Hostname: page-component-848d4c4894-p2v8j Total loading time: 0 Render date: 2024-05-06T12:54:27.970Z Has data issue: false hasContentIssue false

Early onset and progression of non-alcoholic fatty liver disease in young monosodium l-glutamate-induced obese mice

Published online by Cambridge University Press:  01 June 2018

C. F. F. Coelho
Affiliation:
Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, MA, Brazil
L. M. França
Affiliation:
Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, MA, Brazil Health Sciences Graduate Program, Federal University of Maranhão, São Luís, MA, Brazil
J. R. Nascimento
Affiliation:
Laboratory of Immunophysiology, Department of Pathology, Federal University of Maranhão, São Luís, MA, Brazil Health Sciences Graduate Program, Federal University of Maranhão, São Luís, MA, Brazil
A. M. dos Santos
Affiliation:
Department of Public Health, Federal University of Maranhão, São Luís, MA, Brazil Health Sciences Graduate Program, Federal University of Maranhão, São Luís, MA, Brazil
A. P. S. Azevedo-Santos
Affiliation:
Laboratory of Immunophysiology, Department of Pathology, Federal University of Maranhão, São Luís, MA, Brazil Health Sciences Graduate Program, Federal University of Maranhão, São Luís, MA, Brazil
F. R. F. Nascimento
Affiliation:
Laboratory of Immunophysiology, Department of Pathology, Federal University of Maranhão, São Luís, MA, Brazil Health Sciences Graduate Program, Federal University of Maranhão, São Luís, MA, Brazil
A. M. A. Paes*
Affiliation:
Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, MA, Brazil Health Sciences Graduate Program, Federal University of Maranhão, São Luís, MA, Brazil
*
Address for correspondence: A. M. de Andrade Paes, Avenida dos Portugueses, 1966 – Cidade Universitária Dom Delgado, 65080-850 São Luís, MA, Brazil. E-mail: marcuspaes@ufma.br

Abstract

Monosodium l-glutamate (MSG)-induced obesity is a useful model for non-alcoholic fatty liver disease (NAFLD) studies. However, there is limited data on its initiation and progression. Thus, this study aimed to characterize the onset of metabolic and histopathological features of NAFLD and its progression to non-alcoholic steatohepatitis (NASH) in this model. To perform this study, Swiss mice pups were neonatally injected with MSG (4 g/kg/day, s.c.) or equiosmolar saline and followed up to 60, 120 or 180 days old. At each age, blood, liver, as well as periepididymal and retroperitoneal fat pads were collected for morphometric, biochemical and histological analyses, the later according to NAFLD activity score. MSG mice presented hypertriglyceridemia and central obesity at all ages, but peripheral insulin-resistance was verified only in 120- and 180-day-old mice. Hepatic total fat and triglycerides content were higher in MSG mice at all ages. Accordingly, histopathological analysis showed that 60-day-old MSG mice had microvesicular steatosis with occasional ballooning, which evolved into NASH from 120 days old. Retroperitoneal fat accumulation was the only variable to independently correlate with NAFLD activity total score upon multivariate analysis (R2=71.45%). There were no differences in IL-6 and TNF-α serum levels among groups. Overall, this study shows that NAFLD is a precocious outcome in MSG-obese mice, whereas the period comprised between 60 and 120 days old seems to be a crucial metabolic window for comprehending pathophysiological events involved in NAFLD-to-NASH progression in this model.

Type
Original Article
Copyright
© Cambridge University Press and the International Society for Developmental Origins of Health and Disease 2018. 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

1. Chalasani, N, Younossi, Z, Lavine, JE, et al. The diagnosis and management of non-alcoholic fatty liver disease: practice Guideline by the American Association for the Study of Liver Diseases, American College of Gastroenterology, and the American Gastroenterological Association. Hepatology (Baltimore, Md). 2012; 55, 20052023.Google Scholar
2. Younossi, ZM, Koenig, AB, Abdelatif, D, et al. Global epidemiology of nonalcoholic fatty liver disease-meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology (Baltimore, Md). 2016; 64, 7384.Google Scholar
3. Day, CP, James, OF. Steatohepatitis: a tale of two “hits”? Gastroenterology. 1998; 114, 842845.Google Scholar
4. Tiniakos, DG, Vos, MB, Brunt, EM. Nonalcoholic fatty liver disease: pathology and pathogenesis. Annl Rev Pathol. 2010; 5, 145171.Google Scholar
5. Larter, CZ. Not all models of fatty liver are created equal: understanding mechanisms of steatosis development is important. J Gastroenterol Hepatol. 2007; 22, 13531354.Google Scholar
6. Kanuri, G, Bergheim, I. In vitro and in vivo models of non-alcoholic fatty liver disease (NAFLD). Int J Mol Sci. 2013; 14, 1196311980.Google Scholar
7. Tsuneyama, K, Nishitsuji, K, Matsumoto, M, et al. Animal models for analyzing metabolic syndrome-associated liver diseases. Pathol Int. 2017; 67, 539546.Google Scholar
8. Olney, JW. Glutamate-induced neuronal necrosis in the infant mouse hypothalamus. An electron microscopic study. J Neuropathol Exp Neurol. 1971; 30, 7590.Google Scholar
9. Tanaka, K, Shimada, M, Nakao, K, Kusunoki, T. Hypothalamic lesion induced by injection of monosodium glutamate in suckling period and subsequent development of obesity. Exp Neurol. 1978; 62, 191199.Google Scholar
10. Holzwarth-McBride, MA, Sladek, JR Jr., Knigge, KM. Monosodium glutamate induced lesions of the arcurate nucleus. II. Fluorescence histochemistry of catecholamines. Anatomical Record. 1976; 186, 197205.Google Scholar
11. Nemeroff, CB, Lipton, MA, Kizer, JS. Models of neuroendocrine regulation: use of monosodium glutamate as an investigational tool. Dev Neurosci. 1978; 1, 102109.Google Scholar
12. Sasaki, F, Kawai, T, Ohta, M. Immunohistochemical evidence of neurons with GHRH or LHRH in the arcuate nucleus of male mice and their possible role in the postnatal development of adenohypophysial cells. Anatomical Record. 1994; 240, 255260.Google Scholar
13. Remke, H, Wilsdorf, A, Müller, F. Development of hypothalamic obesity in growing rats. Exp Pathol. 1988; 33, 223232.Google Scholar
14. Cameron, DP, Cutbush, L, Opat, F. Effects of monosodium glutamate-induced obesity in mice on carbohydrate metabolism in insulin secretion. Clin Exp Pharmacol Physiol. 1978; 5, 4151.Google Scholar
15. Yamamoto, T, Matsuo, S, Ueshima, Y, et al. Plasma levels of insulin-like growth factor-I are reduced at one week of age in monosodium L-glutamate-treated mice. Endocrine J. 1993; 40, 461465.Google Scholar
16. Hernandez-Bautista, RJ, Alarcon-Aguilar, FJ, Del, CE-VM, et al. Biochemical alterations during the obese-aging process in female and male monosodium glutamate (MSG)-treated mice. Int J Mol Sci. 2014; 15, 1147311494.Google Scholar
17. Tsuneyama, K, Nishida, T, Baba, H, et al. Neonatal monosodium glutamate treatment causes obesity, diabetes, and macrovesicular steatohepatitis with liver nodules in DIAR mice. J Gastroenterol Hepatol. 2014; 29, 17361743.Google Scholar
18. Nagata, M, Suzuki, W, Iizuka, S, et al. Type 2 diabetes mellitus in obese mouse model induced by monosodium glutamate. Exp Anim. 2006; 55, 109115.Google Scholar
19. Dawson, R, Pelleymounter, MA, Millard, WJ, Liu, S, Eppler, B. Attenuation of leptin-mediated effects by monosodium glutamate-induced arcuate nucleus damage. Am J Physiol. 1997; 273(Pt 1), E202E206.Google Scholar
20. Reynolds, CM, Perry, JK, Vickers, MH. Manipulation of the growth hormone-insulin-like growth factor (GH-IGF) axis: a treatment strategy to reverse the effects of early life developmental programming. Int J Mol Sci. 2017; 18, 1729.Google Scholar
21. Franca, LM, Freitas, LN, Chagas, VT, et al. Mechanisms underlying hypertriglyceridemia in rats with monosodium L-glutamate-induced obesity: evidence of XBP-1/PDI/MTP axis activation. Biochem Biophys Res Commun. 2014; 443, 725730.Google Scholar
22. Nakanishi, Y, Tsuneyama, K, Fujimoto, M, et al. Monosodium glutamate (MSG): a villain and promoter of liver inflammation and dysplasia. J Autoimmun. 2008; 30, 4250.Google Scholar
23. Sasaki, Y, Suzuki, W, Shimada, T, et al. Dose dependent development of diabetes mellitus and non-alcoholic steatohepatitis in monosodium glutamate-induced obese mice. Life Sci. 2009; 85, 490498.Google Scholar
24. Bernardis, LL, Patterson, BD. Correlation between ‘Lee index’ and carcass fat content in weanling and adult female rats with hypothalamic lesions. J Endocrinol. 1968; 40, 527528.Google Scholar
25. Guerrero-Romero, F, Simental-Mendia, LE, Gonzalez-Ortiz, M, et al. The product of triglycerides and glucose, a simple measure of insulin sensitivity. Comparison with the euglycemic-hyperinsulinemic clamp. J Clin Endocrinol Metab. 2010; 95, 33473351.Google Scholar
26. Kleiner, DE, Brunt, EM, Van Natta, M, et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology (Baltimore, Md). 2005; 41, 13131321.Google Scholar
27. Hidalgo, B, Goodman, M. Multivariate or multivariable regression? Am J Public Health. 2013; 103, 3940.Google Scholar
28. Imajo, K, Yoneda, M, Kessoku, T, et al. Rodent models of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Int J Mol Sci. 2013; 14, 2183321857.Google Scholar
29. Lubaczeuski, C, Balbo, SL, Ribeiro, RA, et al. Vagotomy ameliorates islet morphofunction and body metabolic homeostasis in MSG-obese rats. Braz J Med Biol Res. 2015; 48, 447457.Google Scholar
30. Dolnikoff, M, Martin-Hidalgo, A, Machado, UF, Lima, FB, Herrera, E. Decreased lipolysis and enhanced glycerol and glucose utilization by adipose tissue prior to development of obesity in monosodium glutamate (MSG) treated-rats. Int J Obes Relat Metab Disord. 2001; 25, 426433.Google Scholar
31. Marçal, AC, Grassiolli, S, da Rocha, DN, et al. The dual effect of isoproterenol on insulin release is suppressed in pancreatic islets from hypothalamic obese rats. Endocrine. 2006; 29, 445449.Google Scholar
32. Bartels, ED, Lauritsen, M, Nielsen, LB. Hepatic expression of microsomal triglyceride transfer protein and in vivo secretion of triglyceride-rich lipoproteins are increased in obese diabetic mice. Diabetes. 2002; 51, 12331239.Google Scholar
33. Baiceanu, A, Mesdom, P, Lagouge, M, Foufelle, F. Endoplasmic reticulum proteostasis in hepatic steatosis. Nat Rev Endocrinol. 2016; 12, 710722.Google Scholar
34. Hazlehurst, JM, Woods, C, Marjot, T, Cobbold, JF, Tomlinson, JW. Non-alcoholic fatty liver disease and diabetes. Metabolism. 2016; 65, 10961108.Google Scholar
35. Donnelly, KL, Smith, CI, Schwarzenberg, SJ, et al. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J Clin Invest. 2005; 115, 13431351.Google Scholar
36. Rhee, EJ, Lee, WY, Cho, YK, Kim, BI, Sung, KC. Hyperinsulinemia and the development of nonalcoholic fatty liver disease in nondiabetic adults. Am J Med. 2011; 124, 6976.Google Scholar
37. Vatner, DF, Majumdar, SK, Kumashiro, N, et al. Insulin-independent regulation of hepatic triglyceride synthesis by fatty acids. Proc Natl Acad Sci USA. 2015; 112, 11431148.Google Scholar
38. Mazidi, M, Kengne, A-P, Katsiki, N, Mikhailidis, DP, Banach, M. Lipid accumulation product and triglycerides/glucose index are useful predictors of insulin resistance. J Diabetes Complications. 2018; 32, 266270.Google Scholar
39. Vasques, AC, Novaes, FS, de Oliveira Mda, S, et al. TyG index performs better than HOMA in a Brazilian population: a hyperglycemic clamp validated study. Diabetes Res Clin Pract. 2011; 93, e98e100.Google Scholar
40. Irace, C, Carallo, C, Scavelli, FB, et al. Markers of insulin resistance and carotid atherosclerosis. A comparison of the homeostasis model assessment and triglyceride glucose index. Int J Clin Pract. 2013; 67, 665672.Google Scholar
41. Zhang, S, Du, T, Li, M, et al. Triglyceride glucose-body mass index is effective in identifying nonalcoholic fatty liver disease in nonobese subjects. Medicine (Baltimore). 2017; 96, e7041.Google Scholar
42. Simental-Mendia, LE, Simental-Mendia, E, Rodriguez-Hernandez, H, Rodriguez-Moran, M, Guerrero-Romero, F. The product of triglycerides and glucose as biomarker for screening simple steatosis and NASH in asymptomatic women. Ann Hepatol. 2016; 15, 715720.Google Scholar
43. Gamboa-Gómez, CI, Simental-Mendía, LE, González-Laredo, RF, et al. In vitro and in vivo assessment of anti-hyperglycemic and antioxidant effects of Oak leaves (Quercus convallata and Quercus arizonica) infusions and fermented beverages. Food Res Int. 2017; 102, 690699.Google Scholar
44. Nunes-Souza, V, Cesar-Gomes, CJ, Da Fonseca, LJ, et al. Aging increases susceptibility to high fat diet-induced metabolic syndrome in C57BL/6 mice: improvement in glycemic and lipid profile after antioxidant therapy. Oxid Med Cell Longev. 2016; 2016, 1987960.Google Scholar
45. Shree, N, Bhonde, RR. Metformin preconditioned adipose derived mesenchymal stem cells is a better option for the reversal of diabetes upon transplantation. Biomed Pharmacother. 2016; 84, 16621667.Google Scholar
46. Sunil, V, Shree, N, Venkataranganna, MV, Bhonde, RR, Majumdar, M. The anti diabetic and anti obesity effect of Memecylon umbellatum extract in high fat diet induced obese mice. Biomed Pharmacother. 2017; 89, 880886.Google Scholar
47. Machado, UF, Shimizu, I, Saito, M. Reduced content and preserved translocation of glucose transporter (GLUT 4) in white adipose tissue of obese mice. Physiol Behav. 1994; 55, 621625.Google Scholar
48. Sasaki, Y, Shimada, T, Iizuka, S, et al. Effects of bezafibrate in nonalcoholic steatohepatitis model mice with monosodium glutamate-induced metabolic syndrome. Eur J Pharmacol. 2011; 662, 18.Google Scholar
49. Takai, A, Kikuchi, K, Kajiyama, Y, et al. Serological and histological examination of a nonalcoholic steatohepatitis mouse model created via the administration of monosodium glutamate. Int Scholarly Res Notices. 2014; 2014, 725351.Google Scholar
50. Yeh, MM, Brunt, EM. Pathological features of fatty liver disease. Gastroenterology. 2014; 147, 754764.Google Scholar
51. Argo, CK, Northup, PG, Al-Osaimi, AM, Caldwell, SH. Systematic review of risk factors for fibrosis progression in non-alcoholic steatohepatitis. J Hepatol. 2009; 51, 371379.Google Scholar
52. Chusyd, DE, Wang, D, Huffman, DM, Nagy, TR. Relationships between rodent white adipose fat pads and human white adipose fat depots. Front Nutri. 2016; 3, 10.Google Scholar
53. Chung, GE, Kim, D, Kwark, MS, et al. Visceral adipose tissue area as an independent risk factor for elevated liver enzyme in nonalcoholic fatty liver disease. Medicine. 2015; 94, e573.Google Scholar
54. Bastard, J-P, Maachi, M, Lagathu, C, et al. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw. 2006; 17, 412.Google Scholar
55. Shi, H, Strader, AD, Woods, SC, Seeley, RJ. The effect of fat removal on glucose tolerance is depot specific in male and female mice. Am J Physiol Endocrinol Metab. 2007; 293, E1012E1020.Google Scholar
56. Thörne, A, Löfgren, P, Hoffstedt, J. Increased visceral adipocyte lipolysis – a pathogenic role in nonalcoholic fatty liver disease? J Clin Endocrinol Metab. 2010; 95, E209E213.Google Scholar
57. Tilg, H, Moschen, AR. Evolution of inflammation in nonalcoholic fatty liver disease: the multiple parallel hits hypothesis. Hepatology (Baltimore, Md). 2010; 52, 18361846.Google Scholar
58. Zhang, N, Huan, Y, Huang, H, et al. Atorvastatin improves insulin sensitivity in mice with obesity induced by monosodium glutamate. Acta Pharmacologica Sinica. 2010; 31, 3542.Google Scholar
59. Tokuyama, K, Himms-Hagen, J. Adrenalectomy prevents obesity in glutamate-treated mice. Am J Physiol Endocrinol Metab. 1989; 257, E139E144.Google Scholar
60. Brattsand, R, Linden, M. Cytokine modulation by glucocorticoids: mechanisms and actions in cellular studies. Aliment Pharmacol Ther. 1996; 10(Suppl. 2), 8190; discussion 82–91.Google Scholar
61. Moreno, G, Perelló, M, Camihort, G, et al. Impact of transient correction of increased adrenocortical activity in hypothalamo-damaged, hyperadipose female rats. Int J Obesity. 2006; 30, 7382.Google Scholar
62. Kratschmar, DV, Calabrese, D, Walsh, J, et al. Suppression of the Nrf2-dependent antioxidant response by glucocorticoids and 11beta-HSD1-mediated glucocorticoid activation in hepatic cells. PLoS One. 2012; 7, e36774.Google Scholar
63. Mueller, KM, Kornfeld, JW, Friedbichler, K, et al. Impairment of hepatic growth hormone and glucocorticoid receptor signaling causes steatosis and hepatocellular carcinoma in mice. Hepatology (Baltimore, Md). 2011; 54, 13981409.Google Scholar
64. Yamazaki, Y, Usui, I, Kanatani, Y, et al. Treatment with SRT1720, a SIRT1 activator, ameliorates fatty liver with reduced expression of lipogenic enzymes in MSG mice. Am J Physiol Endocrinol Metab. 2009; 297, E1179E1186.Google Scholar