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 receptors in podocyte biology and glomerular disease

Abstract

Nuclear receptors have a broad spectrum of biological functions in normal physiology and in the pathology of various diseases, including glomerular disease. The primary therapies for many glomerular diseases are glucocorticoids, which exert their immunosuppressive and direct podocyte protective effects via the glucocorticoid receptor (GR). As glucocorticoids are associated with important adverse effects and a substantial proportion of patients show resistance to these therapies, the beneficial effects of selective GR modulators are now being explored. Peroxisome proliferator-activated receptor-γ (PPARγ) agonism using thiazolidinediones has potent podocyte cytoprotective and nephroprotective effects. Repurposing of thiazolidinediones or identification of novel PPARγ modulators are potential strategies to treat non-diabetic glomerular disease. Retinoic acid receptor-α is the key mediator of the renal protective effects of retinoic acid, and repair of the endogenous retinoic acid pathway offers another potential therapeutic strategy for glomerular disease. Vitamin D receptor, oestrogen receptor and mineralocorticoid receptor modulators regulate podocyte injury in experimental models. Further studies are needed to better understand the mechanisms of these nuclear receptors, evaluate their synergistic pathways and identify their novel modulators. Here, we focus on the role of nuclear receptors in podocyte biology and non-diabetic glomerular disease.

Key points

  • Understanding the role of nuclear receptors in podocyte biology and glomerular disease could lead to novel therapeutic strategies for glomerular disease.

  • Glucocorticoids activate the glucocorticoid receptor and have direct protective effects on podocytes in addition to their immunosuppressive effects; glucocorticoid resistance and adverse effects pose huge challenges to the therapeutic use of these agents.

  • Activation of peroxisome proliferator-activated receptor-γ using thiazolidinediones protects against podocyte injury and has beneficial effects in animal models of glomerular disease.

  • Retinoic acid receptor-α mediates the renal protective effects of retinoic acid; repair of the endogenous retinoic acid synthesis pathway might be a novel strategy to treat glomerular disease.

  • Vitamin D3 activates the vitamin D receptor and ameliorates podocyte injury in experimental models; the available data from clinical trials of vitamin D supplementation in patients with chronic kidney disease are inconclusive.

  • A deeper understanding of the mechanisms, synergistic pathways and modulated targeting of nuclear receptor pathways in podocyte biology could enable the development of more effective therapies for non-diabetic glomerular disease.

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

Fig. 1: GR signalling in podocytes.
Fig. 2: PPARγ signalling in adipocytes and in podocytes.
Fig. 3: RAR signalling in podocytes and glomerular disease.
Fig. 4: VDR signalling in podocytes.
Fig. 5: MR signalling in podocytes.
Fig. 6: ER signalling in podocytes.
Fig. 7: Crosstalk between nuclear receptors signalling pathways in podocytes.

Similar content being viewed by others

References

  1. Overington, J. P., Al-Lazikani, B. & Hopkins, A. L. How many drug targets are there? Nat. Rev. Drug Discov. 5, 993–996 (2006).

    CAS  PubMed  Google Scholar 

  2. Mazaira, G. I. et al. The nuclear receptor field: a historical overview and future challenges. Nucl. Receptor Res. 5, 101320 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Jensen, E. V. On the mechanism of estrogen action. Perspect. Biol. Med. 6, 47–59 (1962).

    CAS  PubMed  Google Scholar 

  4. Hollenberg, S. M. et al. Primary structure and expression of a functional human glucocorticoid receptor cDNA. Nature 318, 635–641 (1985).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Nuclear Receptors Nomenclature Committee. A unified nomenclature system for the nuclear receptor superfamily. Cell 97, 161–163 (1999).

    Google Scholar 

  6. Glass, C. K. & Ogawa, S. Combinatorial roles of nuclear receptors in inflammation and immunity. Nat. Rev. Immunol. 6, 44–55 (2006).

    CAS  PubMed  Google Scholar 

  7. Ogawa, S. et al. Molecular determinants of crosstalk between nuclear receptors and toll-like receptors. Cell 122, 707–721 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Cain, D. W. & Cidlowski, J. A. Specificity and sensitivity of glucocorticoid signaling in health and disease. Best Pract. Res. Clin. Endocrinol. Metab. 29, 545–556 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Zhou, J. & Cidlowski, J. A. The human glucocorticoid receptor: one gene, multiple proteins and diverse responses. Steroids 70, 407–417 (2005).

    CAS  PubMed  Google Scholar 

  10. Weikum, E. R., Knuesel, M. T., Ortlund, E. A. & Yamamoto, K. R. Glucocorticoid receptor control of transcription: precision and plasticity via allostery. Nat. Rev. Mol. Cell Biol. 18, 159–174 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Pratt, W. B. & Toft, D. O. Steroid receptor interactions with heat shock protein and immunophilin chaperones. Endocr. Rev. 18, 306–360 (1997).

    CAS  PubMed  Google Scholar 

  12. Zhao, X., Hwang, D. Y. & Kao, H. Y. The role of glucocorticoid receptors in podocytes and nephrotic syndrome. Nucl. Receptor Res. 5, 101323 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Rhen, T. & Cidlowski, J. A. Antiinflammatory action of glucocorticoids–new mechanisms for old drugs. N. Engl. J. Med. 353, 1711–1723 (2005).

    CAS  PubMed  Google Scholar 

  14. Schacke, H., Docke, W. D. & Asadullah, K. Mechanisms involved in the side effects of glucocorticoids. Pharmacol. Ther. 96, 23–43 (2002).

    CAS  PubMed  Google Scholar 

  15. Ponticelli, C. & Locatelli, F. Glucocorticoids in the treatment of glomerular diseases: pitfalls and pearls. Clin. J. Am. Soc. Nephrol. 13, 815–822 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Ito, K., Chung, K. F. & Adcock, I. M. Update on glucocorticoid action and resistance. J. Allergy Clin. Immunol. 117, 522–543 (2006).

    CAS  PubMed  Google Scholar 

  17. Barnes, P. J. & Adcock, I. M. Glucocorticoid resistance in inflammatory diseases. Lancet 373, 1905–1917 (2009).

    CAS  PubMed  Google Scholar 

  18. Cavallo, T., Graves, K. & Granholm, N. A. Murine lupus nephritis. Effects of glucocorticoid on circulating and tissue-bound immunoreactants. Lab. Invest. 49, 476–481 (1983).

    CAS  PubMed  Google Scholar 

  19. Cavallo, T., Graves, K. & Granholm, N. A. Murine lupus nephritis. Effects of glucocorticoid on glomerular permeability. Lab. Invest. 50, 378–384 (1984).

    CAS  PubMed  Google Scholar 

  20. Holdsworth, S. R. & Bellomo, R. Differential effects of steroids on leukocyte-mediated glomerulonephritis in the rabbit. Kidney Int. 26, 162–169 (1984).

    CAS  PubMed  Google Scholar 

  21. Ito, M., Aono, Y., Suzuki, A., Nagamatsu, T. & Suzuki, Y. Accelerated passive Heymann nephritis in rats as an experimental model for membranous glomerulonephritis and effects of azathioprine and prednisolone on the nephritis. Jpn. J. Pharmacol. 49, 101–110 (1989).

    CAS  PubMed  Google Scholar 

  22. Fujiwara, Y. An ultrastructural study of the effect of the steroid in puromycin aminonucleoside nephrosis rats. Virchows Arch. A Pathol. Anat. Histopathol. 405, 11–24 (1984).

    CAS  PubMed  Google Scholar 

  23. Kawamura, T., Yoshioka, T., Bills, T., Fogo, A. & Ichikawa, I. Glucocorticoid activates glomerular antioxidant enzymes and protects glomeruli from oxidant injuries. Kidney Int. 40, 291–301 (1991).

    CAS  PubMed  Google Scholar 

  24. Agrawal, S. et al. Pioglitazone enhances the beneficial effects of glucocorticoids in experimental nephrotic syndrome. Sci. Rep. 6, 24392 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Bertani, T. et al. Steroids and Adriamycin nephrosis. Appl. Pathol. 2, 32–38 (1984).

    CAS  PubMed  Google Scholar 

  26. Pippin, J. W. et al. Inducible rodent models of acquired podocyte diseases. Am. J. Physiol. Ren. Physiol. 296, F213–F229 (2009).

    CAS  Google Scholar 

  27. Zhou, H. et al. Loss of the podocyte glucocorticoid receptor exacerbates proteinuria after injury. Sci. Rep. 7, 9833 (2017).

    PubMed  PubMed Central  Google Scholar 

  28. Kuppe, C. et al. Investigations of glucocorticoid action in GN. J. Am. Soc. Nephrol. 28, 1408–1420 (2017).

    CAS  PubMed  Google Scholar 

  29. Ransom, R. F., Lam, N. G., Hallett, M. A., Atkinson, S. J. & Smoyer, W. E. Glucocorticoids protect and enhance recovery of cultured murine podocytes via actin filament stabilization. Kidney Int. 68, 2473–2483 (2005).

    CAS  PubMed  Google Scholar 

  30. Wada, T., Pippin, J. W., Marshall, C. B., Griffin, S. V. & Shankland, S. J. Dexamethasone prevents podocyte apoptosis induced by puromycin aminonucleoside: role of p53 and Bcl-2-related family proteins. J. Am. Soc. Nephrol. 16, 2615–2625 (2005).

    CAS  PubMed  Google Scholar 

  31. Ransom, R. F., Vega-Warner, V., Smoyer, W. E. & Klein, J. Differential proteomic analysis of proteins induced by glucocorticoids in cultured murine podocytes. Kidney Int. 67, 1275–1285 (2005).

    CAS  PubMed  Google Scholar 

  32. Wada, T., Pippin, J. W., Nangaku, M. & Shankland, S. J. Dexamethasone’s prosurvival benefits in podocytes require extracellular signal-regulated kinase phosphorylation. Nephron Exp. Nephrol. 109, e8–e19 (2008).

    CAS  PubMed  Google Scholar 

  33. Ohashi, T., Uchida, K., Uchida, S., Sasaki, S. & Nitta, K. Dexamethasone increases the phosphorylation of nephrin in cultured podocytes. Clin. Exp. Nephrol. 15, 688–693 (2011).

    CAS  PubMed  Google Scholar 

  34. Mallipattu, S. K. et al. Kruppel-like factor 15 mediates glucocorticoid-induced restoration of podocyte differentiation markers. J. Am. Soc. Nephrol. 28, 166–184 (2017).

    CAS  PubMed  Google Scholar 

  35. Lewko, B. et al. Dexamethasone-dependent modulation of cyclic GMP synthesis in podocytes. Mol. Cell Biochem. 409, 243–253 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Xie, H. et al. Inhibition of microRNA-30a prevents puromycin aminonucleoside-induced podocytic apoptosis by upregulating the glucocorticoid receptor alpha. Mol. Med. Rep. 12, 6043–6052 (2015).

    CAS  PubMed  Google Scholar 

  37. Wu, J. et al. Downregulation of microRNA-30 facilitates podocyte injury and is prevented by glucocorticoids. J. Am. Soc. Nephrol. 25, 92–104 (2014).

    CAS  PubMed  Google Scholar 

  38. Guess, A. et al. Dose- and time-dependent glucocorticoid receptor signaling in podocytes. Am. J. Physiol. Ren. Physiol. 299, F845–F853 (2010).

    CAS  Google Scholar 

  39. Agrawal, S., Guess, A. J., Benndorf, R. & Smoyer, W. E. Comparison of direct action of thiazolidinediones and glucocorticoids on renal podocytes: protection from injury and molecular effects. Mol. Pharmacol. 80, 389–399 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Agrawal, S., Guess, A. J., Chanley, M. A. & Smoyer, W. E. Albumin-induced podocyte injury and protection are associated with regulation of COX-2. Kidney Int. 86, 1150–1160 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Luetscher, J. A. Jr. & Deming, Q. B. Treatment of nephrosis with cortisone. J. Clin. Invest. 29, 1576–1587 (1950).

    PubMed  PubMed Central  Google Scholar 

  42. Nourbakhsh, N. & Mak, R. H. Steroid-resistant nephrotic syndrome: past and current perspectives. Pediatric Health Med. Ther. 8, 29–37 (2017).

    PubMed  PubMed Central  Google Scholar 

  43. Canetta, P. A. & Radhakrishnan, J. The evidence-based approach to adult-onset idiopathic nephrotic syndrome. Front. Pediatr. 3, 78 (2015).

    PubMed  PubMed Central  Google Scholar 

  44. Agrawal, S. et al. Predicting and defining steroid resistance in pediatric nephrotic syndrome using plasma proteomics. Kidney Int. Rep. 5, 66–80 (2020).

    PubMed  Google Scholar 

  45. Gooding, J. R. et al. Predicting and defining steroid resistance in pediatric nephrotic syndrome using plasma metabolomics. Kidney Int. Rep. 5, 81–93 (2020).

    PubMed  Google Scholar 

  46. Bennett, M. R. et al. A novel biomarker panel to identify steroid resistance in childhood idiopathic nephrotic syndrome. Biomarker Insights 12, 1177271917695832 (2017).

    PubMed  PubMed Central  Google Scholar 

  47. Saleem, M. A. Molecular stratification of idiopathic nephrotic syndrome. Nat. Rev. Nephrol. 15, 750–765 (2019).

    PubMed  Google Scholar 

  48. Newton, R. & Holden, N. S. Separating transrepression and transactivation: a distressing divorce for the glucocorticoid receptor? Mol. Pharmacol. 72, 799–809 (2007).

    CAS  PubMed  Google Scholar 

  49. Gessi, S., Merighi, S. & Borea, P. A. Glucocorticoid’s pharmacology: past, present and future. Curr. Pharm. Des. 16, 3540–3553 (2010).

    CAS  PubMed  Google Scholar 

  50. Zhu, Y., Alvares, K., Huang, Q., Rao, M. S. & Reddy, J. K. Cloning of a new member of the peroxisome proliferator-activated receptor gene family from mouse liver. J. Biol. Chem. 268, 26817–26820 (1993).

    CAS  PubMed  Google Scholar 

  51. Chen, F., Law, S. W. & O’Malley, B. W. Identification of two mPPAR related receptors and evidence for the existence of five subfamily members. Biochem. Biophys. Res. Commun. 196, 671–677 (1993).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Miglio, G. et al. The subtypes of peroxisome proliferator-activated receptors expressed by human podocytes and their role in decreasing podocyte injury. Br. J. Pharmacol. 162, 111–125 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Mukherjee, R., Jow, L., Croston, G. E. & Paterniti, J. R. Jr Identification, characterization, and tissue distribution of human peroxisome proliferator-activated receptor (PPAR) isoforms PPARγ2 versus PPARγ1 and activation with retinoid X receptor agonists and antagonists. J. Biol. Chem. 272, 8071–8076 (1997).

    CAS  PubMed  Google Scholar 

  55. Aprile, M. et al. PPARγΔ5, a naturally occurring dominant-negative splice isoform, impairs PPARγ function and adipocyte differentiation. Cell Rep. 25, 1577–1592.e6 (2018).

    CAS  PubMed  Google Scholar 

  56. Sabatino, L. et al. A novel peroxisome proliferator-activated receptor gamma isoform with dominant negative activity generated by alternative splicing. J. Biol. Chem. 280, 26517–26525 (2005).

    CAS  PubMed  Google Scholar 

  57. Varanasi, U. et al. Identification of a peroxisome proliferator-responsive element upstream of the human peroxisomal fatty acyl coenzyme A oxidase gene. J. Biol. Chem. 271, 2147–2155 (1996).

    CAS  PubMed  Google Scholar 

  58. Chandra, V. et al. Structure of the intact PPAR-γ-RXR–nuclear receptor complex on DNA. Nature 456, 350–356 (2008).

    PubMed  PubMed Central  Google Scholar 

  59. Leo, C. & Chen, J. D. The SRC family of nuclear receptor coactivators. Gene 245, 1–11 (2000).

    CAS  PubMed  Google Scholar 

  60. Siersbaek, R. et al. Molecular architecture of transcription factor hotspots in early adipogenesis. Cell Rep. 7, 1434–1442 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Dubois-Chevalier, J. et al. A dynamic CTCF chromatin binding landscape promotes DNA hydroxymethylation and transcriptional induction of adipocyte differentiation. Nucleic Acids Res. 42, 10943–10959 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Puigserver, P. et al. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell 92, 829–839 (1998).

    CAS  PubMed  Google Scholar 

  63. Lin, J., Puigserver, P., Donovan, J., Tarr, P. & Spiegelman, B. M. Peroxisome proliferator-activated receptor γ coactivator 1β (PGC-1β), a novel PGC-1-related transcription coactivator associated with host cell factor. J. Biol. Chem. 277, 1645–1648 (2002).

    CAS  PubMed  Google Scholar 

  64. Tomaru, T. et al. Isolation and characterization of a transcriptional cofactor and its novel isoform that bind the deoxyribonucleic acid-binding domain of peroxisome proliferator-activated receptor-γ. Endocrinology 147, 377–388 (2006).

    CAS  PubMed  Google Scholar 

  65. Zhu, Y. et al. Isolation and characterization of peroxisome proliferator-activated receptor (PPAR) interacting protein (PRIP) as a coactivator for PPAR. J. Biol. Chem. 275, 13510–13516 (2000).

    CAS  PubMed  Google Scholar 

  66. Ricote, M. & Glass, C. K. PPARs and molecular mechanisms of transrepression. Biochim. Biophys. Acta 1771, 926–935 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Glass, C. K. & Saijo, K. Nuclear receptor transrepression pathways that regulate inflammation in macrophages and T cells. Nat. Rev. Immunol. 10, 365–376 (2010).

    CAS  PubMed  Google Scholar 

  68. Viswakarma, N. et al. Coactivators in PPAR-regulated gene expression. PPAR Res. 2010, 250126 (2010).

    PubMed  PubMed Central  Google Scholar 

  69. Moran-Salvador, E. et al. Role for PPARγ in obesity-induced hepatic steatosis as determined by hepatocyte- and macrophage-specific conditional knockouts. FASEB J. 25, 2538–2550 (2011).

    CAS  PubMed  Google Scholar 

  70. Tontonoz, P., Hu, E. & Spiegelman, B. M. Stimulation of adipogenesis in fibroblasts by PPARγ2, a lipid-activated transcription factor. Cell 79, 1147–1156 (1994).

    CAS  PubMed  Google Scholar 

  71. Lefterova, M. I., Haakonsson, A. K., Lazar, M. A. & Mandrup, S. PPARγ and the global map of adipogenesis and beyond. Trends Endocrinol. Metab. 25, 293–302 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Sun, X., Han, R., Wang, Z. & Chen, Y. Regulation of adiponectin receptors in hepatocytes by the peroxisome proliferator-activated receptor-γ agonist rosiglitazone. Diabetologia 49, 1303–1310 (2006).

    CAS  PubMed  Google Scholar 

  73. Stewart, W. C., Morrison, R. F., Young, S. L. & Stephens, J. M. Regulation of signal transducers and activators of transcription (STATs) by effectors of adipogenesis: coordinate regulation of STATs 1, 5A, and 5B with peroxisome proliferator-activated receptor-γ and C/AAAT enhancer binding protein-α. Biochim. Biophys. Acta 1452, 188–196 (1999).

    CAS  PubMed  Google Scholar 

  74. Olsen, H. & Haldosen, L. A. Peroxisome proliferator-activated receptor gamma regulates expression of signal transducer and activator of transcription 5A. Exp. Cell Res. 312, 1371–1380 (2006).

    CAS  PubMed  Google Scholar 

  75. Prost, S. et al. Human and simian immunodeficiency viruses deregulate early hematopoiesis through a Nef/PPARγ/STAT5 signaling pathway in macaques. J. Clin. Invest. 118, 1765–1775 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Heikkinen, S., Auwerx, J. & Argmann, C. A. PPARgamma in human and mouse physiology. Biochim. Biophys. Acta 1771, 999–1013 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Sato, K. et al. Expression of peroxisome proliferator-activated receptor isoform proteins in the rat kidney. Hypertens. Res. 27, 417–425 (2004).

    CAS  PubMed  Google Scholar 

  78. Yang, T. et al. Expression of peroxisomal proliferator-activated receptors and retinoid X receptors in the kidney. Am. J. Physiol. 277, F966–F973 (1999).

    CAS  PubMed  Google Scholar 

  79. Henique, C. et al. Nuclear factor erythroid 2-related factor 2 drives podocyte-specific expression of peroxisome proliferator-activated receptor γ essential for resistance to crescentic GN. J. Am. Soc. Nephrol. 27, 172–188 (2016).

    CAS  PubMed  Google Scholar 

  80. Long, Q. et al. Peroxisome proliferator-activated receptor-γ increases adiponectin secretion via transcriptional repression of endoplasmic reticulum chaperone protein ERp44. Endocrinology 151, 3195–3203 (2010).

    CAS  PubMed  Google Scholar 

  81. Choi, J. H. et al. Anti-diabetic drugs inhibit obesity-linked phosphorylation of PPARγ by Cdk5. Nature 466, 451–456 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Rutkowski, J. M. et al. Adiponectin promotes functional recovery after podocyte ablation. J. Am. Soc. Nephrol. 24, 268–282 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Pascual, G. et al. A SUMOylation-dependent pathway mediates transrepression of inflammatory response genes by PPAR-γ. Nature 437, 759–763 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Tsukahara, T. et al. Phospholipase D2-dependent inhibition of the nuclear hormone receptor PPARγ by cyclic phosphatidic acid. Mol. Cell 39, 421–432 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. McIntyre, T. M. et al. Identification of an intracellular receptor for lysophosphatidic acid (LPA): LPA is a transcellular PPARγ agonist. Proc. Natl Acad. Sci. USA 100, 131–136 (2003).

    CAS  PubMed  Google Scholar 

  86. Ristow, M., Muller-Wieland, D., Pfeiffer, A., Krone, W. & Kahn, C. R. Obesity associated with a mutation in a genetic regulator of adipocyte differentiation. N. Engl. J. Med. 339, 953–959 (1998).

    CAS  PubMed  Google Scholar 

  87. Barroso, I. et al. Dominant negative mutations in human PPARγ associated with severe insulin resistance, diabetes mellitus and hypertension. Nature 402, 880–883 (1999).

    CAS  PubMed  Google Scholar 

  88. Nikiforova, M. N. et al. RAS point mutations and PAX8-PPARγ rearrangement in thyroid tumors: evidence for distinct molecular pathways in thyroid follicular carcinoma. J. Clin. Endocrinol. Metab. 88, 2318–2326 (2003).

    CAS  PubMed  Google Scholar 

  89. Marques, A. R. et al. Expression of PAX8-PPARγ1 rearrangements in both follicular thyroid carcinomas and adenomas. J. Clin. Endocrinol. Metab. 87, 3947–3952 (2002).

    CAS  PubMed  Google Scholar 

  90. Rochel, N. et al. Recurrent activating mutations of PPARγ associated with luminal bladder tumors. Nat. Commun. 10, 253 (2019).

    PubMed  PubMed Central  Google Scholar 

  91. Halstead, A. M. et al. Bladder-cancer-associated mutations in RXRA activate peroxisome proliferator-activated receptors to drive urothelial proliferation. eLife 6, e30862 (2017).

    PubMed  PubMed Central  Google Scholar 

  92. Toffoli, B. et al. Nephropathy in Pparg-null mice highlights PPARγ systemic activities in metabolism and in the immune system. PLoS ONE 12, e0171474 (2017).

    PubMed  PubMed Central  Google Scholar 

  93. Chinetti, G., Fruchart, J. C. & Staels, B. Peroxisome proliferator-activated receptors (PPARs): nuclear receptors at the crossroads between lipid metabolism and inflammation. Inflamm. Res. 49, 497–505 (2000).

    CAS  PubMed  Google Scholar 

  94. Nakamura, T. et al. Pioglitazone reduces urinary podocyte excretion in type 2 diabetes patients with microalbuminuria. Metabolism 50, 1193–1196 (2001).

    CAS  PubMed  Google Scholar 

  95. Sarafidis, P. A., Stafylas, P. C., Georgianos, P. I., Saratzis, A. N. & Lasaridis, A. N. Effect of thiazolidinediones on albuminuria and proteinuria in diabetes: a meta-analysis. Am. J. Kidney Dis. 55, 835–847 (2010).

    CAS  PubMed  Google Scholar 

  96. Schneider, C. A. et al. Effect of pioglitazone on cardiovascular outcome in diabetes and chronic kidney disease. J. Am. Soc. Nephrol. 19, 182–187 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Kernan, W. N. et al. Pioglitazone after ischemic stroke or transient ischemic attack. N. Engl. J. Med. 374, 1321–1331 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Young, L. H. et al. Cardiac outcomes after ischemic stroke or transient ischemic attack: effects of pioglitazone in patients with insulin resistance without diabetes mellitus. Circulation 135, 1882–1893 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Liu, J. & Wang, L. N. Peroxisome proliferator-activated receptor gamma agonists for preventing recurrent stroke and other vascular events in people with stroke or transient ischaemic attack. Cochrane Database Syst. Rev. 10, CD010693 (2019).

    PubMed  Google Scholar 

  100. Zhou, Y. et al. Pioglitazone for the primary and secondary prevention of cardiovascular and renal outcomes in patients with or at high risk of type 2 diabetes mellitus: a meta-analysis. J. Clin. Endocrinol. Metab. 105, dgz252 (2020).

    PubMed  Google Scholar 

  101. Nissen, S. E. & Wolski, K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N. Engl. J. Med. 356, 2457–2471 (2007).

    CAS  PubMed  Google Scholar 

  102. Wallach, J. D. et al. Updating insights into rosiglitazone and cardiovascular risk through shared data: individual patient and summary level meta-analyses. BMJ 368, l7078 (2020).

    PubMed  PubMed Central  Google Scholar 

  103. van Wijk, J. P., de Koning, E. J., Martens, E. P. & Rabelink, T. J. Thiazolidinediones and blood lipids in type 2 diabetes. Arterioscler. Thromb. Vasc. Biol. 23, 1744–1749 (2003).

    PubMed  Google Scholar 

  104. Deeg, M. A. et al. Pioglitazone and rosiglitazone have different effects on serum lipoprotein particle concentrations and sizes in patients with type 2 diabetes and dyslipidemia. Diabetes Care 30, 2458–2464 (2007).

    CAS  PubMed  Google Scholar 

  105. Mamtani, R. et al. Association between longer therapy with thiazolidinediones and risk of bladder cancer: a cohort study. J. Natl Cancer Inst. 104, 1411–1421 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Graham, D. J. et al. Risk of acute myocardial infarction, stroke, heart failure, and death in elderly Medicare patients treated with rosiglitazone or pioglitazone. JAMA 304, 411–418 (2010).

    CAS  PubMed  Google Scholar 

  107. Buckingham, R. E. et al. Peroxisome proliferator-activated receptor-γ agonist, rosiglitazone, protects against nephropathy and pancreatic islet abnormalities in Zucker fatty rats. Diabetes 47, 1326–1334 (1998).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  109. Tanimoto, M. et al. Effect of pioglitazone on the early stage of type 2 diabetic nephropathy in KK/Ta mice. Metabolism 53, 1473–1479 (2004).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  111. Straus, D. S. & Glass, C. K. Anti-inflammatory actions of PPAR ligands: new insights on cellular and molecular mechanisms. Trends Immunol. 28, 551–558 (2007).

    CAS  PubMed  Google Scholar 

  112. Bouhlel, M. A. et al. PPARγ activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab. 6, 137–143 (2007).

    CAS  PubMed  Google Scholar 

  113. Chinetti, G. et al. Activation of proliferator-activated receptors α and γ induces apoptosis of human monocyte-derived macrophages. J. Biol. Chem. 273, 25573–25580 (1998).

    CAS  PubMed  Google Scholar 

  114. Ma, L. J., Marcantoni, C., Linton, M. F., Fazio, S. & Fogo, A. B. Peroxisome proliferator-activated receptor-γ agonist troglitazone protects against nondiabetic glomerulosclerosis in rats. Kidney Int. 59, 1899–1910 (2001).

    CAS  PubMed  Google Scholar 

  115. Yang, H. C. et al. The PPARγ agonist pioglitazone ameliorates aging-related progressive renal injury. J. Am. Soc. Nephrol. 20, 2380–2388 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Yang, H. C., Ma, L. J., Ma, J. & Fogo, A. B. Peroxisome proliferator-activated receptor-gamma agonist is protective in podocyte injury-associated sclerosis. Kidney Int. 69, 1756–1764 (2006).

    CAS  PubMed  Google Scholar 

  117. Liu, H. F. et al. Thiazolidinedione attenuate proteinuria and glomerulosclerosis in Adriamycin-induced nephropathy rats via slit diaphragm protection. Nephrology 15, 75–83 (2010).

    CAS  PubMed  Google Scholar 

  118. Zuo, Y. et al. Protective effects of PPARγ agonist in acute nephrotic syndrome. Nephrol. Dial. Transplant. 27, 174–181 (2012).

    CAS  PubMed  Google Scholar 

  119. Haraguchi, K., Shimura, H. & Onaya, T. Suppression of experimental crescentic glomerulonephritis by peroxisome proliferator-activated receptor (PPAR)γ activators. Clin. Exp. Nephrol. 7, 27–32 (2003).

    CAS  PubMed  Google Scholar 

  120. Chafin, C. et al. Deletion of PPAR-γ in immune cells enhances susceptibility to antiglomerular basement membrane disease. J. Inflamm. Res. 3, 127–134 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Cho, H. Y. et al. Nrf2-regulated PPARγ expression is critical to protection against acute lung injury in mice. Am. J. Respir. Crit. Care Med. 182, 170–182 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Cho, H. Y., Reddy, S. P., Debiase, A., Yamamoto, M. & Kleeberger, S. R. Gene expression profiling of NRF2-mediated protection against oxidative injury. Free Radic. Biol. Med. 38, 325–343 (2005).

    CAS  PubMed  Google Scholar 

  123. Huang, J., Tabbi-Anneni, I., Gunda, V. & Wang, L. Transcription factor Nrf2 regulates SHP and lipogenic gene expression in hepatic lipid metabolism. Am. J. Physiol. Gastrointest. Liver Physiol. 299, G1211–G1221 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Sonneveld, R. et al. Sildenafil prevents podocyte injury via PPAR-γ-mediated TRPC6 inhibition. J. Am. Soc. Nephrol. 28, 1491–1505 (2017).

    CAS  PubMed  Google Scholar 

  125. Winn, M. P. et al. A mutation in the TRPC6 cation channel causes familial focal segmental glomerulosclerosis. Science 308, 1801–1804 (2005).

    CAS  PubMed  Google Scholar 

  126. Paueksakon, P., Revelo, M. P., Ma, L. J., Marcantoni, C. & Fogo, A. B. Microangiopathic injury and augmented PAI-1 in human diabetic nephropathy. Kidney Int. 61, 2142–2148 (2002).

    CAS  PubMed  Google Scholar 

  127. Okada, T. et al. Thiazolidinediones ameliorate diabetic nephropathy via cell cycle-dependent mechanisms. Diabetes 55, 1666–1677 (2006).

    CAS  PubMed  Google Scholar 

  128. Kanjanabuch, T. et al. PPAR-γ agonist protects podocytes from injury. Kidney Int. 71, 1232–1239 (2007).

    CAS  PubMed  Google Scholar 

  129. Miglio, G. et al. Protective effects of peroxisome proliferator-activated receptor agonists on human podocytes: proposed mechanisms of action. Br. J. Pharmacol. 167, 641–653 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Miceli, I. et al. Stretch reduces nephrin expression via an angiotensin II-AT(1)-dependent mechanism in human podocytes: effect of rosiglitazone. Am. J. Physiol. Ren. Physiol. 298, F381–F390 (2010).

    CAS  Google Scholar 

  131. Zhu, C. et al. Mitochondrial dysfunction mediates aldosterone-induced podocyte damage: a therapeutic target of PPARγ. Am. J. Pathol. 178, 2020–2031 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Zhou, Z. et al. MicroRNA-27a promotes podocyte injury via PPARγ-mediated β-catenin activation in diabetic nephropathy. Cell Death Dis. 8, e2658 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Ahmadian, M. et al. PPARγ signaling and metabolism: the good, the bad and the future. Nat. Med. 19, 557–566 (2013).

    CAS  PubMed  Google Scholar 

  134. Burris, T. P. et al. Nuclear receptors and their selective pharmacologic modulators. Pharmacol. Rev. 65, 710–778 (2013).

    PubMed  Google Scholar 

  135. Choi, J. H. et al. Antidiabetic actions of a non-agonist PPARγ ligand blocking Cdk5-mediated phosphorylation. Nature 477, 477–481 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Marciano, D. P. et al. Pharmacological repression of PPARγ promotes osteogenesis. Nat. Commun. 6, 7443 (2015).

    CAS  PubMed  Google Scholar 

  137. Allenby, G. et al. Retinoic acid receptors and retinoid X receptors: interactions with endogenous retinoic acids. Proc. Natl Acad. Sci. USA 90, 30–34 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Xu, Q. et al. Retinoids in nephrology: promises and pitfalls. Kidney Int. 66, 2119–2131 (2004).

    CAS  PubMed  Google Scholar 

  139. Fisher, G. J. et al. Immunological identification and functional quantitation of retinoic acid and retinoid X receptor proteins in human skin. J. Biol. Chem. 269, 20629–20635 (1994).

    CAS  PubMed  Google Scholar 

  140. Fitzgerald, P., Teng, M., Chandraratna, R. A., Heyman, R. A. & Allegretto, E. A. Retinoic acid receptor alpha expression correlates with retinoid-induced growth inhibition of human breast cancer cells regardless of estrogen receptor status. Cancer Res. 57, 2642–2650 (1997).

    CAS  PubMed  Google Scholar 

  141. Jones, K. A. et al. Localization of the retinoid X receptor alpha gene (RXRA) to chromosome 9q34. Ann. Hum. Genet. 57, 195–201 (1993).

    CAS  PubMed  Google Scholar 

  142. Harrison, E. H. Mechanisms of digestion and absorption of dietary vitamin A. Annu. Rev. Nutr. 25, 87–103 (2005).

    CAS  PubMed  Google Scholar 

  143. Kawaguchi, R. et al. A membrane receptor for retinol binding protein mediates cellular uptake of vitamin A. Science 315, 820–825 (2007).

    CAS  PubMed  Google Scholar 

  144. Liu, L. & Gudas, L. J. Disruption of the lecithin:retinol acyltransferase gene makes mice more susceptible to vitamin A deficiency. J. Biol. Chem. 280, 40226–40234 (2005).

    CAS  PubMed  Google Scholar 

  145. Blaner, W. S. et al. Lipoprotein lipase hydrolysis of retinyl ester. Possible implications for retinoid uptake by cells. J. Biol. Chem. 269, 16559–16565 (1994).

    CAS  PubMed  Google Scholar 

  146. Duester, G. Retinoic acid synthesis and signaling during early organogenesis. Cell 134, 921–931 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Touma, S. E., Perner, S., Rubin, M. A., Nanus, D. M. & Gudas, L. J. Retinoid metabolism and ALDH1A2 (RALDH2) expression are altered in the transgenic adenocarcinoma mouse prostate model. Biochem. Pharmacol. 78, 1127–1138 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Gronemeyer, H. & Miturski, R. Molecular mechanisms of retinoid action. Cell Mol. Biol. Lett. 6, 3–52 (2001).

    CAS  PubMed  Google Scholar 

  149. Na, S. Y. et al. Retinoids inhibit interleukin-12 production in macrophages through physical associations of retinoid X receptor and NFκB. J. Biol. Chem. 274, 7674–7680 (1999).

    CAS  PubMed  Google Scholar 

  150. Benkoussa, M., Brand, C., Delmotte, M. H., Formstecher, P. & Lefebvre, P. Retinoic acid receptors inhibit AP1 activation by regulating extracellular signal-regulated kinase and CBP recruitment to an AP1-responsive promoter. Mol. Cell Biol. 22, 4522–4534 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Simonson, M. S. Anti-AP-1 activity of all-trans retinoic acid in glomerular mesangial cells. Am. J. Physiol. 267, F805–F815 (1994).

    CAS  PubMed  Google Scholar 

  152. Wang, R. et al. All-trans-retinoic acid reduces BACE1 expression under inflammatory conditions via modulation of nuclear factor κB (NFκB) signaling. J. Biol. Chem. 290, 22532–22542 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Balmer, J. E. & Blomhoff, R. Gene expression regulation by retinoic acid. J. Lipid Res. 43, 1773–1808 (2002).

    CAS  PubMed  Google Scholar 

  154. Canon, E., Cosgaya, J. M., Scsucova, S. & Aranda, A. Rapid effects of retinoic acid on CREB and ERK phosphorylation in neuronal cells. Mol. Biol. Cell 15, 5583–5592 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Lonze, B. E. & Ginty, D. D. Function and regulation of CREB family transcription factors in the nervous system. Neuron 35, 605–623 (2002).

    CAS  PubMed  Google Scholar 

  156. Zhao, Q. et al. Rapid induction of cAMP/PKA pathway during retinoic acid-induced acute promyelocytic leukemia cell differentiation. Leukemia 18, 285–292 (2004).

    CAS  PubMed  Google Scholar 

  157. Parrella, E. et al. Phosphodiesterase IV inhibition by piclamilast potentiates the cytodifferentiating action of retinoids in myeloid leukemia cells. Cross-talk between the cAMP and the retinoic acid signaling pathways. J. Biol. Chem. 279, 42026–42040 (2004).

    CAS  PubMed  Google Scholar 

  158. Boskovic, G., Desai, D. & Niles, R. M. Regulation of retinoic acid receptor α by protein kinase C in B16 mouse melanoma cells. J. Biol. Chem. 277, 26113–26119 (2002).

    CAS  PubMed  Google Scholar 

  159. Hughes, P. J., Zhao, Y., Chandraratna, R. A. & Brown, G. Retinoid-mediated stimulation of steroid sulfatase activity in myeloid leukemic cell lines requires RARα and RXR and involves the phosphoinositide 3-kinase and ERK-MAP kinase pathways. J. Cell. Biochem. 97, 327–350 (2006).

    CAS  PubMed  Google Scholar 

  160. Evans, T. R. & Kaye, S. B. Retinoids: present role and future potential. Br. J. Cancer 80, 1–8 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Cabezas-Wallscheid, N. et al. Vitamin A-retinoic acid signaling regulates hematopoietic stem cell dormancy. Cell 169, 807–823.e19 (2017).

    CAS  PubMed  Google Scholar 

  162. Clemens, G. et al. The action of all-trans-retinoic acid (ATRA) and synthetic retinoid analogues (EC19 and EC23) on human pluripotent stem cells differentiation investigated using single cell infrared microspectroscopy. Mol. Biosyst. 9, 677–692 (2013).

    CAS  PubMed  Google Scholar 

  163. Uray, I. P., Dmitrovsky, E. & Brown, P. H. Retinoids and rexinoids in cancer prevention: from laboratory to clinic. Semin. Oncol. 43, 49–64 (2016).

    CAS  PubMed  Google Scholar 

  164. Simoni, D. & Tolomeo, M. Retinoids, apoptosis and cancer. Curr. Pharm. Des. 7, 1823–1837 (2001).

    CAS  PubMed  Google Scholar 

  165. Kitamura, M. et al. Intervention by retinoic acid in oxidative stress-induced apoptosis. Nephrol. Dial., Transplant. 17 (Suppl 9), 84–87 (2002).

    CAS  Google Scholar 

  166. Riahi, R. R., Bush, A. E. & Cohen, P. R. Topical retinoids: therapeutic mechanisms in the treatment of photodamaged skin. Am. J. Clin. Dermatol. 17, 265–276 (2016).

    PubMed  Google Scholar 

  167. Beckenbach, L., Baron, J. M., Merk, H. F., Loffler, H. & Amann, P. M. Retinoid treatment of skin diseases. Eur. J. Dermatol. 25, 384–391 (2015).

    CAS  PubMed  Google Scholar 

  168. Tallman, M. S. All-trans-retinoic acid in acute promyelocytic leukemia and its potential in other hematologic malignancies. Semin. Hematol. 31, 38–48 (1994).

    CAS  PubMed  Google Scholar 

  169. Brzezinski, P., Borowska, K., Chiriac, A. & Smigielski, J. Adverse effects of isotretinoin: a large, retrospective review. Dermatol. Ther. 30, e12483 (2017).

    Google Scholar 

  170. Lehrke, I. et al. Retinoid receptor-specific agonists alleviate experimental glomerulonephritis. Am. J. Physiol. Ren. Physiol. 282, F741–F751 (2002).

    CAS  Google Scholar 

  171. Wagner, J. et al. Retinoic acid reduces glomerular injury in a rat model of glomerular damage. J. Am. Soc. Nephrol. 11, 1479–1487 (2000).

    CAS  PubMed  Google Scholar 

  172. Moreno-Manzano, V. et al. Retinoids as a potential treatment for experimental puromycin-induced nephrosis. Br. J. Pharmacol. 139, 823–831 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Inagaki, T. et al. The retinoic acid-responsive proline-rich protein is identified in promyeloleukemic HL-60 cells. J. Biol. Chem. 278, 51685–51692 (2003).

    CAS  PubMed  Google Scholar 

  174. Schaier, M. et al. Isotretinoin alleviates renal damage in rat chronic glomerulonephritis. Kidney Int. 60, 2222–2234 (2001).

    CAS  PubMed  Google Scholar 

  175. Perez de Lema, G. et al. Retinoic acid treatment protects MRL/lpr lupus mice from the development of glomerular disease. Kidney Int. 66, 1018–1028 (2004).

    CAS  PubMed  Google Scholar 

  176. Vaughan, M. R. et al. ATRA induces podocyte differentiation and alters nephrin and podocin expression in vitro and in vivo. Kidney Int. 68, 133–144 (2005).

    CAS  PubMed  Google Scholar 

  177. Morath, C. et al. Effects of retinoids on the TGF-beta system and extracellular matrix in experimental glomerulonephritis. J. Am. Soc. Nephrol. 12, 2300–2309 (2001).

    CAS  PubMed  Google Scholar 

  178. Dechow, C. et al. Effects of all-trans retinoic acid on renin-angiotensin system in rats with experimental nephritis. Am. J. Physiol. Ren. Physiol. 281, F909–F919 (2001).

    CAS  Google Scholar 

  179. Moreno-Manzano, V., Ishikawa, Y., Lucio-Cazana, J. & Kitamura, M. Suppression of apoptosis by all-trans-retinoic acid. Dual intervention in the c-Jun n-terminal kinase-AP-1 pathway. J. Biol. Chem. 274, 20251–20258 (1999).

    CAS  PubMed  Google Scholar 

  180. Xu, Q., Konta, T. & Kitamura, M. Retinoic acid regulation of mesangial cell apoptosis. Exp. Nephrol. 10, 171–175 (2002).

    CAS  PubMed  Google Scholar 

  181. Ratnam, K. K. et al. Role of the retinoic acid receptor-α in HIV-associated nephropathy. Kidney Int 79, 624–634 (2011).

    CAS  PubMed  Google Scholar 

  182. Dai, Y. et al. Retinoic acid improves nephrotoxic serum-induced glomerulonephritis through activation of podocyte retinoic acid receptor α. Kidney Int. 92, 1444–1457 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Mallipattu, S. K. & He, J. C. The beneficial role of retinoids in glomerular disease. Front. Med. 2, 16 (2015).

    Google Scholar 

  184. Shankland, S. J., Pippin, J. W., Reiser, J. & Mundel, P. Podocytes in culture: past, present, and future. Kidney Int. 72, 26–36 (2007).

    CAS  PubMed  Google Scholar 

  185. Lazzeri, E., Peired, A. J., Lasagni, L. & Romagnani, P. Retinoids and glomerular regeneration. Semin. Nephrol. 34, 429–436 (2014).

    CAS  PubMed  Google Scholar 

  186. He, J. C. et al. Retinoic acid inhibits HIV-1-induced podocyte proliferation through the cAMP pathway. J. Am. Soc. Nephrol. 18, 93–102 (2007).

    CAS  PubMed  Google Scholar 

  187. Zhang, J. et al. Retinoids augment the expression of podocyte proteins by glomerular parietal epithelial cells in experimental glomerular disease. Nephron Exp. Nephrol. 121, e23–e37 (2012).

    CAS  PubMed  Google Scholar 

  188. Merlet-Benichou, C., Vilar, J., Lelievre-Pegorier, M. & Gilbert, T. Role of retinoids in renal development: pathophysiological implication. Curr. Opin. Nephrol. Hypertens. 8, 39–43 (1999).

    CAS  PubMed  Google Scholar 

  189. Gilbert, T. Vitamin A and kidney development. Nephro. Dial. Transplant. 17 (Suppl 9), 78–80 (2002).

    CAS  Google Scholar 

  190. Mallipattu, S. K. et al. Kruppel-like factor 15 (KLF15) is a key regulator of podocyte differentiation. J. Biol. Chem. 287, 19122–19135 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  191. Peired, A. et al. Proteinuria impairs podocyte regeneration by sequestering retinoic acid. J. Am. Soc. Nephrol. 24, 1756–1768 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Standeven, A. M., Teng, M. & Chandraratna, R. A. Lack of involvement of retinoic acid receptor α in retinoid-induced skin irritation in hairless mice. Toxicol. Lett. 92, 231–240 (1997).

    CAS  PubMed  Google Scholar 

  193. Nagpal, S. & Chandraratna, R. A. Recent developments in receptor-selective retinoids. Curr. Pharm. Des. 6, 919–931 (2000).

    CAS  PubMed  Google Scholar 

  194. Advani, A. et al. Role of VEGF in maintaining renal structure and function under normotensive and hypertensive conditions. Proc. Natl Acad. Sci. USA 104, 14448–14453 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Zhong, Y. et al. Novel retinoic acid receptor alpha agonists for treatment of kidney disease. PLoS ONE 6, e27945 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/results/NCT00098020 (2017).

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

    CAS  PubMed  Google Scholar 

  198. Wang, X. X. et al. Diabetic nephropathy is accelerated by farnesoid X receptor deficiency and inhibited by farnesoid X receptor activation in a type 1 diabetes model. Diabetes 59, 2916–2927 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  Google Scholar 

  200. NIH Office of Dietary Supplements. Vitamin D. ODS https://ods.od.nih.gov/factsheets/VitaminD-HealthProfessional/#en1 (2020).

  201. Perez-Gomez, M. V., Ortiz-Arduan, A. & Lorenzo-Sellares, V. Vitamin D and proteinuria: a critical review of molecular bases and clinical experience. Nefrologia 33, 716–726 (2013).

    PubMed  Google Scholar 

  202. Pike, J. W. & Meyer, M. B. The vitamin D receptor: new paradigms for the regulation of gene expression by 1,25-dihydroxyvitamin D(3). Endocrinol. Metab. Clin. North Am. 39, 255–269 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Sertznig, P., Seifert, M., Tilgen, W. & Reichrath, J. Peroxisome proliferator-activated receptor (PPAR) and vitamin D receptor (VDR) signaling pathways in melanoma cells: promising new therapeutic targets? J. Steroid Biochem. Mol. Biol. 121, 383–386 (2010).

    CAS  PubMed  Google Scholar 

  204. Chang, S. W. & Lee, H. C. Vitamin D and health – the missing vitamin in humans. Pediatr. Neonatol. 60, 237–244 (2019).

    PubMed  Google Scholar 

  205. Wang, W., Zhang, J., Wang, H., Wang, X. & Liu, S. Vitamin D deficiency enhances insulin resistance by promoting inflammation in type 2 diabetes. Int. J. Clin. Exp. Pathol. 12, 1859–1867 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Talaei, A., Mohamadi, M. & Adgi, Z. The effect of vitamin D on insulin resistance in patients with type 2 diabetes. Diabetol. Metab. Syndr. 5, 8 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Mutt, S. J. et al. Vitamin D deficiency induces insulin resistance and re-supplementation attenuates hepatic glucose output via the PI3K-AKT-FOXO1 mediated pathway. Mol. Nutr. Food Res. 64, e1900728 (2020).

    PubMed  Google Scholar 

  208. Sonneveld, R. et al. Vitamin D down-regulates TRPC6 expression in podocyte injury and proteinuric glomerular disease. Am. J. Pathol. 182, 1196–1204 (2013).

    CAS  PubMed  Google Scholar 

  209. Wang, X. X. et al. Vitamin D receptor agonist doxercalciferol modulates dietary fat-induced renal disease and renal lipid metabolism. Am. J. Physiol. Ren. Physiol. 300, F801–F810 (2011).

    CAS  Google Scholar 

  210. Garsen, M. et al. Vitamin D attenuates proteinuria by inhibition of heparanase expression in the podocyte. J. Pathol. 237, 472–481 (2015).

    CAS  PubMed  Google Scholar 

  211. Branisteanu, D. D., Leenaerts, P., van Damme, B. & Bouillon, R. Partial prevention of active Heymann nephritis by 1α, 25 dihydroxyvitamin D3. Clin. Exp. Immunol. 94, 412–417 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  212. Makibayashi, K. et al. A vitamin D analog ameliorates glomerular injury on rat glomerulonephritis. Am. J. Pathol. 158, 1733–1741 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Zou, M. S. et al. 1,25-Dihydroxyvitamin D3 decreases adriamycin-induced podocyte apoptosis and loss. Int. J. Med. Sci. 7, 290–299 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Zou, M. S. et al. 1,25-Dihydroxyvitamin D3 ameliorates podocytopenia in rats with adriamycin-induced nephropathy. Intern. Med. 49, 2677–2686 (2010).

    CAS  PubMed  Google Scholar 

  215. Schwarz, U. et al. Effect of 1,25 (OH)2 vitamin D3 on glomerulosclerosis in subtotally nephrectomized rats. Kidney Int. 53, 1696–1705 (1998).

    CAS  PubMed  Google Scholar 

  216. Sanchez-Nino, M. D. et al. Globotriaosylsphingosine actions on human glomerular podocytes: implications for Fabry nephropathy. Nephrol. Dial. Transplant. 26, 1797–1802 (2011).

    CAS  PubMed  Google Scholar 

  217. Agarwal, R. Vitamin D, proteinuria, diabetic nephropathy, and progression of CKD. Clin. J. Am. Soc. Nephrol. 4, 1523–1528 (2009).

    CAS  PubMed  Google Scholar 

  218. Chandel, N. et al. Vitamin D receptor deficit induces activation of renin angiotensin system via SIRT1 modulation in podocytes. Exp. Mol. Pathol. 102, 97–105 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  219. Chandel, N. et al. Epigenetic modulation of human podocyte vitamin D receptor in HIV milieu. J. Mol. Biol. 427, 3201–3215 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  220. Xu, L. et al. Vitamin D and its receptor regulate lipopolysaccharide-induced transforming growth factor-β, angiotensinogen expression and podocytes apoptosis through the nuclear factor-κB pathway. J. Diabetes Investig. 7, 680–688 (2016).

    PubMed  PubMed Central  Google Scholar 

  221. Cheng, X., Zhao, X., Khurana, S., Bruggeman, L. A. & Kao, H. Y. Microarray analyses of glucocorticoid and vitamin D3 target genes in differentiating cultured human podocytes. PLoS ONE 8, e60213 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  222. He, W., Kang, Y. S., Dai, C. & Liu, Y. Blockade of Wnt/β-catenin signaling by paricalcitol ameliorates proteinuria and kidney injury. J. Am. Soc. Nephrol. 22, 90–103 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  223. Khurana, S., Bruggeman, L. A. & Kao, H. Y. Nuclear hormone receptors in podocytes. Cell Biosci. 2, 33 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  224. Li, Y. C. Vitamin D receptor signaling in renal and cardiovascular protection. Semin. Nephrol. 33, 433–447 (2013).

    PubMed  PubMed Central  Google Scholar 

  225. de Borst, M. H. et al. Active vitamin D treatment for reduction of residual proteinuria: a systematic review. J. Am. Soc. Nephrol. 24, 1863–1871 (2013).

    PubMed  PubMed Central  Google Scholar 

  226. Melamed, M. L. & Thadhani, R. I. Vitamin D therapy in chronic kidney disease and end stage renal disease. Clin. J. Am. Soc. Nephrol. 7, 358–365 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  227. de Boer, I. H. et al. Effect of Vitamin D and omega-3 fatty acid supplementation on kidney function in patients with type 2 diabetes: a randomized clinical trial. JAMA 322, 1899–1909 (2019).

    PubMed  PubMed Central  Google Scholar 

  228. Fishbane, S. et al. Oral paricalcitol in the treatment of patients with CKD and proteinuria: a randomized trial. Am. J. Kidney Dis. 54, 647–652 (2009).

    CAS  PubMed  Google Scholar 

  229. Alborzi, P. et al. Paricalcitol reduces albuminuria and inflammation in chronic kidney disease: a randomized double-blind pilot trial. Hypertension 52, 249–255 (2008).

    CAS  PubMed  Google Scholar 

  230. de Zeeuw, D. et al. Selective vitamin D receptor activation with paricalcitol for reduction of albuminuria in patients with type 2 diabetes (VITAL study): a randomised controlled trial. Lancet 376, 1543–1551 (2010).

    PubMed  Google Scholar 

  231. Kramer, H., Berns, J. S., Choi, M. J., Martin, K. & Rocco, M. V. 25-Hydroxyvitamin D testing and supplementation in CKD: an NKF-KDOQI controversies report. Am. J. Kidney Dis. 64, 499–509 (2014).

    CAS  PubMed  Google Scholar 

  232. Le Menuet, D., Viengchareun, S., Muffat-Joly, M., Zennaro, M. C. & Lombes, M. Expression and function of the human mineralocorticoid receptor: lessons from transgenic mouse models. Mol. Cell. Endocrinol. 217, 127–136 (2004).

    PubMed  Google Scholar 

  233. Arriza, J. L. et al. Cloning of human mineralocorticoid receptor complementary DNA: structural and functional kinship with the glucocorticoid receptor. Science 237, 268–275 (1987).

    CAS  PubMed  Google Scholar 

  234. Funder, J. W., Pearce, P. T., Smith, R. & Smith, A. I. Mineralocorticoid action: target tissue specificity is enzyme, not receptor, mediated. Science 242, 583–585 (1988).

    CAS  PubMed  Google Scholar 

  235. Buonafine, M., Bonnard, B. & Jaisser, F. Mineralocorticoid receptor and cardiovascular disease. Am. J. Hypertens. 31, 1165–1174 (2018).

    CAS  PubMed  Google Scholar 

  236. Nishiyama, A. Pathophysiological mechanisms of mineralocorticoid receptor-dependent cardiovascular and chronic kidney disease. Hypertens. Res. 42, 293–300 (2019).

    CAS  PubMed  Google Scholar 

  237. Aldigier, J. C., Kanjanbuch, T., Ma, L. J., Brown, N. J. & Fogo, A. B. Regression of existing glomerulosclerosis by inhibition of aldosterone. J. Am. Soc. Nephrol. 16, 3306–3314 (2005).

    CAS  PubMed  Google Scholar 

  238. Fujihara, C. K. et al. A novel aldosterone antagonist limits renal injury in 5/6 nephrectomy. Sci. Rep. 7, 7899 (2017).

    PubMed  PubMed Central  Google Scholar 

  239. Zhou, X., Ono, H., Ono, Y. & Frohlich, E. D. Aldosterone antagonism ameliorates proteinuria and nephrosclerosis independent of glomerular dynamics in L-NAME/SHR model. Am. J. Nephrol. 24, 242–249 (2004).

    CAS  PubMed  Google Scholar 

  240. Zitt, E. et al. The selective mineralocorticoid receptor antagonist eplerenone is protective in mild anti-GBM glomeru-lonephritis. Int. J. Clin. Exp. Pathol. 4, 606–615 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  241. Qin, D. et al. Aldosterone mediates glomerular inflammation in experimental mesangial proliferative glomerulonephritis. J. Nephrol. 26, 199–206 (2013).

    CAS  PubMed  Google Scholar 

  242. Nagase, M. et al. Podocyte injury underlies the glomerulopathy of Dahl salt-hypertensive rats and is reversed by aldosterone blocker. Hypertension 47, 1084–1093 (2006).

    CAS  PubMed  Google Scholar 

  243. Shibata, S., Nagase, M., Yoshida, S., Kawachi, H. & Fujita, T. Podocyte as the target for aldosterone: roles of oxidative stress and Sgk1. Hypertension 49, 355–364 (2007).

    CAS  PubMed  Google Scholar 

  244. Chen, C. et al. Aldosterone induces apoptosis in rat podocytes: role of PI3-K/Akt and p38MAPK signaling pathways. Nephron Exp. Nephrol. 113, e26–e34 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  245. Nagase, M. et al. Enhanced aldosterone signaling in the early nephropathy of rats with metabolic syndrome: possible contribution of fat-derived factors. J. Am. Soc. Nephrol. 17, 3438–3446 (2006).

    CAS  PubMed  Google Scholar 

  246. Takagi, N. et al. Mineralocorticoid receptor blocker protects against podocyte-dependent glomerulosclerosis. Nephron Extra 2, 17–26 (2012).

    PubMed  PubMed Central  Google Scholar 

  247. Shibata, S. et al. Modification of mineralocorticoid receptor function by Rac1 GTPase: implication in proteinuric kidney disease. Nat. Med. 14, 1370–1376 (2008).

    CAS  PubMed  Google Scholar 

  248. Barrera-Chimal, J., Girerd, S. & Jaisser, F. Mineralocorticoid receptor antagonists and kidney diseases: pathophysiological basis. Kidney Int. 96, 302–319 (2019).

    CAS  PubMed  Google Scholar 

  249. Currie, G. et al. Effect of mineralocorticoid receptor antagonists on proteinuria and progression of chronic kidney disease: a systematic review and meta-analysis. BMC Nephrol. 17, 127 (2016).

    PubMed  PubMed Central  Google Scholar 

  250. Bolignano, D., Palmer, S. C., Navaneethan, S. D. & Strippoli, G. F. Aldosterone antagonists for preventing the progression of chronic kidney disease. Cochrane Database Syst. Rev. 4, CD007004 (2014).

    Google Scholar 

  251. Navaneethan, S. D., Nigwekar, S. U., Sehgal, A. R. & Strippoli, G. F. Aldosterone antagonists for preventing the progression of chronic kidney disease: a systematic review and meta-analysis. Clin. J. Am. Soc. Nephrol. 4, 542–551 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  252. Greene, E. L., Kren, S. & Hostetter, T. H. Role of aldosterone in the remnant kidney model in the rat. J. Clin. Invest. 98, 1063–1068 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  253. Rocha, R., Chander, P. N., Zuckerman, A. & Stier, C. T. Jr. Role of aldosterone in renal vascular injury in stroke-prone hypertensive rats. Hypertension 33, 232–237 (1999).

    CAS  PubMed  Google Scholar 

  254. Deroo, B. J. & Korach, K. S. Estrogen receptors and human disease. J. Clin. Invest. 116, 561–570 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  255. Kummer, S. et al. Estrogen receptor alpha expression in podocytes mediates protection against apoptosis in-vitro and in-vivo. PLoS ONE 6, e27457 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  256. Gross, M. L. et al. Beneficial effects of estrogens on indices of renal damage in uninephrectomized SHRsp rats. J. Am. Soc. Nephrol. 15, 348–358 (2004).

    CAS  PubMed  Google Scholar 

  257. Doublier, S. et al. Testosterone and 17β-estradiol have opposite effects on podocyte apoptosis that precedes glomerulosclerosis in female estrogen receptor knockout mice. Kidney Int. 79, 404–413 (2011).

    CAS  PubMed  Google Scholar 

  258. Gluhovschi, G. et al. Chronic kidney disease and the involvement of estrogen hormones in its pathogenesis and progression. Rom. J. Intern. Med. 50, 135–144 (2012).

    CAS  PubMed  Google Scholar 

  259. Carrero, J. J., Hecking, M., Chesnaye, N. C. & Jager, K. J. Sex and gender disparities in the epidemiology and outcomes of chronic kidney disease. Nat. Rev. Nephrol. 14, 151–164 (2018).

    PubMed  Google Scholar 

  260. Lee, W. L. et al. The benefits of estrogen or selective estrogen receptor modulator on kidney and its related disease–chronic kidney disease–mineral and bone disorder: osteoporosis. J. Chin. Med. Assoc. 76, 365–371 (2013).

    CAS  PubMed  Google Scholar 

  261. Melamed, M. L. et al. Raloxifene, a selective estrogen receptor modulator, is renoprotective: a post-hoc analysis. Kidney Int. 79, 241–249 (2011).

    CAS  PubMed  Google Scholar 

  262. Zhang, J. et al. DNA binding alters coactivator interaction surfaces of the intact VDR-RXR complex. Nat. Struct. Mol. Biol. 18, 556–563 (2011).

    PubMed  PubMed Central  Google Scholar 

  263. Weiss, K. et al. Effect of synthetic ligands of PPAR α, β/δ, γ, RAR, RXR and LXR on the fatty acid composition of phospholipids in mice. Lipids 46, 1013–1020 (2011).

    CAS  PubMed  Google Scholar 

  264. Trasino, S. E., Tang, X. H., Jessurun, J. & Gudas, L. J. Retinoic acid receptor β2 agonists restore glycaemic control in diabetes and reduce steatosis. Diabetes, Obes. Metab. 18, 142–151 (2016).

    CAS  Google Scholar 

  265. Ling, J. & Kumar, R. Crosstalk between NFkB and glucocorticoid signaling: a potential target of breast cancer therapy. Cancer Lett. 322, 119–126 (2012).

    CAS  PubMed  Google Scholar 

  266. Lu, T. C. et al. Retinoic acid utilizes CREB and USF1 in a transcriptional feed-forward loop in order to stimulate MKP1 expression in human immunodeficiency virus-infected podocytes. Mol. Cell Biol. 28, 5785–5794 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  267. Cohen, C. D. et al. Comparative promoter analysis allows de novo identification of specialized cell junction-associated proteins. Proc. Natl Acad. Sci. USA 103, 5682–5687 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  268. Guo, Y. et al. Podocyte-specific induction of kruppel-like factor 15 restores differentiation markers and attenuates kidney injury in proteinuric kidney disease. J. Am. Soc. Nephrol. 29, 2529–2545 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  269. Endlich, N., Nobiling, R., Kriz, W. & Endlich, K. Expression and signaling of parathyroid hormone-related protein in cultured podocytes. Exp. Nephrol. 9, 436–443 (2001).

    CAS  PubMed  Google Scholar 

  270. Endlich, N. & Endlich, K. cAMP pathway in podocytes. Microsc. Res. Tech. 57, 228–231 (2002).

    CAS  PubMed  Google Scholar 

  271. Azeloglu, E. U. et al. Interconnected network motifs control podocyte morphology and kidney function. Sci. Signal. 7, ra12 (2014).

    PubMed  PubMed Central  Google Scholar 

  272. Zhong, Y. et al. Roflumilast enhances the renal protective effects of retinoids in an HIV-1 transgenic mouse model of rapidly progressive renal failure. Kidney Int. 81, 856–864 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  273. Shibata, S. et al. Rac1 GTPase in rodent kidneys is essential for salt-sensitive hypertension via a mineralocorticoid receptor–dependent pathway. J. Clin. Invest. 121, 3233–3243 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  274. Wang, X. X., Jiang, T. & Levi, M. Nuclear hormone receptors in diabetic nephropathy. Nat. Rev. Nephrol. 6, 342–351 (2010).

    CAS  PubMed  Google Scholar 

  275. Yang, J., Zhou, Y. & Guan, Y. PPARγ as a therapeutic target in diabetic nephropathy and other renal diseases. Curr. Opin. Nephrol. Hypertens. 21, 97–105 (2012).

    PubMed  Google Scholar 

  276. Ishibashi, Y. et al. PEDF inhibits AGE-induced podocyte apoptosis via PPAR-gamma activation. Microvasc. Res. 85, 54–58 (2013).

    CAS  PubMed  Google Scholar 

  277. Lee, E. Y. et al. Peroxisome proliferator-activated receptor-δ activation ameliorates albuminuria by preventing nephrin loss and restoring podocyte integrity in type 2 diabetes. Nephrol. Dial. Transplant. 27, 4069–4079 (2012).

    CAS  PubMed  Google Scholar 

  278. Wang, Y. et al. Vitamin D receptor signaling in podocytes protects against diabetic nephropathy. J. Am. Soc. Nephrol. 23, 1977–1986 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  279. Guo, J. et al. GSK-3β and vitamin D receptor are involved in β-catenin and Snail signaling in high glucose-induced epithelial-mesenchymal transition of mouse podocytes. Cell Physiol. Biochem. 33, 1087–1096 (2014).

    CAS  PubMed  Google Scholar 

  280. Verouti, S. N. et al. Vitamin D receptor activators upregulate and rescue podocalyxin expression in high glucose-treated human podocytes. Nephron Exp. Nephrol. 122, 36–50 (2012).

    CAS  PubMed  Google Scholar 

  281. Guo, J. et al. VDR activation reduces proteinuria and high-glucose-induced injury of kidneys and podocytes by regulating Wnt signaling pathway. Cell Physiol. Biochem. 43, 39–51 (2017).

    CAS  PubMed  Google Scholar 

  282. Toyonaga, J. et al. Spironolactone inhibits hyperglycemia-induced podocyte injury by attenuating ROS production. Nephrol. Dial. Transplant. 26, 2475–2484 (2011).

    CAS  PubMed  Google Scholar 

  283. Han, S. Y. et al. Spironolactone prevents diabetic nephropathy through an anti-inflammatory mechanism in type 2 diabetic rats. J. Am. Soc. Nephrol. 17, 1362–1372 (2006).

    CAS  PubMed  Google Scholar 

  284. Zhou, G., Johansson, U., Peng, X. R., Bamberg, K. & Huang, Y. An additive effect of eplerenone to ACE inhibitor on slowing the progression of diabetic nephropathy in the db/db mice. Am. J. Transl. Res. 8, 1339–1354 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  285. Nishiyama, A. et al. Mineralocorticoid receptor blockade enhances the antiproteinuric effect of an angiotensin II blocker through inhibiting podocyte injury in type 2 diabetic rats. J. Pharmacol. Exp. Ther. 332, 1072–1080 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  286. Schjoedt, K. J. et al. Beneficial impact of spironolactone on nephrotic range albuminuria in diabetic nephropathy. Kidney Int. 70, 536–542 (2006).

    CAS  PubMed  Google Scholar 

  287. Mehdi, U. F., Adams-Huet, B., Raskin, P., Vega, G. L. & Toto, R. D. Addition of angiotensin receptor blockade or mineralocorticoid antagonism to maximal angiotensin-converting enzyme inhibition in diabetic nephropathy. J. Am. Soc. Nephrol. 20, 2641–2650 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  288. Bakris, G. L. et al. Effect of finerenone on albuminuria in patients with diabetic nephropathy: a randomized clinical trial. JAMA 314, 884–894 (2015).

    CAS  PubMed  Google Scholar 

  289. Doublier, S., Lupia, E., Catanuto, P. & Elliot, S. J. Estrogens and progression of diabetic kidney damage. Curr. Diabetes Rev. 7, 28–34 (2011).

    CAS  PubMed  Google Scholar 

  290. Han, S. Y. et al. Effect of retinoic acid in experimental diabetic nephropathy. Immunol. Cell Biol. 82, 568–576 (2004).

    CAS  PubMed  Google Scholar 

  291. Wang, X. X. et al. FXR/TGR5 dual agonist prevents progression of nephropathy in diabetes and obesity. J. Am. Soc. Nephrol. 29, 118–137 (2018).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors’ work is supported by funding from the American Heart Association Career Development Award (18CDA34110287) and by the Nationwide Children’s Hospital to S.A.; VA Merit Award IBX000345C, NIH 1R01DK088541 and NIH P01DK56492 to J.C.H.; and by Institut National de Santé et de la Recherche Médicale (INSERM) and research grants from the European Research Council under the European Union’s Seventh Framework Programme (FP7/2007-2013)/ ERC grant agreement no. 107037) and SWITCHES grant (ANR12-BSV1-0039-03) from l’Agence Nationale de la Recherche (ANR) of France to P.-L.T.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to researching data, and writing, reviewing and editing the article. S.A. focused on GR, PPARγ, ER and VDR, and drafted the figures. P.-L.T. focused on PPARγ and MR, and J.C.H. focused on RAR and RXR.

Corresponding author

Correspondence to Shipra Agrawal.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Nephrology thanks P. Brinkkoetter, C. Faul and M. Moeller for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Glossary

Co-regulators

Co-activators or co-repressors that bind to a nuclear receptor in the absence or presence of ligands enabling activation or repression of gene transcription.

Sumoylation

A reversible post-translational protein modification that consists of the covalent labelling of a small protein called SUMO to lysine residues of selected target proteins. Sumoylation is a well-characterized regulator of nuclear functions.

Transactivation

The most common mechanism of nuclear receptor action that involves direct binding of the nuclear receptor to a DNA hormone response element.

Transrepression

The process by which nuclear receptors bind to and deactivate other transcription factors; for example, the glucocorticoid receptor and PPARγ can suppress target genes by inhibiting the activities of other transcription factors, such as AP-1, in a ligand-dependent manner; transrepression does not require the nuclear receptor to bind DNA.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Agrawal, S., He, J.C. & Tharaux, PL. Nuclear receptors in podocyte biology and glomerular disease. Nat Rev Nephrol 17, 185–204 (2021). https://doi.org/10.1038/s41581-020-00339-6

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41581-020-00339-6

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research