Genetic and Physiological Effects of Insulin-Like Growth Factor-1 (IGF-1) on Human Urate Homeostasis

Significance Statement Hyperinsulinemia induces hyperuricemia by activating net renal urate reabsorption in the renal proximal tubule. The basolateral reabsorptive urate transporter GLUT9a appears to be the dominant target for insulin. By contrast, IGF-1 infusion reduces serum urate (SU), through mechanisms unknown. Genetic variants of IGF1R associated with reduced SU have increased IGF-1R expression and interact with genes encoding the GLUT9 and ABCG2 urate transporters, in a sex-specific fashion, which controls the SU level. Activation of IGF-1/IGF-1R signaling in Xenopus oocytes modestly activates GLUT9a and inhibits insulin's stimulatory effect on the transporter, which also activates multiple secretory urate transporters—ABCG2, ABCC4, OAT1, and OAT3. The results collectively suggest that IGF-1 reduces SU by activating secretory urate transporters and inhibiting insulin's action on GLUT9a. Background Metabolic syndrome and hyperinsulinemia are associated with hyperuricemia. Insulin infusion in healthy volunteers elevates serum urate (SU) by activating net urate reabsorption in the renal proximal tubule, whereas IGF-1 infusion reduces SU by mechanisms unknown. Variation within the IGF1R gene also affects SU levels. Methods Colocalization analyses of a SU genome-wide association studies signal at IGF1R and expression quantitative trait loci signals in cis using COLOC2, RT-PCR, Western blotting, and urate transport assays in transfected HEK 293T cells and in Xenopus laevis oocytes. Results Genetic association at IGF1R with SU is stronger in women and is mediated by control of IGF1R expression. Inheritance of the urate-lowering homozygous genotype at the SLC2A9 locus is associated with a differential effect of IGF1R genotype between men and women. IGF-1, through IGF-1R, stimulated urate uptake in human renal proximal tubule epithelial cells and transfected HEK 293T cells, through activation of IRS1, PI3/Akt, MEK/ERK, and p38 MAPK; urate uptake was inhibited in the presence of uricosuric drugs, specific inhibitors of protein tyrosine kinase, PI3 kinase (PI3K), ERK, and p38 MAPK. In X. laevis oocytes expressing ten individual urate transporters, IGF-1 through endogenous IGF-1R stimulated urate transport mediated by GLUT9, OAT1, OAT3, ABCG2, and ABCC4 and inhibited insulin's stimulatory action on GLUT9a and OAT3. IGF-1 significantly activated Akt and ERK. Specific inhibitors of PI3K, ERK, and PKC significantly affected IGF-1 stimulation of urate transport in oocytes. Conclusions The combined results of infusion, genetics, and transport experiments suggest that IGF-1 reduces SU by activating urate secretory transporters and inhibiting insulin's action.

[1]  T. Merriman,et al.  Genetic and Physiological Effects of Insulin on Human Urate Homeostasis , 2021, Frontiers in Physiology.

[2]  B. Ban,et al.  Association Between Uric Acid and Insulin-Like Growth Factor-1 in Type 2 Diabetes Mellitus , 2021, International journal of general medicine.

[3]  M. Mann,et al.  Distinct signaling by insulin and IGF-1 receptors and their extra- and intracellular domains , 2021, Proceedings of the National Academy of Sciences.

[4]  G. You,et al.  Regulation of organic anion transporters: Role in physiology, pathophysiology, and drug elimination. , 2020, Pharmacology & therapeutics.

[5]  T. Merriman,et al.  The ABCG2 Q141K hyperuricemia and gout associated variant illuminates the physiology of human urate excretion , 2020, Nature Communications.

[6]  N. Osada,et al.  Dysfunctional missense variant of OAT10/SLC22A13 decreases gout risk and serum uric acid levels , 2019, Annals of the rheumatic diseases.

[7]  D. Mount,et al.  Interaction Between ITM2B and GLUT9 Links Urate Transport to Neurodegenerative Disorders , 2019, Front. Physiol..

[8]  Y. Okada,et al.  Genomic dissection of 43 serum urate-associated loci provides multiple insights into molecular mechanisms of urate control , 2019, bioRxiv.

[9]  G. You,et al.  Insulin-like growth factor 1 modulates the phosphorylation, expression, and activity of organic anion transporter 3 through protein kinase A signaling pathway , 2019, Acta pharmaceutica Sinica. B.

[10]  G. Sesti,et al.  The polymorphism rs35767 at IGF1 locus is associated with serum urate levels , 2018, Scientific Reports.

[11]  T. Merriman,et al.  An update on the genetics of hyperuricaemia and gout , 2018, Nature Reviews Rheumatology.

[12]  C. Duan,et al.  IGF-Binding Proteins: Why Do They Exist and Why Are There So Many? , 2018, Front. Endocrinol..

[13]  A. Goumenou,et al.  Cetuximab Prevents Methotrexate-Induced Cytotoxicity in Vitro through Epidermal Growth Factor Dependent Regulation of Renal Drug Transporters , 2017, Molecular pharmaceutics.

[14]  D. Mount,et al.  Uricosuric targets of tranilast , 2017, Pharmacology research & perspectives.

[15]  Janina M. Jeff,et al.  Analysis of ABCG2 and other urate transporters in uric acid homeostasis in chronic kidney disease: potential role of remote sensing and signaling , 2016, Clinical kidney journal.

[16]  D. Mount,et al.  The molecular physiology of uric acid homeostasis. , 2015, Annual review of physiology.

[17]  G. Sesti,et al.  Low circulating insulin-like growth factor-1 levels are associated with high serum uric acid in nondiabetic adult subjects. , 2014, Nutrition, metabolism, and cardiovascular diseases : NMCD.

[18]  A. V. Rij,et al.  Twenty-eight loci that influence serum urate levels: analysis of association with gout , 2014, Annals of the rheumatic diseases.

[19]  A. Schürmann,et al.  Early-onset metabolic syndrome in mice lacking the intestinal uric acid transporter SLC2A9 , 2014, Nature Communications.

[20]  K. Yan,et al.  Expression of SLC2A9 Isoforms in the Kidney and Their Localization in Polarized Epithelial Cells , 2014, PloS one.

[21]  C. Wallace,et al.  Bayesian Test for Colocalisation between Pairs of Genetic Association Studies Using Summary Statistics , 2013, PLoS genetics.

[22]  Fabian J Theis,et al.  Genome-wide association analyses identify 18 new loci associated with serum urate concentrations , 2012, Nature Genetics.

[23]  C. Cheeseman,et al.  Human SLC2A9a and SLC2A9b isoforms mediate electrogenic transport of urate with different characteristics in the presence of hexoses. , 2012, American journal of physiology. Renal physiology.

[24]  T. Nakanishi,et al.  Extra-Renal Elimination of Uric Acid via Intestinal Efflux Transporter BCRP/ABCG2 , 2012, PloS one.

[25]  Y. Oh The insulin-like growth factor system in chronic kidney disease: Pathophysiology and therapeutic opportunities , 2012, Kidney research and clinical practice.

[26]  Y. Shinohara,et al.  Decreased extra-renal urate excretion is a common cause of hyperuricemia , 2012, Nature Communications.

[27]  N. Shinomiya,et al.  Identification of ABCG2 Dysfunction as a Major Factor Contributing to Gout , 2011, Nucleosides, nucleotides & nucleic acids.

[28]  Tadashi Yamamoto,et al.  Expression and localization of insulin‐like growth factor binding proteins in normal and proteinuric kidney glomeruli , 2010, Nephrology.

[29]  M. Hiasa,et al.  Type 1 Sodium-dependent Phosphate Transporter (SLC17A1 Protein) Is a Cl−-dependent Urate Exporter* , 2010, The Journal of Biological Chemistry.

[30]  K. Geering,et al.  Mouse GLUT9: evidences for a urate uniporter. , 2009, American journal of physiology. Renal physiology.

[31]  E. Boerwinkle,et al.  Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout , 2009, Proceedings of the National Academy of Sciences.

[32]  S. Barros,et al.  Activation of Protein Kinase Cζ Increases OAT1 (SLC22A6)- and OAT3 (SLC22A8)-mediated Transport* , 2009, Journal of Biological Chemistry.

[33]  I. Hisatome,et al.  Plasma Urate Level Is Directly Regulated by a Voltage-driven Urate Efflux Transporter URATv1 (SLC2A9) in Humans* , 2008, Journal of Biological Chemistry.

[34]  P. Elliott,et al.  SLC2A9 Is a High-Capacity Urate Transporter in Humans , 2008, PLoS medicine.

[35]  A. Ashworth,et al.  Identification of genetic variants that influence circulating IGF1 levels: a targeted search strategy. , 2008, Human molecular genetics.

[36]  F. Russel,et al.  The breast cancer resistance protein transporter ABCG2 is expressed in the human kidney proximal tubule apical membrane. , 2008, Kidney international.

[37]  T. Litman,et al.  Localization of the ABCG2 mitoxantrone resistance-associated protein in normal tissues. , 2006, Cancer letters.

[38]  J. Chang-Claude,et al.  Polymorphisms of genes coding for insulin-like growth factor 1 and its major binding proteins, circulating levels of IGF-I and IGFBP-3 and breast cancer risk: results from the EPIC study , 2006, British Journal of Cancer.

[39]  S. Wright,et al.  Involvement of tyrosine kinase and PI3K in the regulation of OAT3-mediated estrone sulfate transport in isolated rabbit renal proximal tubules. , 2005, American journal of physiology. Renal physiology.

[40]  E. Henriksen,et al.  Enhanced insulin action on glucose transport and insulin signaling in 7-day unweighted rat soleus muscle. , 2004, Journal of applied physiology.

[41]  J. Kim,et al.  Identification of a Novel Voltage-driven Organic Anion Transporter Present at Apical Membrane of Renal Proximal Tubule* , 2003, Journal of Biological Chemistry.

[42]  Xiao‐ping Yang,et al.  Organic anion transporter 3 (Slc22a8) is a dicarboxylate exchanger indirectly coupled to the Na+ gradient. , 2003, American journal of physiology. Renal physiology.

[43]  Hirotaka Matsuo,et al.  Molecular identification of a renal urate–anion exchanger that regulates blood urate levels , 2002, Nature.

[44]  C. Ward,et al.  Structure and function of the type 1 insulin-like growth factor receptor , 2000, Cellular and Molecular Life Sciences CMLS.

[45]  D. Leroith,et al.  Insulin-Like Growth Factor I Is Essential for Postnatal Growth in Response to Growth Hormone , 1999 .

[46]  K. Siddle,et al.  Phosphoinositide 3-kinase: the key switch mechanism in insulin signalling. , 1998, The Biochemical journal.

[47]  L. Zhu,et al.  Molecular cloning and characterization of Xenopus insulin-like growth factor-1 receptor: its role in mediating insulin-induced Xenopus oocyte maturation and expression during embryogenesis. , 1998, Endocrinology.

[48]  R. Bigazzi,et al.  Effect of insulin on renal sodium and uric acid handling in essential hypertension. , 1996, American journal of hypertension.

[49]  D. Leroith,et al.  The Role of the Tyrosine Kinase Domain of the Insulin-like Growth Factor-I Receptor in Intracellular Signaling, Cellular Proliferation, and Tumorigenesis (*) , 1995, The Journal of Biological Chemistry.

[50]  C T Roberts,et al.  Molecular and cellular aspects of the insulin-like growth factor I receptor. , 1995, Endocrine reviews.

[51]  J. Tytgat,et al.  Subunit stoichiometry of a mammalian K+ channel determined by construction of multimeric cDNAs , 1992, Neuron.

[52]  A. Shuldiner,et al.  Genes encoding receptors for insulin and insulin-like growth factor I are expressed in Xenopus oocytes and embryos. , 1991, Proceedings of the National Academy of Sciences of the United States of America.

[53]  M. Lane,et al.  The insulin-like growth factor 1 (IGF-1) receptor is responsible for mediating the effects of insulin, IGF-1, and IGF-2 in Xenopus laevis oocytes. , 1991, The Journal of biological chemistry.

[54]  E. Van Obberghen,et al.  Insulin and insulin-like-growth-factor-I (IGF-I) receptors in Xenopus laevis oocytes. Comparison with insulin receptors from liver and muscle. , 1991, The Biochemical journal.

[55]  E. Krebs,et al.  Microinjection of a protein-tyrosine-phosphatase inhibits insulin action in Xenopus oocytes. , 1990, Proceedings of the National Academy of Sciences of the United States of America.

[56]  E. Krebs,et al.  Insulin and progesterone activate a common synthetic ribosomal protein S6 peptide kinase in Xenopus oocytes , 1988, FEBS letters.

[57]  C Collins,et al.  Insulin‐like growth factor I receptor primary structure: comparison with insulin receptor suggests structural determinants that define functional specificity. , 1986, The EMBO journal.

[58]  P. H. Seeburg,et al.  Human insulin receptor and its relationship to the tyrosine kinase family of oncogenes , 1985, Nature.

[59]  Yuan Zhang,et al.  Insulin-like growth factor-1 receptor-mediated inhibition of A-type K(+) current induces sensory neuronal hyperexcitability through the phosphatidylinositol 3-kinase and extracellular signal-regulated kinase 1/2 pathways, independently of Akt. , 2014, Endocrinology.

[60]  William A. Richardson,et al.  Homozygous SLC2A9 mutations cause severe renal hypouricemia. , 2010, Journal of the American Society of Nephrology : JASN.

[61]  H. Saito Pathophysiological regulation of renal SLC22A organic ion transporters in acute kidney injury: pharmacological and toxicological implications. , 2010, Pharmacology & therapeutics.

[62]  William A. Richardson,et al.  SLC2A9 is a newly identified urate transporter influencing serum urate concentration, urate excretion and gout , 2008, Nature Genetics.

[63]  H. Werner,et al.  Similarities and differences between insulin and IGF-I: Structures, receptors, and signalling pathways , 2008, Archives of Physiology and Biochemistry.

[64]  C. Gieger,et al.  SLC2A9 influences uric acid concentrations with pronounced sex-specific effects , 2008, Nature Genetics.

[65]  M. Resnick,et al.  Growth, immortalization, and differentiation potential of normal adult human proximal tubule cells , 2004, In Vitro Cellular & Developmental Biology - Animal.

[66]  J. T. ter Maaten,et al.  Renal handling of urate and sodium during acute physiological hyperinsulinaemia in healthy subjects. , 1997, Clinical science.

[67]  E. Ferrannini,et al.  Effect of insulin on uric acid excretion in humans. , 1995, The American journal of physiology.

[68]  E. Renard,et al.  Insulin and IGF-1 signaling in oocyte maturation. , 1994, Hormone research.

[69]  M. Czech,et al.  The Insulinlike Growth Factor Receptors , 1986 .

[70]  M. Rechler,et al.  The nature and regulation of the receptors for insulin-like growth factors. , 1985, Annual review of physiology.

[71]  P. Pilch,et al.  Stimulation of tyrosine-specific phosphorylation in vitro by insulin-like growth factor I , 1983, Nature.