Features of the Course of Non-alcoholic Fatty Liver Disease in Experimental Animals at High Altitudes

Authors

  • Nurgul Toktogulova Department of Therapy, Pediatrics and Dentistry Specialization, Kyrgyz-Russian Slavic University Named after B.N. Yeltsin (KRSU), Bishkek, Kyrgyzstan
  • Rustam Tuhvatshin Department of Pathological Physiology, Kyrgyz State Medical Academy Named after I.K. Akhunbaev (KSMA), Bishkek, Kyrgyzstan

DOI:

https://doi.org/10.3889/oamjms.2021.7506

Keywords:

Non-alcoholic fatty liver disease, Modeling, Hypobaric hypoxia

Abstract

Background: Nearly 25% of adults worldwide are affected by non-alcoholic fatty liver disease (NAFLD). taged changes in the liver from steatosis progress to non-alcoholic steatohepatitis (NASH) and its complicated forms such as fibrosis, cirrhosis, and hepatocellular carcinoma. There are very few data in the literature on the development of NAFLD in conditions of high altitude. There are no data on the state of pro- and anti-inflammatory cytokines in NAFLD in high altitude conditions. Thus, simulating NAFLD on animals in artificial highlands will help find answers to these questions.

Aim: to study the features of the course of non-alcoholic fatty liver disease (NAFLD) in experimental animals in artificial high-mountain conditions.

Material and methods: The study was carried out on 180 male Wistar rats. 7 groups of experimental animals were formed, which were divided into control and experimental groups. The rats of the control group were on a standard diet. Non-alcoholic fatty liver disease was modeled by keeping animals on a diet (Ackermann et al., 2005) rich in fructose and fat in low and high mountain conditions (in a pressure chamber 6000 m above sea level) for 35 and 70 days. In all groups of animals, the following was determined: the concentration of total bilirubin (TB), the activity of the enzymes aspartate aminotransferase (AST), alanine aminotransferase (ALT), the level of total cholesterol (TC), low-density lipoprotein cholesterol (LDL), the total protein content in plasma (TP), pro- and anti-inflammatory cytokines.

Results: In animals on a diet enriched with fructose and fat, it equally led to the inhibition of the synthetic function of the liver, both in high altitude and in low altitudes. Liver enzyme levels were uncertain. AST levels were high in all major groups, with a similar upward trend at 5 and 10 weeks on the fructose-fortified diet. The greatest shift was observed on the part of ALT in animals under conditions of hypobaric hypoxia, the growth of which was statistically significantly lower than in the low- altitude groups. The opposite picture was observed in pigment metabolism. Indicators of total cholesterol and LDL increased almost twofold in the main high- altitude groups, and were significantly higher than the indicators of low- altitude animals with p <0.001. The activity of pro- and anti-inflammatory cytokines in the main group, when the animals were raised in the pressure chamber, statistically significantly increased after 5 weeks compared to the low-altitude group by more than 2 times and statistically significantly correlated with cytolysis syndromes, hypercholesterolemia and impaired synthetic function against the background of liver hypoergosis.

Conclusion: Non-alcoholic fatty liver disease in animals on a special diet enriched with fructose under conditions of hypobaric hypoxia is characterized by deeper violations of pigment metabolism, pro- and anti-inflammatory cytokines and lipid spectrum with simultaneously statistically significant low alanine aminotransferase indices compared to low-altitude groups on an identical diet.

Downloads

Download data is not yet available.

Metrics

Metrics Loading ...

Plum Analytics Artifact Widget Block

References

Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism. 2016;65:1038-48. https://doi.org/10.1016/j.metabol.2015.12.012 PMid:26823198 DOI: https://doi.org/10.1016/j.metabol.2015.12.012

Foroughi M, Maghsoudi Z, Khayyatzadeh S, Ghiasvand R, Askari G, Iraj B. Relationship between non-alcoholic fatty liver disease and inflammation in patients with non-alcoholic fatty liver. Adv Biomed Res. 2016;5:28. https://doi.org/10.4103/2277-9175.176368 PMid:27014655 DOI: https://doi.org/10.4103/2277-9175.176368

Basaranoglu M, Basaranoglu G, Bugianesi E. Carbohydrate intake and nonalcoholic fatty liver disease: Fructose as a weapon of mass destruction. Hepatobiliary Surg Nutr. 2015;4(2):109-16. https://doi.org/10.3978/j.issn.2304-3881.2014.11.05 PMid:26005677

Chung M, Ma J, Patel K, Berger S, Lau J, Lichtenstein AH. Fructose, high-fructose corn syrup, sucrose, and nonalcoholic fatty liver disease or indexes of liver health: A systematic review and meta-analysis. Am J Clin Nutr. 2014;100(3):833-49. https://doi.org/10.3945/ajcn.114.086314 PMid:25099546 DOI: https://doi.org/10.3945/ajcn.114.086314

Schwarz JM, Noworolski SM, Wen MJ, Dyachenko A, Prior JL, Weinberg ME, et al. Effect of a high-fructose weight-maintaining diet on lipogenesis and liver fat. J Clin Endocrinol Metab. 2015;100(6):2434-42. https://doi.org/10.1210/jc.2014-3678 PMid:25825943 DOI: https://doi.org/10.1210/jc.2014-3678

Sanyal D, Mukherjee P, Raychaudhuri M, Ghosh S, Mukherjee S, Chowdhury S. Profile of liver enzymes in non-alcoholic fatty liver disease in patients with impaired glucose tolerance and newly detected untreated type 2 diabetes. Indian J Endocrinol Metab. 2015;19(5):597-601. https://doi.org/10.4103/2230-8210.163172 PMid:26425466 DOI: https://doi.org/10.4103/2230-8210.163172

Arvind A, Osganian SA, Cohen DE, Corey KE. Lipid and lipoprotein metabolism in liver disease. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman J, et al, editors. Endotext. South Dartmouth, MA: MDText, Inc.; 2019.

Deprince A, Haas JT, Staels B. Dysregulated lipid metabolism links NAFLD to cardiovascular disease. Mol Metab. 2020;42:101092. https://doi.org/10.1016/j.molmet.2020.101092 PMid:33010471 DOI: https://doi.org/10.1016/j.molmet.2020.101092

Song K, Zhang Y, Ga Q, Bai Z, Ge RL. High-altitude chronic hypoxia ameliorates obesity-induced non-alcoholic fatty liver disease in mice by regulating mitochondrial and AMPK signaling. Life Sci. 2020;252:117633. https://doi.org/10.1016/j.lfs.2020.117633 PMid:32289432 DOI: https://doi.org/10.1016/j.lfs.2020.117633

Ackerman Z, Oron-Herman M, Grozovski M, Rosenthal T, Pappo O, Link G, et al. Fructose-induced fatty liver disease: Hepatic effects of blood pressure and plasma triglyceride reduction. Hypertension. 2005;45(5):1012-8. https://doi.org/10.1161/01.HYP.0000164570.20420.67 PMid:15824194 DOI: https://doi.org/10.1161/01.HYP.0000164570.20420.67

Meerson FZ. Adaptation to high-altitude hypoxia. In: Physiology of Adaptation Processes. Moscow: Nauka; 1986. p. 635.

Dekker MJ, Su Q, Baker C, Rutledge AC, Adeli K. Fructose: A highly lipogenic nutrient implicated in insulin resistance, hepatic steatosis, and the metabolic syndrome. Am J Physiol Endocrinol Metab. 2010;299(5):E685-94. https://doi.org/10.1152/ajpendo.00283.2010 PMid:20823452 DOI: https://doi.org/10.1152/ajpendo.00283.2010

Zámbó V, Simon-Szabó L, Szelényi P, Kereszturi E, Bánhegyi G, Csala M. Lipotoxicity in the liver. World J Hepatol. 2013;5(10):550-7. https://doi.org/10.4254/wjh.v5.i10.550 PMid:24179614 DOI: https://doi.org/10.4254/wjh.v5.i10.550

Baffy G. Kupffer cells in non-alcoholic fatty liver disease: The emerging view. J Hepatol. 2009;51(1):212-23. https://doi.org/10.1016/j.jhep.2009.03.008 PMid:19447517 DOI: https://doi.org/10.1016/j.jhep.2009.03.008

Galván-Peña S, O’Neill LA. Metabolic reprograming in macrophage polarization. Front Immunol. 2014;5:420. https://doi.org/10.3389/fimmu.2014.00420 PMid:25228902 DOI: https://doi.org/10.3389/fimmu.2014.00420

Tilg H, Diehl AM. Cytokines in alcoholic and nonalcoholic steatohepatitis. N Engl J Med. 2000;343(20):1467-76. https://doi.org/10.1056/nejm200011163432007 PMid:11078773 DOI: https://doi.org/10.1056/NEJM200011163432007

Ye J, Gao Zh, Yin J, He Q. Hypoxia is a potential risk factor for chronic inflammation and adiponectin reduction in adipose tissue of ob/ob and dietary obese mice. Am J Physiol Endocrinol Metab. 2007;293(4):E1118-28. https://doi.org/10.1152/ajpendo.00435.2007 PMid:17666485 DOI: https://doi.org/10.1152/ajpendo.00435.2007

Yefuni SN, Shpector VA. Hypoxic states and their classification. Anesteziol Reanimatol. 1981;2:3-12.

Calderon R, Llerena LA, Munive L, Kruger F. Intravenous glucose tolerance test in pregnancy in women living in chronic hypoxia. Diabetes. 1966;15(2):130-2. https://doi.org/10.2337/diab.15.2.130 PMid:5907157 DOI: https://doi.org/10.2337/diab.15.2.130

Davidson MB, Aoki VS. Fasting glucose homeostasis in rats after chronic exposure to hypoxia. Am J Physiol. 1970;219(2):378-83. https://doi.org/10.1152/ajplegacy.1970.219.2.378 PMid:5448067 DOI: https://doi.org/10.1152/ajplegacy.1970.219.2.378

Larsen JJ, Hansen JM, Olsen NV, Galbo H, Dela F. The effect of altitude hypoxia on glucose homeostasis in men. J Physiol. 1997;504(1):241-9. https://doi.org/10.1111/j.1469-7793.1997.241bf.x PMid:9350634 DOI: https://doi.org/10.1111/j.1469-7793.1997.241bf.x

Polotsky VY, Li J, Punjabi NM, Rubin AE, Smith PL, Schwartz AR, et al. Intermittent hypoxia increases insulin resistance in genetically obese mice. J Physiol. 2003;552(1):253-64. https://doi.org/10.1113/jphysiol.2003.048173 PMid:12878760 DOI: https://doi.org/10.1113/jphysiol.2003.048173

Siques P, Brito J, Naveas N, Pulido R, De la Cruz JJ, Mamani M, et al. Plasma and liver lipid profiles in rats exposed to chronic hypobaric hypoxia: Changes in metabolic pathways. High Alt Med Biol. 2014;15(3):388-95. https://doi.org/10.1089/ham.2013.1134 PMid:25185022 DOI: https://doi.org/10.1089/ham.2013.1134

Downloads

Published

2021-11-19

How to Cite

1.
Toktogulova N, Tuhvatshin R. Features of the Course of Non-alcoholic Fatty Liver Disease in Experimental Animals at High Altitudes. Open Access Maced J Med Sci [Internet]. 2021 Nov. 19 [cited 2024 Mar. 28];9(A):1092-6. Available from: https://oamjms.eu/index.php/mjms/article/view/7506