Next Article in Journal
Controlling Liquid Crystal Configuration and Phase Using Multiple Molecular Triggers
Next Article in Special Issue
Curcuma phaeocaulis Inhibits NLRP3 Inflammasome in Macrophages and Ameliorates Nanoparticle-Induced Airway Inflammation in Mice
Previous Article in Journal
A New Butyrate Releaser Exerts a Protective Action against SARS-CoV-2 Infection in Human Intestine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Comparison of the Gene Expression Profiles of Non-Alcoholic Fatty Liver Disease between Animal Models of a High-Fat Diet and Methionine-Choline-Deficient Diet

by
Mohammed Abdullah Alshawsh
1,*,
Abdulsamad Alsalahi
1,
Salah Abdalrazak Alshehade
2,
Sultan Ayesh Mohammed Saghir
3,
Ahmad Faheem Ahmeda
4,5,
Raghdaa Hamdan Al Zarzour
2 and
Ayman Moawad Mahmoud
6
1
Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
2
Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia (USM), Gelugor 11800, Malaysia
3
Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Medical Sciences, Al-Hussein Bin Talal University, Ma’an 71111, Jordan
4
Department of Basic Medical Sciences, College of Medicine, Ajman University, Ajman P.O. Box 346, United Arab Emirates
5
Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
6
Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(3), 858; https://doi.org/10.3390/molecules27030858
Submission received: 31 December 2021 / Revised: 21 January 2022 / Accepted: 24 January 2022 / Published: 27 January 2022

Abstract

:
Non-alcoholic fatty liver disease (NAFLD) embraces several forms of liver disorders involving fat disposition in hepatocytes ranging from simple steatosis to the severe stage, namely, non-alcoholic steatohepatitis (NASH). Recently, several experimental in vivo animal models for NAFLD/NASH have been established. However, no reproducible experimental animal model displays the full spectrum of pathophysiological, histological, molecular, and clinical features associated with human NAFLD/NASH progression. Although methionine-choline-deficient (MCD) diet and high-fat diet (HFD) models can mimic histological and metabolic abnormalities of human disease, respectively, the molecular signaling pathways are extremely important for understanding the pathogenesis of the disease. This review aimed to assess the differences in gene expression patterns and NAFLD/NASH progression pathways among the most common dietary animal models, i.e., HFD- and MCD diet-fed animals. Studies showed that the HFD and MCD diet could induce either up- or downregulation of the expression of genes and proteins that are involved in lipid metabolism, inflammation, oxidative stress, and fibrogenesis pathways. Interestingly, the MCD diet model could spontaneously develop liver fibrosis within two to four weeks and has significant effects on the expression of genes that encode proteins and enzymes involved in the liver fibrogenesis pathway. However, such effects in the HFD model were found to occur after 24 weeks with insulin resistance but appear to cause less severe fibrosis. In conclusion, assessing the abnormal gene expression patterns caused by different diet types provides valuable information regarding the molecular mechanisms of NAFLD/NASH and predicts the clinical progression of the disease. However, expression profiling studies concerning genetic variants involved in the development and progression of NAFLD/NASH should be conducted.

1. Introduction

As a term, non-alcoholic fatty liver disease (NAFLD) was proposed to describe the histopathological fatty changes in hepatocytes in which alcohol consumption is not involved as the etiological inducer [1]. In general, NAFLD embraces several forms of liver disorders involving fat disposition in hepatocytes ranging from the simple steatosis to the advanced stage, namely, non-alcoholic steatohepatitis (NASH) [2]. Moreover, NAFLD is known as the hepatic manifestation of metabolic syndrome since it is associated with metabolic disorders, such as obesity, type 2 diabetes mellitus, dyslipidemia, and insulin resistance [1]. NASH is characterized by the finding of hepatic steatosis with inflammation due to the excessive disposition of fats in hepatocytes, which makes hepatocytes predisposed to oxidative stress and the subsequent inflammatory and fibrosis cascades [3]. However, obesity or simple hepatic steatosis is normally not sufficient to develop inflammation and fibrosis, thus the “second-hit” hypothesis was proposed to further overstate the liver injury [4]. On the other hand, NAFLD is considered a multisystem disease as it is significantly related to extra-hepatic complications, including type 2 diabetes, chronic kidney disease [5], cardiovascular disease [6], and even neurological diseases [7]. These complications make it difficult to develop a comprehensive experimental model that fully mimics the underlying mechanisms of NAFLD. Depending on the study objectives, many models have been developed to elucidate the pathophysiological mechanisms of NAFLD, including genetically modified animals, such as leptin deficiency (ob/ob) mice [6], leptin receptor (LepR) deficiency (db/db) mice [8], KK-Ay mice [9], phosphatase and tensin homolog (PTEN)-deficient mice [10], CD36-deficient mice [11], peroxisome proliferator-activated receptor alpha (PPAR-α) knockout mice [12], acyl CoA oxidase (AOX)-null mice [13], methionine adenosyltransferase-1A (MAT1A)-null mice [14], nuclear factor erythroid 2-related factor 2 (Nrf2)-deficient mice [15], Zucker (fa/fa) rats [16], Otsuka Long-Evans Tokushima Fatty (OLETF) rats [17], and Koletsky f/f rats [18]. Furthermore, NAFLD can be induced by feeding mice different kinds of diets, including a high-fat diet (HFD) [19], high-carbohydrate diet (HCD) [20], methionine- and choline-deficient (MCD) diet [21], and fast food (FF) diet [22]. Furthermore, to reach the NASH and fibrosis stage, some models need to be triggered by a second stimulus or agent (“second hit”), such as tunicamycin [23], dexamethasone [24], and carbon tetrachloride (CCl4) [25]. It was found that male C57BL/6J mice fed a high-fat, -sucrose, and -cholesterol diet with high fructose or glucose water and injected with CCl4 showed the closest similarity to the human NAFLD pattern [26]. However, the most common dietary animal models used to elucidate the molecular and cellular progression of NAFLD either induce hepatotoxicity with MCD or over-nutrition disorders with HFD [4] as both cover most NAFLD manifestations. Although a significant number of studies regarding the cellular and molecular pathogenicity of NAFLD/NASH have been conducted on those two dietary animal models, gene expression could provide an extra essential approach to elucidate the complex pathogenesis of NAFLD. Consequently, the current review aims to outline the differences in the gene expression profiles between HFD- and MCD diet-fed animal models, which can provide valuable information as both models represent a different manifestation to obtain a comprehensive understanding of the progression of NAFLD/NASH.

2. Challenges Encountered in NAFLD Animal Models

In humans, NAFLD is the outcome of a complex of genetic and environmental factors [9]. Since NAFLD is a polygenic disease, a full understanding of its pathogenicity has still been limited because of the genetic heterogeneity within populations, the suppression of a certain gene by another [27], and ethical issues regarding exposing humans to the whole experimental approaches and techniques during the study of NAFLD compared to what animals are exposed to [28]. For such reasons, several animal models were developed to achieve a better understanding of NAFLD. Firstly, relative similarities regarding physiological, metabolic, and anatomical features exist between humans and animals. Secondly, animal models provide a valuable opportunity to study the pathogenesis of NAFLD in a single homogeneous population since the effects of variations in age, genetic heterogeneity, gender, and diet are minimized in animal models [27]. In addition, the safer and easier collection of liver biopsies is an advantage of animal models over humans [28]. Nonetheless, an animal model that successfully reflects human NASH should feature the pathological profile of NASH, such as steatosis, inflammation, and fibrosis [3]. The latter fact directs us to another challenge: how to distinguish between bland steatosis and steatohepatitis. Simple steatosis and steatohepatitis share similar metabolic determinants, which supports the idea of looking for clear-cut metabolic boundaries to distinguish steatosis from steatohepatitis [28] and which animal model could solve such a challenge or achieve the transition from simple steatosis to steatohepatitis exactly as it develops in humans [28]. It has been reported that bland steatosis is well recognized by the presence of fatty infiltration while steatohepatitis involves the presence of both fatty infiltration and inflammation [29].
Regardless of the pathogenesis of NASH, over-nutrition is the critical factor responsible for the development of NAFLD since it leads to the most common metabolic disorders, namely, insulin resistance, glucose intolerance, obesity, and dyslipidemia, which are considered risk factors for the establishment of NASH [28]. Simple steatosis usually develops after insulin resistance, which results in a reversible cumulative disposition of fat in hepatocytes due to increases in lipid delivery to hepatocytes and the uptake of lipids by hepatocytes, an increase in the biosynthesis of triglycerides and fatty acids in the cytoplasm of hepatocytes, failure in the biosynthesis of very-low-density lipoprotein, and the export of triglycerides and the impairment in β-oxidation of hepatic mitochondria. To achieve the transition of steatosis to NASH, the combination of oxidative stress, lipid peroxidation, cell death, and proinflammatory cytokine-mediated liver injury constitutes the second hit [27]. Therefore, many etiological pathways are involved in the development of NASH, including mainly oxidative stress, lipid peroxidation, and inflammation [30]. Although the genetically altered ob/ob mice is the most common model used for studying obesity and NAFLD, it does not acquire NASH spontaneously and requires a second hit with lipopolysaccharide to trigger inflammatory events [31].
HFD- and MCD-fed animal models were found to be appropriate models for the study of NAFLD/NASH for humans since the HFD-fed animal model can mimic the metabolic abnormalities of NAFLD and other spectra of oxidative stress and inflammation but is unable to reach advanced stages, such as fibrosis and cirrhosis [4]. The MCD-fed animal model provides the histological hallmark of NASH because of its vulnerability to transition from simple steatosis to steatohepatitis and can reach fibrosis stages [4]. Furthermore, the HFD model develops insulin resistance [32] unlike the MCD-fed model, which shows lower insulin resistance levels [33]. Further, the HFD-fed model cannot mimic the whole features of histological changes that are associated with NASH as in humans while the MCD model fails to establish the transition from NAFLD (simple steatosis) to the advanced stage (NASH) [34]. Interestingly, the histological changes in the liver can be recovered among HFD-fed animals but not among MCD-fed animals after stopping the tested diet [35]. Table 1 shows a comparison between the diet models. However, the effects may differ depending on the mice strains, for example, feeding MCD to C57BL/6 mice [36], FVB/NJ mice [33], and db/db mice [37] resulted in noticeably lower serum insulin levels compared to chow diet-fed mice, but db/m mice showed a slight increase in serum insulin levels [37]. Moreover, to emphasize the important difference between intracellular oxidative stress caused by different types of diet, a recent study compared the mitochondrial (mt)DNA content between two mice models. A significantly lower mtDNA copy number was found in the MCD group compared to the HFD group, which could be attributed to the upregulation of both mitochondrial biogenesis and degradation-related genes. Interestingly, after stopping feeding, the MCD-fed group showed irreversible and consistently low levels of mtDNA, unlike the HFD group, which recovered to similar levels to those observed in the control chow-fed mice [38]. This finding justifies the increase in body weight with time and fat accumulation without fibrosis among the HFD group while a decrease in body weight and steatohepatitis with fibrosis was observed among the MCD group [35]. In general, high liver steatosis results in a higher average mtDNA copy number [39] while high liver steatohepatitis results in lower mtDNA copy numbers [40].

3. Molecular Signaling Pathways of NAFLD/NASH

The changes in the gene expression profile of NAFLD associated with HFD or MCD-fed models among different studies are discussed and categorized into four different molecular pathways, namely, lipid metabolism, inflammation, oxidative stress, and fibrogenesis. Moreover, the expressions of the key genes encoding for these four pathways are discussed in detail in the following sections.

3.1. Lipid Metabolism Pathway

Studies showed that HFD could induce either up- or downregulation of the expression of genes encoding proteins or enzymes that are involved in lipogenesis [42,43,44,45,46,47]. In addition, upregulation was associated with genes encoding hepatic cholesterol [44], leptin [46], and fatty acid biosynthesis [42,44,45] while downregulation was associated with genes encoding proteins or enzymes involved in hepatic β-oxidation [42,44,45], cholesterol clearance [42,43], and insulin resistance [30,47]. Moreover, the onset of changes in gene expression was delayed, i.e., 8–24 weeks after exposure to HFD (Table 2). Such changes in the gene expression of the lipid metabolism pathway indicated that feeding animals a HFD for a longer period could primarily induce simple steatosis with insulin resistance similar to the early stage of NAFLD in humans. On the other hand, feeding animals an MCD diet could induce lipogenesis to a limited extent as evidenced by the overexpression of a limited number of genes encoding proteins or enzymes involved in lipogenesis [48,49]. In addition, it seems that changes in the gene expression of lipid metabolism pathways were more restricted to adipocytes rather than hepatocytes. Moreover, the onset of changes in such genes involved in the lipid metabolism pathway occurred much earlier than in the HFD animal model (Table 2).

3.2. Inflammatory Pathway

It seems that most key regulatory genes involved in the inflammation pathway are upregulated by HFD following the initiation of lipogenesis [22,44,45,50]. Particularly, TNF-α was found to be the gene that was expressed earlier compared to the other genes encoding other inflammatory markers (Table 3). Similarly, genes encoding proteins involved in the inflammation pathway are upregulated by the MCD diet (Table 3). However, the gene expression of inflammatory markers in the MCD diet animal model occurs much earlier than that in the HFD animal model.

3.3. Oxidative Stress Pathway

Genes encoding proteins or enzymes involved in oxidative stress pathways are subjected to either up- or downregulation by HFD. Generally, genes encoding antioxidant enzymes, such as GPX1 and CAT, are downregulated by HFD [30] while genes encoding proteins and enzymes involved in the oxidative stress pathway are upregulated [22,44,45] (Table 4). In addition, such changes in the gene expression of oxidative stress occur mostly after a longer period of exposure to HFD (16–25 weeks). On the other hand, few studies have shown that the changes in the expression of genes encoding proteins involved in oxidative stress are more prominent in the MCD diet animal model (Table 4).

3.4. Fibrogenesis Pathway

NASH with fibrosis is characterized by extensive accumulation of the extracellular matrix (ECM), which is usually followed by extensive tissue damage [57]. The fibrogenesis process in the liver is thought to mainly be regulated by hepatic stellate cells [58], and its activation produces collagen I (COL I), collagen III (COL III), and transforming growth factor-β (TGF-β) [4] in response to overexpression of X-box-binding protein 1 (XBP1) [59], NACHT, leucine-rich-repeat, pyrin domain-containing protein 3 (NLRP3) [60], and platelet-derived growth factor (PDGF) [57]. Collagen is the principal component of the extracellular matrix or scars. A recent study showed that NASH with fibrosis is associated with upregulation of hepatic fibrillar collagen genes, namely, COL I and COL III [61]. Most studies on animals fed an HFD diet for a duration of less than 4 months showed that there were no significant changes in the expression of genes encoding proteins involved in the fibrogenesis pathway; however, such changes were found to be significant in studies with a longer duration from 24-25 weeks of exposure to HFD [22,45] (Table 5). On the other hand, the MCD diet was able to induce significant changes in the expression of genes that encode proteins involved in the fibrogenesis pathway much earlier than HFD and most of the related genes, such as COL1A1, COL1A2, MMP-9, MMP-13, TIMP-1, and TGF-β, were upregulated within 2 weeks of feeding with the MCD diet [51,52] (Table 5).

4. Conclusions

In conclusion, both models reflect different changes in the expression of genes encoding proteins and enzymes involved in several pathways of NAFLD/NASH pathogenesis. The MCD diet animal model can spontaneously develop liver injury characterized by a fibrosis pattern in a short period while the HFD animal model requires a longer duration of feeding with HFD to stimulate the progression of steatosis to mild steatohepatitis. In this study, it is clear that the difference between the two dietary animal models may be related to the differences in the animal strains and as a result, it might also be associated with differences in the genetic makeup of individual patients. The variation between HFD- and MCD diet-fed animal models in the patterns of gene expression involved in the development of NAFLD and steatohepatitis provides an insight into the molecular mechanisms of NAFLD/NASH and could help to find a selective target for an effective treatment. Further expression profiling studies concerning the genetic variants involved in both the development and progression of NAFLD/NASH using suitable animal models should be conducted.

Author Contributions

Conceptualization, M.A.A., A.A. and S.A.A.; methodology and data collection, A.A. and S.A.A.; resources, M.A.A., S.A.M.S., A.F.A., R.H.A.Z., A.M.M.; supervision, M.A.A., R.H.A.Z.; Original draft preparation, A.A. and S.A.A.; review and editing, M.A.A., S.A.M.S., A.F.A., R.H.A.Z., A.M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by a research grant from the University of Malaya, Malaysia, project number (ST070-2021).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rinella, M.E. Nonalcoholic fatty liver disease a systematic review. JAMA-J. Am. Med. Assoc. 2015, 313, 2263–2273. [Google Scholar] [CrossRef] [PubMed]
  2. Mota, M.; Banini, B.A.; Cazanave, S.C.; Sanyal, A.J. Molecular mechanisms of lipotoxicity and glucotoxicity in nonalcoholic fatty liver disease. Metabolism. 2016, 65, 1049–1061. [Google Scholar] [CrossRef] [Green Version]
  3. Wree, A.; Marra, F. The inflammasome in liver disease. J. Hepatol. 2016, 65, 1055–1056. [Google Scholar] [CrossRef] [PubMed]
  4. Peng, C.; Stewart, A.G.; Woodman, O.L.; Ritchie, R.H.; Qin, C.X. Non-Alcoholic Steatohepatitis: A Review of Its Mechanism, Models and Medical Treatments. Front. Pharmacol. 2020, 11, 603926. [Google Scholar] [CrossRef]
  5. Byrne, C.D.; Targher, G. NAFLD: A multisystem disease. J. Hepatol. 2015, 62, S47–S64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Targher, G.; Corey, K.E.; Byrne, C.D. NAFLD, and cardiovascular and cardiac diseases: Factors influencing risk, prediction and treatment. Diabetes Metab. 2021, 47, 101215. [Google Scholar] [CrossRef] [PubMed]
  7. Hadjihambi, A. Cerebrovascular alterations in NAFLD: Is it increasing our risk of Alzheimer’s disease? Anal. Biochem. 2022, 636, 114387. [Google Scholar] [CrossRef] [PubMed]
  8. Kondo, Y.; Hasegawa, G.; Okada, H.; Senmaru, T.; Fukui, M.; Nakamura, N.; Sawada, M.; Kitawaki, J.; Okanoue, T.; Kishimoto, Y.; et al. Leprdb/db Mice with Senescence Marker Protein-30 Knockout (Leprdb/dbSmp30Y/-) Exhibit Increases in Small Dense-LDL and Severe Fatty Liver Despite Being Fed a Standard Diet. PLoS ONE 2013, 8, e65698. [Google Scholar] [CrossRef] [Green Version]
  9. Okumura, K.; Ikejima, K.; Kon, K.; Abe, W.; Yamashina, S.; Enomoto, N.; Takei, Y.; Sato, N. Exacerbation of dietary steatohepatitis and fibrosis in obese, diabetic KK-Ay mice. Hepatol. Res. 2006, 36, 217–228. [Google Scholar] [CrossRef]
  10. Sato, W.; Horie, Y.; Kataoka, E.; Ohshima, S.; Dohmen, T.; Iizuka, M.; Sasaki, J.; Sasaki, T.; Hamada, K.; Kishimoto, H.; et al. Hepatic gene expression in hepatocyte-specific Pten deficient mice showing steatohepatitis without ethanol challenge. Hepatol. Res. 2006, 34, 256–265. [Google Scholar] [CrossRef]
  11. Rada, P.; González-Rodríguez, Á.; García-Monzón, C.; Valverde, Á.M. Understanding lipotoxicity in NAFLD pathogenesis: Is CD36 a key driver? Cell Death Dis. 2020, 11, 802. [Google Scholar] [CrossRef] [PubMed]
  12. Wang, Y.; Nakajima, T.; Gonzalez, F.J.; Tanaka, N. PPARs as metabolic regulators in the liver: Lessons from liver-specific PPAR-null mice. Int. J. Mol. Sci. 2020, 21, 2061. [Google Scholar] [CrossRef] [Green Version]
  13. Cook, W.S.; Jain, S.; Jia, Y.; Cao, W.Q.; Yeldandi, A.V.; Reddy, J.K.; Rao, M.S. Peroxisome proliferator-activated receptor α-responsive genes induced in the newborn but not prenatal liver of peroxisomal fatty acyl-CoA oxidase null mice. Exp. Cell Res. 2001, 268, 70–76. [Google Scholar] [CrossRef]
  14. Lu, S.C.; Alvarez, L.; Huang, Z.Z.; Chen, L.; An, W.; Corrales, F.J.; Avila, M.A.; Kanel, G.; Mato, J.M. Methionine adenosyltransferase 1A knockout mice are predisposed to liver injury and exhibit increased expression of genes involved in proliferation. Proc. Natl. Acad. Sci. USA 2001, 98, 5560–5565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Chowdhry, S.; Nazmy, M.H.; Meakin, P.J.; Dinkova-Kostova, A.T.; Walsh, S.V.; Tsujita, T.; Dillon, J.F.; Ashford, M.L.J.; Hayes, J.D. Loss of Nrf2 markedly exacerbates nonalcoholic steatohepatitis. Free Radic. Biol. Med. 2010, 48, 357–371. [Google Scholar] [CrossRef] [PubMed]
  16. Matsumoto, Y.; Fujita, S.; Yamagishi, A.; Shirai, T.; Maeda, Y.; Suzuki, T.; Kobayashi, K.I.; Inoue, J.; Yamamoto, Y. Brown Rice Inhibits Development of Nonalcoholic Fatty Liver Disease in Obese Zucker (fa/fa) Rats by Increasing Lipid Oxidation Via Activation of Retinoic Acid Synthesis. J. Nutr. 2021, 151, 2705–2713. [Google Scholar] [CrossRef] [PubMed]
  17. Song, Y.S.; Fang, C.H.; So, B.I.; Park, J.Y.; Lee, Y.; Shin, J.H.; Jun, D.W.; Kim, H.; Kim, K.S. Time course of the development of nonalcoholic fatty liver disease in the otsuka long-evans tokushima fatty rat. Gastroenterol. Res. Pract. 2013, 2013, 342648. [Google Scholar] [CrossRef] [Green Version]
  18. Rhinehart, E.K.; Kalra, S.P.; Kalra, P.S. Neuropeptidergic characterization of the leptin receptor mutated obese Koletsky rat. Regul. Pept. 2004, 119, 3–10. [Google Scholar] [CrossRef]
  19. Al Zarzour, R.; Alshawsh, M.; Asif, M.; Al-Mansoub, M.; Mohamed, Z.; Ahmad, M.; Abdul Majid, A.; Asmawi, M.; Kaur, G.; Al-dualimi, D.; et al. Adipocytokine Regulation and Antiangiogenic Activity Underlie the Molecular Mechanisms of Therapeutic Effects of Phyllanthus niruri against Non-Alcoholic Fatty Liver Disease. Nutrients 2018, 10, 1057. [Google Scholar] [CrossRef] [Green Version]
  20. Prisingkorn, W.; Prathomya, P.; Jakovlić, I.; Liu, H.; Zhao, Y.H.; Wang, W.M. Transcriptomics, metabolomics and histology indicate that high-carbohydrate diet negatively affects the liver health of blunt snout bream (Megalobrama amblycephala). BMC Genomics 2017, 18, 856. [Google Scholar] [CrossRef] [Green Version]
  21. Lan, Q.; Ren, Z.; Chen, Y.; Cui, G.; Choi, I.C.; Ung, C.O.L.; Yu, H.H.; Lee, S.M.Y. Hepatoprotective effect of Qushihuayu formula on non-alcoholic steatohepatitis induced by MCD diet in rat. Chinese Med. 2021, 16, 27. [Google Scholar] [CrossRef] [PubMed]
  22. Charlton, M.; Krishnan, A.; Viker, K.; Sanderson, S.; Cazanave, S.; McConico, A.; Al., E.; Masuoko, H.; Gores, G. Fast food diet mouse: Novel small animal model of NASH with ballooning, progressive fibrosis, and high physiological fidelity to the human condition. Am. J. Physiol.-Gastrointest. Liver Physiol. 2011, 301, G825–G834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Kim, S.H.; Kwon, D.Y.; Kwak, J.H.; Lee, S.; Lee, Y.H.; Yun, J.; Son, T.G.; Jung, Y.S. Tunicamycin-induced ER stress is accompanied with oxidative stress via abrogation of sulfur amino acids metabolism in the liver. Int. J. Mol. Sci. 2018, 19, 4114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Rahimi, L.; Rajpal, A.; Ismail-Beigi, F. Glucocorticoid-induced fatty liver disease. Diabetes, Metab. Syndr. Obes. Targets Ther. 2020, 13, 1133–1145. [Google Scholar] [CrossRef] [Green Version]
  25. Zhang, G.; Wang, X.; Chung, T.Y.; Ye, W.; Hodge, L.; Zhang, L.; Chng, K.; Xiao, Y.F.; Wang, Y.J. Carbon tetrachloride (CCl4) accelerated development of non-alcoholic fatty liver disease (NAFLD)/steatohepatitis (NASH) in MS-NASH mice fed western diet supplemented with fructose (WDF). BMC Gastroenterol. 2020, 20, 339. [Google Scholar] [CrossRef]
  26. Tsuchida, T.; Lee, Y.A.; Fujiwara, N.; Ybanez, M.; Allen, B.; Martins, S.; Fiel, M.I.; Goossens, N.; Chou, H.I.; Hoshida, Y.; et al. A simple diet- and chemical-induced murine NASH model with rapid progression of steatohepatitis, fibrosis and liver cancer. J. Hepatol. 2018, 69, 385–395. [Google Scholar] [CrossRef]
  27. Anstee, Q.M.; Goldin, R.D. Mouse models in non-alcoholic fatty liver disease and steatohepatitis research. Int. J. Exp. Pathol. 2006, 87, 1–16. [Google Scholar] [CrossRef]
  28. Larter, C.Z.; Yeh, M.M. Animal models of NASH: Getting both pathology and metabolic context right. J. Gastroenterol. Hepatol. 2008, 23, 1635–1648. [Google Scholar] [CrossRef] [Green Version]
  29. Hebbard, L.; George, J. Animal models of nonalcoholic fatty liver disease. Nat. Rev. Gastroenterol. Hepatol. 2011, 8, 34–44. [Google Scholar] [CrossRef]
  30. Tipoe, G.L.; Ho, C.T.; Liong, E.C.; Leung, T.M.; Lau, T.Y.H.; Fung, M.L.; Nanji, A.A. Voluntary oral feeding of rats not requiring a very high fat diet is a clinically relevant animal model of non-alcoholic fatty liver disease (NAFLD). Histol. Histopathol. 2009, 24, 1161–1169. [Google Scholar] [CrossRef]
  31. Wouters, K.; van Gorp, P.J.; Bieghs, V.; Gijbels, M.J.; Duimel, H.; Lütjohann, D.; Kerksiek, A.; van Kruchten, R.; Maeda, N.; Staels, B.; et al. Dietary cholesterol, rather than liver steatosis, leads to hepatic inflammation in hyperlipidemic mouse models of nonalcoholic steatohepatitis. Hepatology 2008, 48, 474–486. [Google Scholar] [CrossRef] [PubMed]
  32. Lee, K.C.; Hsieh, Y.C.; Yang, Y.Y.; Chan, C.C.; Huang, Y.H.; Lin, H.C. Aliskiren Reduces Hepatic steatosis and Epididymal Fat Mass and Increases Skeletal Muscle Insulin Sensitivity in High-Fat Diet-Fed Mice. Sci. Rep. 2016, 6, 18899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Rinella, M.E.; Green, R.M. The methionine-choline deficient dietary model of steatohepatitis does not exhibit insulin resistance. J. Hepatol. 2004, 40, 47–51. [Google Scholar] [CrossRef] [PubMed]
  34. Im, Y.R.; Hunter, H.; de Gracia Hahn, D.; Duret, A.; Cheah, Q.; Dong, J.; Fairey, M.; Hjalmarsson, C.; Li, A.; Lim, H.K.; et al. A Systematic Review of Animal Models of NAFLD Finds High-Fat, High-Fructose Diets Most Closely Resemble Human NAFLD. Hepatology 2021, 74, 1884–1901. [Google Scholar] [CrossRef] [PubMed]
  35. Itagaki, H.; Shimizu, K.; Morikawa, S.; Ogawa, K.; Ezaki, T. Morphological and functional characterization of non-alcoholic fatty liver disease induced by a methionine-choline-deficient diet in C57BL/6 mice. Int. J. Clin. Exp. Pathol. 2013, 6, 2683–2696. [Google Scholar]
  36. Kim, S.H.; Lim, Y.; Bin Park, J.; Kwak, J.-H.; Kim, K.-J.; Kim, J.-H.; Song, H.; Cho, J.Y.; Hwang, D.Y.; Kim, K.S.; et al. Comparative study of fatty liver induced by methionine and choline-deficiency in C57BL/6N mice originating from three different sources. Lab. Anim. Res. 2017, 33, 157. [Google Scholar] [CrossRef] [Green Version]
  37. Rinella, M.E.; Elias, M.S.; Smolak, R.R.; Fu, T.; Borensztajn, J.; Green, R.M. Mechanisms of hepatic steatosis in mice fed a lipogenic methionine choline-deficient diet. J. Lipid Res. 2008, 49, 1068–1076. [Google Scholar] [CrossRef] [Green Version]
  38. Arao, Y.; Kawai, H.; Kamimura, K.; Kobayashi, T.; Nakano, O.; Hayatsu, M.; Ushiki, T.; Terai, S. Effect of methionine/choline-deficient diet and high-fat diet-induced steatohepatitis on mitochondrial homeostasis in mice. Biochem. Biophys. Res. Commun. 2020, 527, 365–371. [Google Scholar] [CrossRef]
  39. Kamfar, S.; Alavian, S.M.; Houshmand, M.; Yadegarazari, R.; Zarei, B.S.; Khalaj, A.; Shabab, N.; Saidijam, M. Liver mitochondrial DNA copy number and deletion levels may contribute to nonalcoholic fatty liver disease susceptibility. Hepat. Mon. 2016, 16, e40774. [Google Scholar] [CrossRef] [Green Version]
  40. Pirola, C.J.; Scian, R.; Gianotti, T.F.; Dopazo, H.; Rohr, C.; Martino, J.S.; Castaño, G.O.; Sookoian, S. Epigenetic modifications in the biology of nonalcoholic fatty liver disease: The role of DNA hydroxymethylation and TET proteins. Medicine 2015, 94, e1480. [Google Scholar] [CrossRef]
  41. Chiba, T.; Suzuki, S.; Sato, Y.; Itoh, T.; Umegaki, K. Evaluation of methionine content in a high-fat and choline-deficient diet on body weight gain and the development of non-alcoholic steatohepatitis in mice. PLoS ONE 2016, 11, e0164191. [Google Scholar] [CrossRef] [PubMed]
  42. Yang, D.J.; Chang, Y.Y.; Hsu, C.L.; Liu, C.W.; Lin, Y.I.L.; Lin, Y.U.H.; Liu, K.C.; Chen, Y.I.C. Antiobesity and hypolipidemic effects of polyphenol-rich longan (dimocarpus longans lour.) flower water extract in hypercaloric-dietary rats. J. Agric. Food Chem. 2010, 58, 2020–2027. [Google Scholar] [CrossRef] [PubMed]
  43. Dutta, P.K.; Tripathi, S.; Mehrotra, G.K.; Dutta, J. Perspectives for chitosan based antimicrobial films in food applications. Food Chem. 2009, 114, 1173–1182. [Google Scholar] [CrossRef]
  44. Xie, Z.; Li, H.; Wang, K.; Lin, J.; Wang, Q.; Zhao, G.; Jia, W.; Zhang, Q. Analysis of transcriptome and metabolome profiles alterations in fatty liver induced by high-fat diet in rat. Metabolism. 2010, 59, 554–560. [Google Scholar] [CrossRef]
  45. Matsuzawa, N.; Takamura, T.; Kurita, S.; Misu, H.; Ota, T.; Ando, H.; Yokoyama, M.; Honda, M.; Zen, Y.; Nakanuma, Y.; et al. Lipid-induced oxidative stress causes steatohepatitis in mice fed an atherogenic diet. Hepatology 2007, 46, 1392–1403. [Google Scholar] [CrossRef]
  46. Milagro, F.I.; Campión, J.; Martíez, J.A. Weight gain induced by high-fat feeding involves increased liver oxidative stress. Obesity 2006, 14, 1118–1123. [Google Scholar] [CrossRef]
  47. Xing, L.J.; Zhang, L.; Liu, T.; Hua, Y.Q.; Zheng, P.Y.; Ji, G. Berberine reducing insulin resistance by up-regulating IRS-2 mRNA expression in nonalcoholic fatty liver disease (NAFLD) rat liver. Eur. J. Pharmacol. 2011, 668, 467–471. [Google Scholar] [CrossRef]
  48. Larter, C.Z.; Yeh, M.M.; Williams, J.; Bell-Anderson, K.S.; Farrell, G.C. MCD-induced steatohepatitis is associated with hepatic adiponectin resistance and adipogenic transformation of hepatocytes. J. Hepatol. 2008, 49, 407–416. [Google Scholar] [CrossRef]
  49. Nagasawa, T.; Inada, Y.; Nakano, S.; Tamura, T.; Takahashi, T.; Maruyama, K.; Yamazaki, Y.; Kuroda, J.; Shibata, N. Effects of bezafibrate, PPAR pan-agonist, and GW501516, PPARδ agonist, on development of steatohepatitis in mice fed a methionine- and choline-deficient diet. Eur. J. Pharmacol. 2006, 536, 182–191. [Google Scholar] [CrossRef]
  50. Wang, Y.; Ausman, L.M.; Russell, R.M.; Greenberg, A.S.; Wang, X.D. Increased apoptosis in high-fat diet-induced nonalcoholic steatohepatitis in rats is associated with c-Jun NH2-terminal kinase activation and elevated proapoptotic bax. J. Nutr. 2008, 138, 1866–1871. [Google Scholar] [CrossRef] [Green Version]
  51. Oz, H.S.; Chen, T.S.; Neuman, M. Methionine deficiency and hepatic injury in a dietary steatohepatitis model. Dig. Dis. Sci. 2008, 53, 767–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. George, J.; Pera, N.; Phung, N.; Leclercq, I.; Hou, J.Y.; Farrell, G. Lipid peroxidation, stellate cell activation and hepatic fibrogenesis in a rat model of chronic steatohepatitis. J. Hepatol. 2003, 39, 756–764. [Google Scholar] [CrossRef]
  53. Tetri, L.H.; Basaranoglu, M.; Brunt, E.M.; Yerian, L.M.; Neuschwander-Tetri, B.A. Severe NAFLD with hepatic necroinflammatory changes in mice fed trans fats and a high-fructose corn syrup equivalent. Am. J. Physiol.-Gastrointest. Liver Physiol. 2008, 295, G987–G995. [Google Scholar] [CrossRef] [PubMed]
  54. Koppe, S.W.P.; Sahai, A.; Malladi, P.; Whitington, P.F.; Green, R.M. Pentoxifylline attenuates steatohepatitis induced by the methionine choline deficient diet. J. Hepatol. 2004, 41, 592–598. [Google Scholar] [CrossRef] [PubMed]
  55. Henao-Mejia, J.; Elinav, E.; Jin, C.; Hao, L.; Mehal, W.Z.; Strowig, T.; Thaiss, C.A.; Kau, A.L.; Eisenbarth, S.C.; Jurczak, M.J.; et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012, 482, 179–185. [Google Scholar] [CrossRef] [Green Version]
  56. Deng, X.Q.; Chen, L.L.; Li, N.X. The expression of SIRT1 in nonalcoholic fatty liver disease induced by high-fat diet in rats. Liver Int. 2007, 27, 708–715. [Google Scholar] [CrossRef]
  57. Zhang, S.L.; Ma, L.; Zhao, J.; You, S.P.; Ma, X.T.; Ye, X.Y.; Liu, T. The phenylethanol glycoside liposome inhibits PDGF-induced HSC activation via regulation of the FAK/PI3K/AkT signaling pathway. Molecules 2019, 24, 3282. [Google Scholar] [CrossRef] [Green Version]
  58. Li, Z.; Zhao, L.; Xia, Y.; Chen, J.; Hua, M.; Sun, Y. Schisandrin b attenuates hepatic stellate cell activation and promotes apoptosis to protect against liver fibrosis. Molecules 2021, 26, 6882. [Google Scholar] [CrossRef]
  59. Kim, R.S.; Hasegawa, D.; Goossens, N.; Tsuchida, T.; Athwal, V.; Sun, X.; Robinson, C.L.; Bhattacharya, D.; Chou, H.I.; Zhang, D.Y.; et al. The XBP1 Arm of the Unfolded Protein Response Induces Fibrogenic Activity in Hepatic Stellate Cells Through Autophagy. Sci. Rep. 2016, 6, 39342. [Google Scholar] [CrossRef]
  60. Wree, A.; Eguchi, A.; Mcgeough, M.D.; Pena, C.A.; Johnson, C.D.; Canbay, A.; Hoffman, H.M.; Feldstein, A.E. NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. Hepatology 2014, 59, 898–910. [Google Scholar] [CrossRef] [Green Version]
  61. Pellicano, A.J.; Spahn, K.; Zhou, P.; Goldberg, I.D.; Narayan, P. Collagen characterization in a model of nonalcoholic steatohepatitis with fibrosis; a call for development of targeted therapeutics. Molecules 2021, 26, 3316. [Google Scholar] [CrossRef] [PubMed]
Table 1. A comparison between the NAFLD model mice fed MCD and HFD.
Table 1. A comparison between the NAFLD model mice fed MCD and HFD.
ComparisonHFD *MCD **HFD & MCD ***
Body weightHigherLowerNo change
Liver-to-body weight ratioNo change Slightly lowerHigher
Serum biomarkersTCHigherLowerLower
TGSlightly lowerLowerLower
ASTLowerHigherNA
GlucoseHigherLowerSlightly higher
InsulinHigherLowerLower
SteatosisHigherHigher (but not as high as HFD)Higher
FibrosisNo changeHigherNo change
Inflammation lobularSlightly higherHigherHigher
Hepatocellular ballooningSlightly higherSlightly higherNo change
* Compared (HFD 60% of energy as fat) to a standard chow diet as control ([22]); ** Compared to a standard chow diet as the control ([33,35,36]); *** Compared to an L-amino acid rodent diet as the control ([41]). TC: total cholesterols; TG: total triglycerides; AST: alanine aminotransferase; NA: not available.
Table 2. Gene expression profiles of hepatic lipid metabolism in HFD and MCD diet-fed animal models.
Table 2. Gene expression profiles of hepatic lipid metabolism in HFD and MCD diet-fed animal models.
Gene SymbolNomenclatureRoleDiet TypeDuration
(Weeks)
Animal ModelGene ExpressionReference
ADIPOQAdiponectin, C1Q, and collagen domain containingRequired for normal glucose and fat homeostasisHFD8 Female SD ratsDown[30]
ADIPOR1Adiponectin receptor 1Required for normal glucose and fat homeostasisMCD3 C57BL/6J micUp[48]
ADIPOR2Adiponectin receptor 2Required for normal glucose and fat homeostasis5 Male C57BL/6N miceUp[49]
AMPKα25′AMP-activated protein kinase catalytic subunit alpha-2Inhibits protein, carbohydrate, and lipid biosynthesis5 Male C57BL/6N miceUp
CPT-1ACarnitine palmitoyltransferase-1 alphaMitochondrial oxidation of long-chain fatty acidsHFD24 Male C57BL/6J miceDown[45]
16 Male Wistar ratsDown[44]
CYP4A10Cytochrome P450, family 4, subfamily A, polypeptide 10Involved in the metabolism of fatty acidsMCD3 C57BL/6J miceDown[48]
CYPA14Cytochrome P450, family 14, subfamily AInvolved in the metabolism of fatty acids3 C57BL/6J micUp
L-FABPLiver-type fatty acid-binding proteinPlays a role in lipoprotein-mediated cholesterol uptake4db/db miceDown[37]
4db/m miceDown
FATP-1Long-chain fatty acid transport protein 1Mediates the ATP-dependent import of long-chain fatty acids into the cell4db/m miceUp
db/db miceUp
3 C57BL/6J miceDown[48]
4db/m miceUp[37]
FATP-2Very long-chain acyl-CoA synthaseActivates long-chain and very-long-chain fatty acids
db/db miceDown
FATP-3Solute carrier family 27 member 3Acyl-CoA ligase activity for long-chain and very-long-chain fatty acidsdb/m miceUp
db/db miceDown
FATP-4Long-chain fatty acid transport protein 4Involved in the translocation of long-chain fatty acids across the plasma membranedb/m miceUp
db/db miceUp
3 C57BL/6J miceUp[48]
FATP-5Bile acyl-CoA synthaseCatalyzes the activation of bile acids via the formation of bile acid CoA thioesters4db/m miceNo change[37]
db/db miceDown
FASNFatty acid synthaseCatalyzes the de novo biosynthesis of long-chain saturated fatty acidsMCD4 db/db miceDown
db/m miceDown
HFD9Male SD ratsUp[42]
24Male C57BL/6J miceUp[45]
G3PDHGlycerol-3-phosphate dehydrogenaseGlycolysisHFD8 Male Wistar ratsUp[46]
HMGCR3-Hydroxy-3-methylglutaryl-coenzyme A reductaseCholesterol biosynthesisHFD6 Male Golden Syrian hamstersUp[43]
HMGCS13-Hydroxy-3-methylglutaryl-coenzyme A synthase 1Cholesterol biosynthesisHFD16 Male Wistar ratsDown[44]
IRS-2Insulin receptor substrate-2Controls various cellular processes by insulinHFD12 Male Wister ratsDown[47]
LDLRLow-density lipoprotein receptorClearance of cholesterolHFD9 Male SD ratsDown[42]
6 Male Golden Syrian HamstersDown[43]
LEPLeptinRegulation of energy balance and body weight controlHFD8 Male Wistar ratsUp[46]
MTMR4Myotubularin-related protein 4Phosphatase activity and protein serine/threonine phosphatase activityHFD16 Male Wistar ratsUp[44]
PPAR-APeroxisome proliferator-activated receptor alphaβ-Oxidation and energy expenditureHFD9 Male SD ratsDown[42]
16 Male Wistar ratsDown[44]
24 Male C57BL/6J miceDown[45]
PPAR-γPeroxisome proliferator-activated receptor gammaControls the peroxisomal β-oxidation pathway of fatty acidsMCD3 C57BL/6J micUp[48]
SREBF1Sterol regulatory element-binding protein 1
(isoform SREBP-1a)
Stimulates both lipogenic and cholesterogenic gene expressionHFD16 Male Wistar ratsUp[44]
9 Male SD ratsUp[42]
24 Male C57BL/6J miceUp[45]
MCD4 db/db miceDown[37]
db/m miceDown
Sterol regulatory element-binding protein 1
(Isoform SREBP-1c)
Primarily controls the expression of the lipogenic genedb/db miceUp
db/m miceDown
SCD-1Stearoyl-CoA desaturase-1Plays an important role in lipid biosynthesisMCD4db/db miceDown
db/m miceDown
HFD16 Male Wistar ratsUp[44]
Table 3. Gene expression profiles of hepatic inflammation in HFD and MCD diet-fed animal models.
Table 3. Gene expression profiles of hepatic inflammation in HFD and MCD diet-fed animal models.
Gene SymbolNomenclatureRoleDiet TypeDuration
(Weeks)
Animal ModelGene ExpressionReference
CFHComplement component factor HPlays an essential role in maintaining a well-balanced immune responseHFD16Male Wistar ratsUp[44]
COX-2Prostaglandin-endoperoxide synthase 2 (cyclooxygenase-2)A particular role in the inflammatory responseHFD8Female SD ratsUp[30]
CXCL1Chemokine (C-X-C motif) ligand 1Plays a role in inflammation and as a chemoattractant for neutrophilsHFD16Male Wistar ratsUp[44]
CXCL14Chemokine (C-X-C motif) ligand 14Chemotactic for B-lymphocytesHFD16Male Wistar ratsDown
IL-1βInterleukin-1 betaInduces prostaglandin synthesis, neutrophil influx and activation, and T cell activationMCD2Male SD ratsUp[51]
5Male C57BL/6N miceUp[49]
IL-6Interleukin-6Regulation of the immune responseMCD2Male SD ratsUp[51]
5Male C57BL/6N miceUp[49]
MCP-1Monocyte chemoattractant protein-1Exhibits chemotactic activity for monocytes and basophilsMCD5Male C57BL/6N miceUp
NF-κB1Nuclear factor-kappa B subunit 1Stimulates many biological processes such as inflammation, immunity, differentiation, cell growth, tumorigenesis, and apoptosisMCD5Male C57BL/6N miceUp
iNOS2Inducible nitric oxide synthaseInvolved in inflammation, enhances the synthesis of proinflammatory mediators, such as IL-6 and IL-8HFD8Female SD ratsUp[30]
TGF-β1Transforming growth factor-beta 1Multifunctional protein that regulates the growth and differentiation of various cell typesMCD2Male SD ratsUp[51]
5Male C57BL/6N miceUp[49]
12Male SD ratsUp[52]
TNF-αTumor necrosis factor-alphaA key mediator of cell death, and induces insulin resistanceHFD6Male SD ratsUp[50]
24Male C57BL/6J miceUp[45]
8Female SD ratsUp[30]
16Male C57BL/6 miceUp[53]
25Male C57BL/6 miceUp[22]
MCD2Male and female C57BL/6J miceUp[54]
4C57BL/6 miceUp[55]
2Male SD ratsUp[51]
5Male C57BL/6N miceUp[49]
Table 4. Gene expression profiles of hepatic oxidative stress in HFD and MCD diet-fed animal models.
Table 4. Gene expression profiles of hepatic oxidative stress in HFD and MCD diet-fed animal models.
Gene SymbolNomenclatureRoleDiet TypeDuration
(Weeks)
Animal ModelGene ExpressionReference
AOXAlternative oxidase mitochondrial precursorCatalyzes the cyanide-resistant oxidation of ubiquinolMCD4db/db miceDown[37]
db/m MiceUp
CATCatalaseProtects cells from the toxic effects of hydrogen peroxideHFD8Female SD ratsDown[30]
CHOP (DDIT3)C/EBP homologous protein (DNA damage-inducible transcript 3)Endoplasmic reticulum (ER) stress responseHFD25Male C57BL/6 miceUp[22]
CPT-1Carnitine O-palmitoyltransferase 1Plays role in mitochondrial uptake of long-chain fatty acidsMCD4db/db miceUp[37]
4db/m miceUp
CPT-2Carnitine O-palmitoyl transferase 2, mitochondrialIntra-mitochondrial synthesis of acylcarnitinesMCD4db/db miceUp
4db/m miceUp
GAB1GRB2-associated binding protein 1Plays a role in FGFR1 signalingHFD16Male Wistar ratsUp[44]
GADD45GGrowth arrest and DNA-damage-inducible, gammaMediates activation of stress-responsive MTK1/MEKK4 MAPKKKHFD16Male Wistar ratsUp
Gp91phox (CYBB)Cytochrome B-245, beta polypeptideA critical component of the membrane-bound oxidase of phagocytes that generates superoxideHFD24Male C57BL/6J miceUp[45]
GPX1Glutathione peroxidaseProtects from oxidative breakdownHFD8Female SD ratsDown[30]
LCADLong-chain specific acyl-CoA dehydrogenase, mitochondrialCatalyzes the first step of mitochondrial fatty acid beta-oxidationMCD4db/db miceNo change[37]
4db/m miceUp
L-FABPFatty acid-binding protein, liverPlays a role in lipoprotein-mediated cholesterol uptakeMCD4db/db miceDown
4db/m miceDown
NFE2L2Nuclear factor, erythroid 2-Like 2Transcription factorHFD16Male Wistar ratsUp[44]
P22phox (CYPA)Cytochrome B-245, alpha polypeptideCritical component of the membrane-bound oxidaseHFD24Male C57BL/6J miceUp[45]
P47phox (NCF1)Neutrophil cytosolic factor 1Activation of the latent NADPH oxidase (necessary for superoxide production)HFD24Male C57BL/6J miceUp
PERKProtein kinase R (PKR)-like endoplasmic reticulum kinasePlays a role in the early steps of protein synthesisHFD25Male C57BL/6 miceDown[22]
SIRT1Sirtuin 1 (silent mating type information regulation 2 homolog)Deacetylase, ADP-ribosyl transferase, and other deacylase activitiesHFD12Male Wistar ratsDown[56]
Table 5. Gene expression profiles of hepatic fibrogenesis in HFD and MCD diet-fed animal models.
Table 5. Gene expression profiles of hepatic fibrogenesis in HFD and MCD diet-fed animal models.
Gene SymbolNomenclatureRoleDiet TypeDuration
(Weeks)
Animal ModelGene ExpressionReference
ACTA2
(ASMA)
Anti-smooth muscle actinActivation to myofibroblast-like cellHFD25C57BL/6 miceUp[22]
COL1A1Collagen type 1 alpha 1Fibrillar forming collagenHFD25C57BL/6 miceUp[22]
24Male C57BL/6J miceUp[45]
MCD2Male SD ratsUp[51]
17Male SD ratsUp[52]
COL1A2Collagen type I alpha 2HFD24Male C57BL/6J miceUp[45]
COL4A1Collagen type IV alpha 1HFD24Male C57BL/6J miceUp[45]
HGFHepatocyte growth factorHepatotropic factor, which acts as a growth factorHFD25C57BL/6 miceUp[22]
LUMLumicanExtracellular matrix structural constituentHFD25C57BL/6 miceUp[22]
MMP-13Matrix metalloproteinase-13Degradation of extracellular matrix proteinsMCD2Male SD ratsUp[51]
MMP-9Matrix metalloproteinase-9MCD2Male SD ratsUp[51]
PAI-1Plasminogen activator inhibitor 1Inhibitor of tissue-type plasminogen activator (PLAT) and urokinase-type plasminogen activator (PLAU)HFD24Male C57BL/6J miceUp[45]
SOCS1Suppressor of cytokine signaling 1Prevents uncontrolled cytokine signalingMCD2Male SD ratsUp[51]
TIMP-1Tissue inhibitor matrix metalloproteinase 1 (TIMP Metallopeptidase Inhibitor 1)Inhibitor of collagenases by forming one to one complexesMCD17Male SD ratsUp[52]
HFD25C57BL/6 miceUp[22]
TGF-β1Transforming growth factor-beta 1Acts as a regulator of extracellular matrix storageHFD25C57BL/6 miceUp[22]
24Male C57BL/6J miceUp[45]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Alshawsh, M.A.; Alsalahi, A.; Alshehade, S.A.; Saghir, S.A.M.; Ahmeda, A.F.; Al Zarzour, R.H.; Mahmoud, A.M. A Comparison of the Gene Expression Profiles of Non-Alcoholic Fatty Liver Disease between Animal Models of a High-Fat Diet and Methionine-Choline-Deficient Diet. Molecules 2022, 27, 858. https://doi.org/10.3390/molecules27030858

AMA Style

Alshawsh MA, Alsalahi A, Alshehade SA, Saghir SAM, Ahmeda AF, Al Zarzour RH, Mahmoud AM. A Comparison of the Gene Expression Profiles of Non-Alcoholic Fatty Liver Disease between Animal Models of a High-Fat Diet and Methionine-Choline-Deficient Diet. Molecules. 2022; 27(3):858. https://doi.org/10.3390/molecules27030858

Chicago/Turabian Style

Alshawsh, Mohammed Abdullah, Abdulsamad Alsalahi, Salah Abdalrazak Alshehade, Sultan Ayesh Mohammed Saghir, Ahmad Faheem Ahmeda, Raghdaa Hamdan Al Zarzour, and Ayman Moawad Mahmoud. 2022. "A Comparison of the Gene Expression Profiles of Non-Alcoholic Fatty Liver Disease between Animal Models of a High-Fat Diet and Methionine-Choline-Deficient Diet" Molecules 27, no. 3: 858. https://doi.org/10.3390/molecules27030858

Article Metrics

Back to TopTop