Mol. Biol. Cell 14, 1279–1294 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Sladek, F. M. & Seidel, S. D. in Nuclear Receptors and Disease (eds Burris, T. & McCabe, E. R. B.) 309–361 (Academic Press, London, 2001).

    Google Scholar 

  103. Schrem, H., Klempnauer, J. & Borlak, J. Liver-enriched transcription factors in liver function and development. Part I: the hepatocyte nuclear factor network and liver-specific gene expression. Pharmacol. Rev. 54, 129–158 (2002).

    Article  CAS  PubMed  Google Scholar 

  104. Ryffel, G. U. Mutations in the human genes encoding the transcription factors of the hepatocyte nuclear factor (HNF)1 and HNF4 families: functional and pathological consequences. J. Mol. Endocrinol. 27, 11–29 (2001).

    Article  CAS  PubMed  Google Scholar 

  105. Mohlke, K. L. & Boehnke, M. The role of HNF4A variants in the risk of type 2 diabetes. Curr. Diab. Rep. 5, 149–156 (2005).

    Article  CAS  PubMed  Google Scholar 

  106. Du, J. et al. Canonical transient receptor potential 1 channel is involved in contractile function of glomerular mesangial cells. J. Am. Soc. Nephrol. 18, 1437–1445 (2007).

    Article  CAS  PubMed  Google Scholar 

  107. Abramowitz, J. & Birnbaumer, L. Physiology and pathophysiology of canonical transient receptor potential channels. FASEB J. 23, 297–328 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Forman, B. M. et al. Identification of a nuclear receptor that is activated by farnesol metabolites. Cell 81, 687–693 (1995).

    Article  CAS  PubMed  Google Scholar 

  109. Seol, W., Choi, H. S. & Moore, D. D. Isolation of proteins that interact specifically with the retinoid X receptor: two novel orphan receptors. Mol. Endocrinol. 9, 72–85 (1995).

    CAS  PubMed  Google Scholar 

  110. Makishima, M. et al. Identification of a nuclear receptor for bile acids. Science 284, 1362–1365 (1999).

    Article  CAS  PubMed  Google Scholar 

  111. Parks, D. J. et al. Bile acids: natural ligands for an orphan nuclear receptor. Science 284, 1365–1368 (1999).

    Article  CAS  PubMed  Google Scholar 

  112. Wang, H., Chen, J., Hollister, K., Sowers, L. C. & Forman, B. M. Endogenous bile acids are ligands for the nuclear receptor FXR/BAR. Mol. Cell 3, 543–553 (1999).

    Article  CAS  PubMed  Google Scholar 

  113. Claudel, T. et al. Bile acid-activated nuclear receptor FXR suppresses apolipoprotein A-I transcription via a negative FXR response element. J. Clin. Invest. 109, 961–971 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Thomas, C., Pellicciari, R., Pruzanski, M., Auwerx, J. & Schoonjans, K. Targeting bile-acid signalling for metabolic diseases. Nat. Rev. Drug Discov. 7, 678–693 (2008).

    Article  CAS  PubMed  Google Scholar 

  115. Lefebvre, P., Cariou, B., Lien, F., Kuipers, F. & Staels, B. Role of bile acids and bile acid receptors in metabolic regulation. Physiol. Rev. 89, 147–191 (2009).

    Article  CAS  PubMed  Google Scholar 

  116. Fiorucci, S. et al. The nuclear receptor SHP mediates inhibition of hepatic stellate cells by FXR and protects against liver fibrosis. Gastroenterology 127, 1497–1512 (2004).

    Article  CAS  PubMed  Google Scholar 

  117. Li, Y. T., Swales, K. E., Thomas, G. J., Warner, T. D. & Bishop-Bailey, D. Farnesoid X receptor ligands inhibit vascular smooth muscle cell inflammation and migration. Arterioscler. Thromb. Vasc. Biol. 27, 2606–2611 (2007).

    Article  PubMed  CAS  Google Scholar 

  118. Hartman, H. B. et al. Activation of farnesoid X receptor prevents atherosclerotic lesion formation in LDLR−/− and apoE−/− mice. J. Lipid Res. 50, 1090–1100 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Wang, X. X. et al. The farnesoid X receptor modulates renal lipid metabolism and diet-induced renal inflammation, fibrosis, and proteinuria. Am. J. Physiol. Renal Physiol. 297, F1587–F1596 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Hong, C. & Tontonoz, P. Coordination of inflammation and metabolism by PPAR and LXR nuclear receptors. Curr. Opin. Genet. Dev. 18, 461–467 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Zhang, Y. et al. Liver X receptor agonist TO-901317 upregulates SCD1 expression in renal proximal straight tubule. Am. J. Physiol. Renal Physiol. 290, F1065–F1073 (2006).

    Article  CAS  PubMed  Google Scholar 

  122. Kuipers, I. et al. Activation of liver X receptor-alpha reduces activation of the renal and cardiac renin–angiotensin–aldosterone system. Lab. Invest. doi:10.1038/labinvest.2010.7.

    Article  CAS  PubMed  Google Scholar 

  123. Giguère, V. Transcriptional control of energy homeostasis by the estrogen-related receptors. Endocr. Rev. 29, 677–696 (2008).

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Moshe Levi.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wang, X., Jiang, T. & Levi, M. Nuclear hormone receptors in diabetic nephropathy. Nat Rev Nephrol 6, 342–351 (2010). https://doi.org/10.1038/nrneph.2010.56

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrneph.2010.56

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing