Diabetes Metab J.  2014 Feb;38(1):13-24. 10.4093/dmj.2014.38.1.13.

The Hijacking of Cellular Signaling and the Diabetes Epidemic: Mechanisms of Environmental Disruption of Insulin Action and Glucose Homeostasis

Affiliations
  • 1Committee on Molecular Metabolism and Nutrition, Kovler Diabetes Center, Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Chicago, Chicago, IL, USA. rsargis@medicine.bsd.uchicago.edu

Abstract

The burgeoning epidemic of metabolic disease causes significant societal and individual morbidity and threatens the stability of health care systems around the globe. Efforts to understand the factors that contribute to metabolic derangements are critical for reversing these troubling trends. While excess caloric consumption and physical inactivity superimposed on a susceptible genetic background are central drivers of this crisis, these factors alone fail to fully account for the magnitude and rapidity with which metabolic diseases have increased in prevalence worldwide. Recent epidemiological evidence implicates endocrine disrupting chemicals in the pathogenesis of metabolic diseases. These compounds represent a diverse array of chemicals to which humans are exposed via multiple routes in adulthood and during development. Furthermore, a growing ensemble of animal- and cell-based studies provides preclinical evidence supporting the hypothesis that environmental contaminants contribute to the development of metabolic diseases, including diabetes. Herein are reviewed studies linking specific endocrine disruptors to impairments in glucose homeostasis as well as tying these compounds to disturbances in insulin secretion and impairments in insulin signal transduction. While the data remains somewhat incomplete, the current body of evidence supports the hypothesis that our chemically polluted environment may play a contributing role in the current metabolic crisis.

Keyword

Diabetes mellitus; Endocrine disrupting chemicals; Endocrine disruptors; Insulin; Insulin resistance; Insulin signaling; Pollution

MeSH Terms

Delivery of Health Care
Diabetes Mellitus
Endocrine Disruptors
Glucose*
Homeostasis*
Humans
Insulin Resistance
Insulin*
Metabolic Diseases
Prevalence
Signal Transduction
Endocrine Disruptors
Glucose
Insulin

Figure

  • Fig. 1 Insulin signaling targets of endocrine disrupting chemicals (EDCs). Multiple studies have examined the effects of EDCs on various aspects of insulin synthesis, release, and cellular action. The molecular targets identified from these various studies are summarized. Of note, this figure synthesizes data from various model systems, including multiple different targets of insulin action (i.e., adipose tissue, liver, and muscle). The data has been combined for clarity but should not be understood to mean that the EDCs shown exert similar effects in all biological tissues or in all species. TCDD, 2,3,7,8-tetrachlorodibenzodioxin; BPA, bisphenol A; DEHP, diethylhexylphthalate; PCB, polychlorinated biphenyl; PDK, phosphoinositide-dependent kinase; mTORC2, mammalian target of rapamycin complex 2; POP, persistent organic pollutant; GLUT4, glucose transporter 4.

  • Fig. 2 Endocrine disrupting chemical (EDC) modulation of glucocorticoid activity. The effects of multiple studies examining the effects of EDCs on the glucocorticoid signaling pathway are summarized, including effects at various concentrations and in different model systems. As such, these studies should not be interpreted to mean that each EDCs operates similarly in all tissues; however, pooling of the data suggests potential mechanisms of synergy among various EDCs that modulate activity of this signaling pathway. 11β-HSD, 11β-hydroxysteroid dehydrogenase; BPA, bisphenol A; PCB, polychlorinated biphenyl; GR-GRE, glucocorticoid receptor-glucocorticoid response element.


Cited by  1 articles

Association between Blood Mercury Level and Visceral Adiposity in Adults
Jong Suk Park, Kyoung Hwa Ha, Ka He, Dae Jung Kim
Diabetes Metab J. 2017;41(2):113-120.    doi: 10.4093/dmj.2017.41.2.113.


Reference

1. International Diabetes Federation: Diabetes atlas. updated 2013 Nov 15. Available from: http://www.idf.org/diabetesatlas.
2. American Diabetes Association. Economic costs of diabetes in the U.S. in 2012. Diabetes Care. 2013; 36:1033–1046.
3. Neel BA, Sargis RM. The paradox of progress: environmental disruption of metabolism and the diabetes epidemic. Diabetes. 2011; 60:1838–1848.
4. Kuo CC, Moon K, Thayer KA, Navas-Acien A. Environmental chemicals and type 2 diabetes: an updated systematic review of the epidemiologic evidence. Curr Diab Rep. 2013; 13:831–849.
5. Regnier SM, Sargis RM. Adipocytes under assault: environmental disruption of adipose physiology. Biochim Biophys Acta. 2014; 1842:520–533.
6. Hill DS, Wlodarczyk BJ, Mitchell LE, Finnell RH. Arsenate-induced maternal glucose intolerance and neural tube defects in a mouse model. Toxicol Appl Pharmacol. 2009; 239:29–36.
7. Matsui H, Wada O, Manabe S, Ushijima Y, Fujikura T. Species difference in sensitivity to the diabetogenic action of triphenyltin hydroxide. Experientia. 1984; 40:377–378.
8. Ogino K, Inukai T, Miura Y, Matsui H, Takemura Y. Triphenyltin chloride induces glucose intolerance by the suppression of insulin release from hamster pancreatic beta-cells. Exp Clin Endocrinol Diabetes. 1996; 104:409–411.
9. Martinelli MI, Mocchiutti NO, Bernal CA. Dietary di(2-ethylhexyl) phthalate-impaired glucose metabolism in experimental animals. Hum Exp Toxicol. 2006; 25:531–538.
10. Baker NA, Karounos M, English V, Fang J, Wei Y, Stromberg A, Sunkara M, Morris AJ, Swanson HI, Cassis LA. Coplanar polychlorinated biphenyls impair glucose homeostasis in lean C57BL/6 mice and mitigate beneficial effects of weight loss on glucose homeostasis in obese mice. Environ Health Perspect. 2013; 121:105–110.
11. Paul DS, Walton FS, Saunders RJ, Styblo M. Characterization of the impaired glucose homeostasis produced in C57BL/6 mice by chronic exposure to arsenic and high-fat diet. Environ Health Perspect. 2011; 119:1104–1109.
12. Alonso-Magdalena P, Vieira E, Soriano S, Menes L, Burks D, Quesada I, Nadal A. Bisphenol A exposure during pregnancy disrupts glucose homeostasis in mothers and adult male offspring. Environ Health Perspect. 2010; 118:1243–1250.
13. Nash JT, Szabo DT, Carey GB. Polybrominated diphenyl ethers alter hepatic phosphoenolpyruvate carboxykinase enzyme kinetics in male Wistar rats: implications for lipid and glucose metabolism. J Toxicol Environ Health A. 2013; 76:142–156.
14. Ruzzin J, Petersen R, Meugnier E, Madsen L, Lock EJ, Lillefosse H, Ma T, Pesenti S, Sonne SB, Marstrand TT, Malde MK, Du ZY, Chavey C, Fajas L, Lundebye AK, Brand CL, Vidal H, Kristiansen K, Froyland L. Persistent organic pollutant exposure leads to insulin resistance syndrome. Environ Health Perspect. 2010; 118:465–471.
15. Yan YH, Chou CC, Lee CT, Liu JY, Cheng TJ. Enhanced insulin resistance in diet-induced obese rats exposed to fine particles by instillation. Inhal Toxicol. 2011; 23:507–519.
16. Miyawaki J, Sakayama K, Kato H, Yamamoto H, Masuno H. Perinatal and postnatal exposure to bisphenol a increases adipose tissue mass and serum cholesterol level in mice. J Atheroscler Thromb. 2007; 14:245–252.
17. Sakurai T, Miyazawa S, Hashimoto T. Effects of di-(2-ethylhexyl) phthalate administration on carbohydrate and fatty acid metabolism in rat liver. J Biochem. 1978; 83:313–320.
18. Feige JN, Gerber A, Casals-Casas C, Yang Q, Winkler C, Bedu E, Bueno M, Gelman L, Auwerx J, Gonzalez FJ, Desvergne B. The pollutant diethylhexyl phthalate regulates hepatic energy metabolism via species-specific PPARalpha-dependent mechanisms. Environ Health Perspect. 2010; 118:234–241.
19. Seefeld MD, Corbett SW, Keesey RE, Peterson RE. Characterization of the wasting syndrome in rats treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Appl Pharmacol. 1984; 73:311–322.
20. Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006; 444:840–846.
21. Pont A, Rubino JM, Bishop D, Peal R. Diabetes mellitus and neuropathy following Vacor ingestion in man. Arch Intern Med. 1979; 139:185–187.
22. Karam JH, Lewitt PA, Young CW, Nowlain RE, Frankel BJ, Fujiya H, Freedman ZR, Grodsky GM. Insulinopenic diabetes after rodenticide (Vacor) ingestion: a unique model of acquired diabetes in man. Diabetes. 1980; 29:971–978.
23. Kurita H, Yoshioka W, Nishimura N, Kubota N, Kadowaki T, Tohyama C. Aryl hydrocarbon receptor-mediated effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on glucose-stimulated insulin secretion in mice. J Appl Toxicol. 2009; 29:689–694.
24. Novelli M, Piaggi S, De Tata V. 2,3,7,8-Tetrachlorodibenzo-p-dioxin-induced impairment of glucose-stimulated insulin secretion in isolated rat pancreatic islets. Toxicol Lett. 2005; 156:307–314.
25. Piaggi S, Novelli M, Martino L, Masini M, Raggi C, Orciuolo E, Masiello P, Casini A, De Tata V. Cell death and impairment of glucose-stimulated insulin secretion induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the beta-cell line INS-1E. Toxicol Appl Pharmacol. 2007; 220:333–340.
26. Ebner K, Matsumura F, Enan E, Olsen H. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) alters pancreatic membrane tyrosine phosphorylation following acute treatment. J Biochem Toxicol. 1993; 8:71–81.
27. Wassermann D, Wassermann M, Lemesch C. Ultrastructure of beta-cells of the endocrine pancreas in rats receiving polychlorinated biphenyls. Environ Physiol Biochem. 1975; 5:322–340.
28. Douillet C, Currier J, Saunders J, Bodnar WM, Matousek T, Styblo M. Methylated trivalent arsenicals are potent inhibitors of glucose stimulated insulin secretion by murine pancreatic islets. Toxicol Appl Pharmacol. 2013; 267:11–15.
29. Fu J, Woods CG, Yehuda-Shnaidman E, Zhang Q, Wong V, Collins S, Sun G, Andersen ME, Pi J. Low-level arsenic impairs glucose-stimulated insulin secretion in pancreatic beta cells: involvement of cellular adaptive response to oxidative stress. Environ Health Perspect. 2010; 118:864–870.
30. Diaz-Villasenor A, Burns AL, Salazar AM, Sordo M, Hiriart M, Cebrian ME, Ostrosky-Wegman P. Arsenite reduces insulin secretion in rat pancreatic beta-cells by decreasing the calcium-dependent calpain-10 proteolysis of SNAP-25. Toxicol Appl Pharmacol. 2008; 231:291–299.
31. El Muayed M, Raja MR, Zhang X, MacRenaris KW, Bhatt S, Chen X, Urbanek M, O'Halloran TV, Lowe WL Jr. Accumulation of cadmium in insulin-producing β cells. Islets. 2012; 4:405–416.
32. Chang KC, Hsu CC, Liu SH, Su CC, Yen CC, Lee MJ, Chen KL, Ho TJ, Hung DZ, Wu CC, Lu TH, Su YC, Chen YW, Huang CF. Cadmium induces apoptosis in pancreatic β-cells through a mitochondria-dependent pathway: the role of oxidative stress-mediated c-Jun N-terminal kinase activation. PLoS One. 2013; 8:e54374.
33. Chen YW, Huang CF, Yang CY, Yen CC, Tsai KS, Liu SH. Inorganic mercury causes pancreatic beta-cell death via the oxidative stress-induced apoptotic and necrotic pathways. Toxicol Appl Pharmacol. 2010; 243:323–331.
34. Alonso-Magdalena P, Morimoto S, Ripoll C, Fuentes E, Nadal A. The estrogenic effect of bisphenol A disrupts pancreatic beta-cell function in vivo and induces insulin resistance. Environ Health Perspect. 2006; 114:106–112.
35. Fischer LJ, Zhou HR, Wagner MA. Polychlorinated biphenyls release insulin from RINm5F cells. Life Sci. 1996; 59:2041–2049.
36. Quesada I, Fuentes E, Viso-Leon MC, Soria B, Ripoll C, Nadal A. Low doses of the endocrine disruptor bisphenol-A and the native hormone 17beta-estradiol rapidly activate transcription factor CREB. FASEB J. 2002; 16:1671–1673.
37. Miura Y, Matsui H. Triphenyltin impairs a protein kinase A (PKA)-dependent increase of cytosolic Na+ and Ca2+ and PKA-independent increase of cytosolic Ca2+ associated with insulin secretion in hamster pancreatic beta-cells. Toxicol Appl Pharmacol. 2006; 216:363–372.
38. Fischer LJ, Wagner MA, Madhukar BV. Potential involvement of calcium, CaM kinase II, and MAP kinases in PCB-stimulated insulin release from RINm5F cells. Toxicol Appl Pharmacol. 1999; 159:194–203.
39. Kim YH, Shim YJ, Shin YJ, Sul D, Lee E, Min BH. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces calcium influx through T-type calcium channel and enhances lysosomal exocytosis and insulin secretion in INS-1 cells. Int J Toxicol. 2009; 28:151–161.
40. Soriano S, Alonso-Magdalena P, Garcia-Arevalo M, Novials A, Muhammed SJ, Salehi A, Gustafsson JA, Quesada I, Nadal A. Rapid insulinotropic action of low doses of bisphenol-A on mouse and human islets of Langerhans: role of estrogen receptor β. PLoS One. 2012; 7:e31109.
41. Alonso-Magdalena P, Laribi O, Ropero AB, Fuentes E, Ripoll C, Soria B, Nadal A. Low doses of bisphenol A and diethylstilbestrol impair Ca2+ signals in pancreatic alpha-cells through a nonclassical membrane estrogen receptor within intact islets of Langerhans. Environ Health Perspect. 2005; 113:969–977.
42. Grun F, Blumberg B. Minireview: the case for obesogens. Mol Endocrinol. 2009; 23:1127–1134.
43. Kanayama T, Kobayashi N, Mamiya S, Nakanishi T, Nishikawa J. Organotin compounds promote adipocyte differentiation as agonists of the peroxisome proliferator-activated receptor gamma/retinoid X receptor pathway. Mol Pharmacol. 2005; 67:766–774.
44. Grun F, Watanabe H, Zamanian Z, Maeda L, Arima K, Cubacha R, Gardiner DM, Kanno J, Iguchi T, Blumberg B. Endocrine-disrupting organotin compounds are potent inducers of adipogenesis in vertebrates. Mol Endocrinol. 2006; 20:2141–2155.
45. Masuno H, Iwanami J, Kidani T, Sakayama K, Honda K. Bisphenol a accelerates terminal differentiation of 3T3-L1 cells into adipocytes through the phosphatidylinositol 3-kinase pathway. Toxicol Sci. 2005; 84:319–327.
46. Sargis RM, Johnson DN, Choudhury RA, Brady MJ. Environmental endocrine disruptors promote adipogenesis in the 3T3-L1 cell line through glucocorticoid receptor activation. Obesity (Silver Spring). 2010; 18:1283–1288.
47. Arsenescu V, Arsenescu RI, King V, Swanson H, Cassis LA. Polychlorinated biphenyl-77 induces adipocyte differentiation and proinflammatory adipokines and promotes obesity and atherosclerosis. Environ Health Perspect. 2008; 116:761–768.
48. Li X, Pham HT, Janesick AS, Blumberg B. Triflumizole is an obesogen in mice that acts through peroxisome proliferator activated receptor gamma (PPARγ). Environ Health Perspect. 2012; 120:1720–1726.
49. Ravussin E, Smith SR. Increased fat intake, impaired fat oxidation, and failure of fat cell proliferation result in ectopic fat storage, insulin resistance, and type 2 diabetes mellitus. Ann N Y Acad Sci. 2002; 967:363–378.
50. Moreno-Aliaga MJ, Matsumura F. Endrin inhibits adipocyte differentiation by selectively altering expression pattern of CCAAT/enhancer binding protein-alpha in 3T3-L1 cells. Mol Pharmacol. 1999; 56:91–101.
51. Hsu HF, Tsou TC, Chao HR, Kuo YT, Tsai FY, Yeh SC. Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on adipogenic differentiation and insulin-induced glucose uptake in 3T3-L1 cells. J Hazard Mater. 2010; 182:649–655.
52. Shimba S, Wada T, Tezuka M. Arylhydrocarbon receptor (AhR) is involved in negative regulation of adipose differentiation in 3T3-L1 cells: AhR inhibits adipose differentiation independently of dioxin. J Cell Sci. 2001; 114(Pt 15):2809–2817.
53. Wauson EM, Langan AS, Vorce RL. Sodium arsenite inhibits and reverses expression of adipogenic and fat cell-specific genes during in vitro adipogenesis. Toxicol Sci. 2002; 65:211–219.
54. Steffens AA, Hong GM, Bain LJ. Sodium arsenite delays the differentiation of C2C12 mouse myoblast cells and alters methylation patterns on the transcription factor myogenin. Toxicol Appl Pharmacol. 2011; 250:154–161.
55. Yen YP, Tsai KS, Chen YW, Huang CF, Yang RS, Liu SH. Arsenic inhibits myogenic differentiation and muscle regeneration. Environ Health Perspect. 2010; 118:949–956.
56. Turer AT, Scherer PE. Adiponectin: mechanistic insights and clinical implications. Diabetologia. 2012; 55:2319–2326.
57. Greenberg AS, Obin MS. Obesity and the role of adipose tissue in inflammation and metabolism. Am J Clin Nutr. 2006; 83:461S–465S.
58. Kawakami T, Sugimoto H, Furuichi R, Kadota Y, Inoue M, Setsu K, Suzuki S, Sato M. Cadmium reduces adipocyte size and expression levels of adiponectin and Peg1/Mest in adipose tissue. Toxicology. 2010; 267:20–26.
59. Zuo Z, Chen S, Wu T, Zhang J, Su Y, Chen Y, Wang C. Tributyltin causes obesity and hepatic steatosis in male mice. Environ Toxicol. 2011; 26:79–85.
60. Kidani T, Kamei S, Miyawaki J, Aizawa J, Sakayama K, Masuno H. Bisphenol A downregulates Akt signaling and inhibits adiponectin production and secretion in 3T3-L1 adipocytes. J Atheroscler Thromb. 2010; 17:834–843.
61. Hugo ER, Brandebourg TD, Woo JG, Loftus J, Alexander JW, Ben-Jonathan N. Bisphenol A at environmentally relevant doses inhibits adiponectin release from human adipose tissue explants and adipocytes. Environ Health Perspect. 2008; 116:1642–1647.
62. Xu X, Liu C, Xu Z, Tzan K, Zhong M, Wang A, Lippmann M, Chen LC, Rajagopalan S, Sun Q. Long-term exposure to ambient fine particulate pollution induces insulin resistance and mitochondrial alteration in adipose tissue. Toxicol Sci. 2011; 124:88–98.
63. Hong EG, Ko HJ, Cho YR, Kim HJ, Ma Z, Yu TY, Friedline RH, Kurt-Jones E, Finberg R, Fischer MA, Granger EL, Norbury CC, Hauschka SD, Philbrick WM, Lee CG, Elias JA, Kim JK. Interleukin-10 prevents diet-induced insulin resistance by attenuating macrophage and cytokine response in skeletal muscle. Diabetes. 2009; 58:2525–2535.
64. Xiao Z, Mohamood AS, Uddin S, Gutfreund R, Nakata C, Marshall A, Kimura H, Caturegli P, Womer KL, Huang Y, Jie C, Chakravarti S, Schneck JP, Yagita H, Hamad AR. Inhibition of Fas ligand in NOD mice unmasks a protective role for IL-10 against insulitis development. Am J Pathol. 2011; 179:725–732.
65. Sun Q, Yue P, Deiuliis JA, Lumeng CN, Kampfrath T, Mikolaj MB, Cai Y, Ostrowski MC, Lu B, Parthasarathy S, Brook RD, Moffatt-Bruce SD, Chen LC, Rajagopalan S. Ambient air pollution exaggerates adipose inflammation and insulin resistance in a mouse model of diet-induced obesity. Circulation. 2009; 119:538–546.
66. Kern PA, Dicker-Brown A, Said ST, Kennedy R, Fonseca VA. The stimulation of tumor necrosis factor and inhibition of glucose transport and lipoprotein lipase in adipose cells by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Metabolism. 2002; 51:65–68.
67. Nishiumi S, Yoshida M, Azuma T, Yoshida K, Ashida H. 2,3,7,8-tetrachlorodibenzo-p-dioxin impairs an insulin signaling pathway through the induction of tumor necrosis factor-alpha in adipocytes. Toxicol Sci. 2010; 115:482–491.
68. Sonne C, Fonfara S, Dietz R, Kirkegaard M, Letcher RJ, Shahmiri S, Andersen S, Moller P. Multiple cytokine and acute-phase protein gene transcription in West Greenland sledge dogs (Canis familiaris) dietary exposed to organic environmental pollutants. Arch Environ Contam Toxicol. 2007; 53:110–118.
69. Wang J, Lv X, Du Y. Inflammatory response and insulin signaling alteration induced by PCB77. J Environ Sci (China). 2010; 22:1086–1090.
70. Howell G 3rd, Mangum L. Exposure to bioaccumulative organochlorine compounds alters adipogenesis, fatty acid uptake, and adipokine production in NIH3T3-L1 cells. Toxicol In Vitro. 2011; 25:394–402.
71. Ben-Jonathan N, Hugo ER, Brandebourg TD. Effects of bisphenol A on adipokine release from human adipose tissue: implications for the metabolic syndrome. Mol Cell Endocrinol. 2009; 304:49–54.
72. Sargis RM, Neel BA, Brock CO, Lin Y, Hickey AT, Carlton DA, Brady MJ. The novel endocrine disruptor tolylfluanid impairs insulin signaling in primary rodent and human adipocytes through a reduction in insulin receptor substrate-1 levels. Biochim Biophys Acta. 2012; 1822:952–960.
73. Sun XJ, Liu F. Phosphorylation of IRS proteins Yin-Yang regulation of insulin signaling. Vitam Horm. 2009; 80:351–387.
74. Zheng Z, Xu X, Zhang X, Wang A, Zhang C, Huttemann M, Grossman LI, Chen LC, Rajagopalan S, Sun Q, Zhang K. Exposure to ambient particulate matter induces a NASH-like phenotype and impairs hepatic glucose metabolism in an animal model. J Hepatol. 2013; 58:148–154.
75. Rajesh P, Sathish S, Srinivasan C, Selvaraj J, Balasubramanian K. Phthalate is associated with insulin resistance in adipose tissue of male rat: role of antioxidant vitamins. J Cell Biochem. 2013; 114:558–569.
76. Xue P, Hou Y, Zhang Q, Woods CG, Yarborough K, Liu H, Sun G, Andersen ME, Pi J. Prolonged inorganic arsenite exposure suppresses insulin-stimulated AKT S473 phosphorylation and glucose uptake in 3T3-L1 adipocytes: involvement of the adaptive antioxidant response. Biochem Biophys Res Commun. 2011; 407:360–365.
77. Paul DS, Harmon AW, Devesa V, Thomas DJ, Styblo M. Molecular mechanisms of the diabetogenic effects of arsenic: inhibition of insulin signaling by arsenite and methylarsonous acid. Environ Health Perspect. 2007; 115:734–742.
78. Indumathi D, Jayashree S, Selvaraj J, Sathish S, Mayilvanan C, Akilavalli N, Balasubramanian K. Effect of bisphenol-A on insulin signal transduction and glucose oxidation in skeletal muscle of adult male albino rat. Hum Exp Toxicol. 2013; 32:960–971.
79. Jayashree S, Indumathi D, Akilavalli N, Sathish S, Selvaraj J, Balasubramanian K. Effect of Bisphenol-A on insulin signal transduction and glucose oxidation in liver of adult male albino rat. Environ Toxicol Pharmacol. 2013; 35:300–310.
80. Rengarajan S, Parthasarathy C, Anitha M, Balasubramanian K. Diethylhexyl phthalate impairs insulin binding and glucose oxidation in Chang liver cells. Toxicol In Vitro. 2007; 21:99–102.
81. Han JC, Park SY, Hah BG, Choi GH, Kim YK, Kwon TH, Kim EK, Lachaal M, Jung CY, Lee W. Cadmium induces impaired glucose tolerance in rat by down-regulating GLUT4 expression in adipocytes. Arch Biochem Biophys. 2003; 413:213–220.
82. Sakurai K, Kawazuma M, Adachi T, Harigaya T, Saito Y, Hashimoto N, Mori C. Bisphenol A affects glucose transport in mouse 3T3-F442A adipocytes. Br J Pharmacol. 2004; 141:209–214.
83. Lind L, Lind PM. Can persistent organic pollutants and plastic-associated chemicals cause cardiovascular disease? J Intern Med. 2012; 271:537–553.
84. Viluksela M, Unkila M, Pohjanvirta R, Tuomisto JT, Stahl BU, Rozman KK, Tuomisto J. Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on liver phosphoenolpyruvate carboxykinase (PEPCK) activity, glucose homeostasis and plasma amino acid concentrations in the most TCDD-susceptible and the most TCDD-resistant rat strains. Arch Toxicol. 1999; 73:323–336.
85. Zhang W, Sargis RM, Volden PA, Carmean CM, Sun XJ, Brady MJ. PCB 126 and other dioxin-like PCBs specifically suppress hepatic PEPCK expression via the aryl hydrocarbon receptor. PLoS One. 2012; 7:e37103.
86. Neel BA, Brady MJ, Sargis RM. The endocrine disrupting chemical tolylfluanid alters adipocyte metabolism via glucocorticoid receptor activation. Mol Endocrinol. 2013; 27:394–406.
87. Garbrecht MR, Krozowski ZS, Snyder JM, Schmidt TJ. Reduction of glucocorticoid receptor ligand binding by the 11-beta hydroxysteroid dehydrogenase type 2 inhibitor, Thiram. Steroids. 2006; 71:895–901.
88. Johansson M, Johansson N, Lund BO. Xenobiotics and the glucocorticoid receptor: additive antagonistic effects on tyrosine aminotransferase activity in rat hepatoma cells. Basic Clin Pharmacol Toxicol. 2005; 96:309–315.
89. Johansson M, Nilsson S, Lund BO. Interactions between methylsulfonyl PCBs and the glucocorticoid receptor. Environ Health Perspect. 1998; 106:769–772.
90. Prasanth GK, Divya LM, Sadasivan C. Bisphenol-A can bind to human glucocorticoid receptor as an agonist: an in silico study. J Appl Toxicol. 2010; 30:769–774.
91. Atanasov AG, Tam S, Rocken JM, Baker ME, Odermatt A. Inhibition of 11 beta-hydroxysteroid dehydrogenase type 2 by dithiocarbamates. Biochem Biophys Res Commun. 2003; 308:257–262.
92. Meyer A, Strajhar P, Murer C, Da Cunha T, Odermatt A. Species-specific differences in the inhibition of human and zebrafish 11β-hydroxysteroid dehydrogenase 2 by thiram and organotins. Toxicology. 2012; 301:72–78.
93. Wang J, Sun B, Hou M, Pan X, Li X. The environmental obesogen bisphenol A promotes adipogenesis by increasing the amount of 11β-hydroxysteroid dehydrogenase type 1 in the adipose tissue of children. Int J Obes (Lond). 2013; 37:999–1005.
94. Bodwell JE, Kingsley LA, Hamilton JW. Arsenic at very low concentrations alters glucocorticoid receptor (GR)-mediated gene activation but not GR-mediated gene repression: complex dose-response effects are closely correlated with levels of activated GR and require a functional GR DNA binding domain. Chem Res Toxicol. 2004; 17:1064–1076.
95. Gosse JA, Taylor VF, Jackson BP, Hamilton JW, Bodwell JE. Monomethylated trivalent arsenic species disrupt steroid receptor interactions with their DNA response elements at non-cytotoxic cellular concentrations. J Appl Toxicol. Epub 2013 Jun 14. http://dx.doi.org/10.1002/jat.2898.
96. Oliveira E, Pinheiro CR, Santos-Silva AP, Trevenzoli IH, Abreu-Villaca Y, Nogueira Neto JF, Reis AM, Passos MC, Moura EG, Lisboa PC. Nicotine exposure affects mother's and pup's nutritional, biochemical, and hormonal profiles during lactation in rats. J Endocrinol. 2010; 205:159–170.
97. Chang LL, Wun WS, Wang PS. In utero and neonate exposure to nonylphenol develops hyperadrenalism and metabolic syndrome later in life. I. First generation rats (F(1)). Toxicology. 2012; 301:40–49.
98. Beausoleil C, Ormsby JN, Gies A, Hass U, Heindel JJ, Holmer ML, Nielsen PJ, Munn S, Schoenfelder G. Low dose effects and non-monotonic dose responses for endocrine active chemicals: science to practice workshop: workshop summary. Chemosphere. 2013; 93:847–856.
99. Angelin B, Larsson TE, Rudling M. Circulating fibroblast growth factors as metabolic regulators: a critical appraisal. Cell Metab. 2012; 16:693–705.
100. Cardon CM, Rutter J. PAS kinase: integrating nutrient sensing with nutrient partitioning. Semin Cell Dev Biol. 2012; 23:626–630.
101. Fajans SS, Bell GI. MODY: history, genetics, pathophysiology, and clinical decision making. Diabetes Care. 2011; 34:1878–1884.
Full Text Links
  • DMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr