Editorial Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Jun 8, 2015; 7(10): 1297-1301
Published online Jun 8, 2015. doi: 10.4254/wjh.v7.i10.1297
Pathophysiological mechanisms involved in non-alcoholic steatohepatitis and novel potential therapeutic targets
Fátima Higuera-de la Tijera, Liver Clinic, Gastroenterology Department, Mexico’s General Hospital “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico
Alfredo I Servín-Caamaño, Diabetes, Obesity and Metabolic Syndrome Clinic, Internal Medicine Department, Mexico’s General Hospital “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico
Author contributions: Higuera-de la Tijera F prepared and wrote the editorial; Servín-Caamaño AI made corrections and carried out final editing of the manuscript.
Conflict-of-interest: The authors involved in this manuscript (Fátima Higuera-de la Tijera, Alfredo I Servín-Caamaño) have no conflicting commercial, personal, political, intellectual, or religious interests.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Fátima Higuera-de la Tijera, MD, MSc, Liver Clinic, Gastroenterology Department, Mexico’s General Hospital “Dr. Eduardo Liceaga”, Eje 2A Sur (Dr. Balmis) 148, Cuauhtémoc, Doctores, Mexico City 06726, Mexico. fatimahiguera@yahoo.com.mx
Telephone: +52-55-27892000
Received: January 14, 2015
Peer-review started: January 18, 2015
First decision: February 7, 2015
Revised: February 21, 2015
Accepted: March 16, 2015
Article in press: March 18, 2015
Published online: June 8, 2015

Abstract

Non-alcoholic fatty liver disease (NAFLD) is a major health care problem and represents the hepatic expression of the metabolic syndrome. NAFLD is classified as non-alcoholic fatty liver (NAFL) or simple steatosis, and non-alcoholic steatohepatitis (NASH). NASH is characterized by the presence of steatosis and inflammation with or without fibrosis. The physiopathology of NAFL and NASH and their progression to cirrhosis involve several parallel and interrelated mechanisms, such as, insulin resistance (IR), lipotoxicity, inflammation, oxidative stress, and recently the gut-liver axis interaction has been described. Incretin-based therapies could play a role in the treatment of NAFLD. Glucagon-like peptide-1 (GLP-1) is an intestinal mucosa-derived hormone which is secreted into the bloodstream in response to nutrient ingestion; it favors glucose-stimulated insulin secretion, inhibition of postprandial glucagon secretion and delayed gastric emptying. It also promotes weight loss and is involved in lipid metabolism. Once secreted, GLP-1 is quickly degraded by dipeptidyl peptidase-4 (DPP-4). Therefore, DPP-4 inhibitors are able to extend the activity of GLP-1. Currently, GLP-1 agonists and DPP-4 inhibitors represent attractive options for the treatment of NAFLD and NASH. The modulation of lipid and glucose metabolism through nuclear receptors, such as the farsenoid X receptor, also constitutes an attractive therapeutic target. Obeticholic acid is a potent activator of the farnesoid X nuclear receptor and reduces liver fat content and fibrosis in animal models. Ursodeoxycholic acid (UDCA) is a hydrophilic bile acid with immunomodulatory, anti-inflammatory, antiapoptotic, antioxidant and anti-fibrotic properties. UDCA can improve IR and modulate lipid metabolism through its interaction with nuclear receptors such as, TGR5, farnesoid X receptor-α, or the small heterodimeric partner. Finally, pharmacologic modulation of the gut microbiota could have a role in the therapy of NAFLD and NASH. Probiotics prevent bacterial translocation and epithelial invasion, inhibit mucosal adherence by bacteria, and stimulate host immunity. In animal models, probiotics prevent obesity, decrease transaminase levels, and improve IR and liver histology in NASH.

Key Words: Insulin resistance, Lipotoxicity, Non-alcoholic steatohepatitis, Physiopathology, Therapeutic targets

Core tip: Non-alcoholic fatty liver disease (NAFLD) is an important health care problem. The pathophysiology of NAFLD and non-alcoholic steatohepatitis (NASH) and their progression are multifactorial and complex processes, where multi-parallel simultaneous hits derived from the gut and adipose tissue promote a pro-inflammatory response and liver injury. All of these represent attractive therapeutic targets. Pharmacological agents such as glucagon-like peptide-1 agonists, dipeptidyl peptidase-4 inhibitors, ursodeoxycholic acid, obeticholic acid and probiotics need to be explored in clinical trials specifically for treating NAFLD and NASH.



INTRODUCTION

Currently, non-alcoholic fatty liver disease (NAFLD) is recognized as a major health care problem, and it is the most common cause of raised transaminases in western countries, where it is considered the first cause of liver disease. Worldwide its incidence and prevalence are increasing[1-4]. By 2020, non-alcoholic steatohepatitis (NASH) could be the main cause of liver transplantation[5].

NALFD is characterized by fat accumulation, mainly as triglycerides, in the hepatocytes; it is associated with clinical factors such as obesity, dyslipidemia, and diabetes; its diagnosis requires the exclusion of other conditions that could be associated with steatosis, such as, significant alcohol consumption, viral hepatitis, use of steatogenic medications or hereditary disorders[1,6].

Histologically, NAFLD is classified as non-alcoholic fatty liver (NAFL) or simple steatosis, and NASH. NASH is characterized by the presence of steatosis accompanied by inflammatory infiltrate, ballooning of hepatocytes, and the presence of Mallory-Denk bodies; any degree of fibrosis can be present, but this is not a mandatory finding. Patients with NASH, mainly those with advanced fibrosis, are at higher risk for developing decompensated cirrhosis, hepatocellular carcinoma[7-9], or even death due to cardiovascular disease as a result of early atherosclerosis[10].

NAFLD represents the hepatic expression of the metabolic syndrome, and its physiopathology involves several mechanisms, such as, glucose intolerance, insulin resistance (IR)[11,12], enhanced lipogenesis and lipotoxicity[12,13], hepatic and systemic inflammation[14], and oxidative stress[15].

The “multi-parallel hits” hypothesis is the most accepted for understanding the pathogenesis of NAFL and NASH and their progression to cirrhosis. This hypothesis proposes that many simultaneous hits derived from the gut and adipose tissue may promote inflammation and liver injury[13].

Unhealthy lifestyles are clearly related to NAFL and NASH. Excess energy intake through a diet rich in fat and carbohydrates[14-17] leads to failure of adipocytes to adapt in terms of proliferation and differentiation[18]. In the liver, free fatty acids (FFAs) are the main source for the synthesis of triglycerides. Similarly, excess dietary fat and de novo lipogenesis are two main factors contributing to the production of diacylglycerol and lysophosphatidyl choline, two non-triglyceride metabolites, which are responsible for lipotoxicity[19,20]. FFAs and cholesterol can also accumulate in the mitochondria leading to inflammation and liver injury mediated by tumor necrosis factor alpha and reactive oxygen species[13,21,22].

As NAFL and NASH are frequently associated with overweight, an important objective in the treatment of NAFL and NASH is to encourage weight loss; this can be achieved through lifestyle modification including a hypocaloric diet and/or aerobic exercise. The loss of at least 5% of body weight is necessary to improve steatosis, but a loss greater than 10% may be needed to improve steatohepatitis[23].

Pharmacological agents that could be useful in NAFL and NASH include glucagon-like peptide-1 (GLP-1) agonists. GLP-1 is an intestinal mucosa-derived hormone which is secreted into the bloodstream in response to nutrient ingestion; it favors glucose-stimulated insulin secretion, inhibition of postprandial glucagon secretion and delayed gastric emptying[24]. GLP-1 agonists also promote weight loss. In one study, treatment with liraglutide 1.2 mg once daily for 12 wk improved eating behavior in obese women with polycystic ovary syndrome (PCOS) and resulted in an average weight loss of 3.8 ± 0.1 kg (P < 0.001)[25]. In another study, short-term combined treatment with liraglutide 1.2 mg once daily and metformin 1000 mg twice daily was associated with significant weight loss and a decrease in waist circumference in obese women with PCOS who had previously been poor responders to metformin monotherapy[26]. In a cohort of obese nondiabetic women, short-term treatment with exenatide was also associated with a modest weight loss and decreased waist circumference[27].

GLP-1 is also involved in lipid metabolism; studies in animal models and in diabetic patients have found that GLP-1 agonists suppress postprandial elevations in lipids and lipoproteins[28-30]; result in a decrease in serum triglycerides, total cholesterol, low density lipoprotein-cholesterol and serum high density lipoprotein-cholesterol levels and reduce the development of atherosclerosis[31-34]. In mice, treatment with GLP-1 agonists was related to a reduction in the hepatic content of triglycerides[35]. Besides its property of enhancing insulin sensitivity, other possible mechanisms through which GLP-1 agonists may improve the lipid profile and metabolism are: Activation of peroxisome proliferator-activated receptor-α on the hepatic cell surface, which reduces the synthesis of apolipoprotein C, degrades fat in plasma, and removes triglycerides[36-39].

Once secreted, GLP-1 is quickly degraded by dipeptidyl peptidase-4 (DPP-4). DPP-4 inhibitors prolong the activity of incretins, GLP-1 and glucose-dependent insulinotropic polypeptide[24]. As the receptor for GLP-1 has been shown to exist in various cells, including hepatocytes[40,41], DPP-4 inhibitors may have pleiotropic effects independent of lowering plasma glucose level and stimulating insulin secretion. In a retrospective study which included diabetic patients who received treatment with DPP-4 inhibitors and with abnormal transaminase levels, Kanazawa et al[42] found that transaminase levels decreased after six months of treatment with DPP-4 inhibitors.

Ursodeoxycholic acid (UDCA) is not approved for treating NASH; however, it is a hydrophilic bile acid with immunomodulatory, anti-inflammatory, antiapoptotic, antioxidant and anti-fibrotic properties. It also reduces the mitochondrial membrane permeability and the release of hydrolytic enzymes from damaged hepatocytes. In a recent study which included patients with biopsy-proven NASH, high-dose UDCA for 12 mo reduced transaminase levels, the degree of fibrosis, and improved IR independently of the change in body weight. UDCA modulated lipid and glucose metabolism through its interaction with nuclear receptors such as, TGR5, farnesoid X receptor-α, or the small heterodimeric partner[43].

Obeticholic acid is a potent activator of the farnesoid X nuclear receptor that reduces liver fat content and fibrosis in animal models of NAFLD. In a multicenter, double-blind, placebo-controlled, randomized clinical trial, treatment with obeticholic acid in adult patients with NASH for 72 wk improved the histological features of NASH, but its long-term benefits and safety require further study[44].

Recently, it was demonstrated that the gut-liver axis plays a key role in the pathogenesis of obesity, NAFL, NASH and their progression. The gut microbiota is composed of bacteria, viruses, yeasts and parasites. Gut microbes are able to interact actively with the host immune system modulating inflammation, IR, intestinal permeability, and endotoxemia[45]. Pharmacologic modulation of the gut microbiota could have a role in the therapy of NAFL and NASH. Probiotics prevent bacterial translocation and epithelial invasion, inhibit mucosal adherence by bacteria, and stimulate host immunity. In animal models, probiotics also prevent obesity, improve IR and liver histology in NASH, and decrease transaminase levels[46-50]. In patients with NAFLD the use of probiotics improved transaminases, the cytokine profile and oxidative stress[51].

In summary, NAFLD is an important health care problem. The pathophysiology of NAFL and NASH and their progression involve multifactorial and complex processes, where multi-parallel simultaneous hits derived from the gut and adipose tissue promote a pro-inflammatory response and liver injury. All of these represent attractive therapeutic targets. Pharmacological agents such as GLP-1 agonists, DPP-4 inhibitors, UDCA, obeticholic acid and probiotics need to be explored in clinical trials specifically for treating NAFL and NASH.

Footnotes

P- Reviewer: Buechler C, Hwu CM, Manesis EK

S- Editor: Ji FF L- Editor: Webster JR E- Editor: Liu SQ

References
1.  Ong JP, Younossi ZM. Epidemiology and natural history of NAFLD and NASH. Clin Liver Dis. 2007;11:1-16, vii.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 316]  [Cited by in F6Publishing: 329]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
2.  LaBrecque DR, Abbas Z, Anania F, Ferenci P, Khan AG, Goh KL, Hamid SS, Isakov V, Lizarzabal M, Peñaranda MM. World Gastroenterology Organisation global guidelines: Nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. J Clin Gastroenterol. 2014;48:467-473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 245]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
3.  Bellentani S, Scaglioni F, Marino M, Bedogni G. Epidemiology of non-alcoholic fatty liver disease. Dig Dis. 2010;28:155-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 607]  [Cited by in F6Publishing: 625]  [Article Influence: 44.6]  [Reference Citation Analysis (0)]
4.  Lazo M, Clark JM. The epidemiology of nonalcoholic fatty liver disease: a global perspective. Semin Liver Dis. 2008;28:339-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 521]  [Cited by in F6Publishing: 503]  [Article Influence: 31.4]  [Reference Citation Analysis (0)]
5.  Mahady SE, George J. Management of nonalcoholic steatohepatitis: an evidence-based approach. Clin Liver Dis. 2012;16:631-645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 34]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
6.  Lomonaco R, Chen J, Cusi K. An Endocrine Perspective of Nonalcoholic Fatty Liver Disease (NAFLD). Ther Adv Endocrinol Metab. 2011;2:211-225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
7.  Paredes AH, Torres DM, Harrison SA. Nonalcoholic fatty liver disease. Clin Liver Dis. 2012;16:397-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
8.  Brunt EM, Kleiner DE, Wilson LA, Belt P, Neuschwander-Tetri BA. Nonalcoholic fatty liver disease (NAFLD) activity score and the histopathologic diagnosis in NAFLD: distinct clinicopathologic meanings. Hepatology. 2011;53:810-820.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 785]  [Cited by in F6Publishing: 843]  [Article Influence: 64.8]  [Reference Citation Analysis (0)]
9.  Angulo P, Hui JM, Marchesini G, Bugianesi E, George J, Farrell GC, Enders F, Saksena S, Burt AD, Bida JP. The NAFLD fibrosis score: a noninvasive system that identifies liver fibrosis in patients with NAFLD. Hepatology. 2007;45:846-854.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1917]  [Cited by in F6Publishing: 2018]  [Article Influence: 118.7]  [Reference Citation Analysis (1)]
10.  Gastaldelli A, Kozakova M, Højlund K, Flyvbjerg A, Favuzzi A, Mitrakou A, Balkau B. Fatty liver is associated with insulin resistance, risk of coronary heart disease, and early atherosclerosis in a large European population. Hepatology. 2009;49:1537-1544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 258]  [Cited by in F6Publishing: 258]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
11.  Chitturi S, Abeygunasekera S, Farrell GC, Holmes-Walker J, Hui JM, Fung C, Karim R, Lin R, Samarasinghe D, Liddle C. NASH and insulin resistance: Insulin hypersecretion and specific association with the insulin resistance syndrome. Hepatology. 2002;35:373-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 807]  [Cited by in F6Publishing: 803]  [Article Influence: 36.5]  [Reference Citation Analysis (0)]
12.  Cusi K. Role of insulin resistance and lipotoxicity in non-alcoholic steatohepatitis. Clin Liver Dis. 2009;13:545-563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 160]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
13.  Tilg H, Moschen AR. Evolution of inflammation in nonalcoholic fatty liver disease: the multiple parallel hits hypothesis. Hepatology. 2010;52:1836-1846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1543]  [Cited by in F6Publishing: 1629]  [Article Influence: 116.4]  [Reference Citation Analysis (0)]
14.  Jeffery RW, Harnack LJ. Evidence implicating eating as a primary driver for the obesity epidemic. Diabetes. 2007;56:2673-2676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 56]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
15.  Ishimoto T, Lanaspa MA, Rivard CJ, Roncal-Jimenez CA, Orlicky DJ, Cicerchi C, McMahan RH, Abdelmalek MF, Rosen HR, Jackman MR. High-fat and high-sucrose (western) diet induces steatohepatitis that is dependent on fructokinase. Hepatology. 2013;58:1632-1643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 220]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
16.  Panchal SK, Poudyal H, Iyer A, Nazer R, Alam MA, Diwan V, Kauter K, Sernia C, Campbell F, Ward L. High-carbohydrate, high-fat diet-induced metabolic syndrome and cardiovascular remodeling in rats. J Cardiovasc Pharmacol. 2011;57:611-624.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 103]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
17.  Zhou X, Han D, Xu R, Li S, Wu H, Qu C, Wang F, Wang X, Zhao Y. A model of metabolic syndrome and related diseases with intestinal endotoxemia in rats fed a high fat and high sucrose diet. PLoS One. 2014;9:e115148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 65]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
18.  Gregor MF, Hotamisligil GS. Thematic review series: Adipocyte Biology. Adipocyte stress: the endoplasmic reticulum and metabolic disease. J Lipid Res. 2007;48:1905-1914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 409]  [Cited by in F6Publishing: 409]  [Article Influence: 24.1]  [Reference Citation Analysis (0)]
19.  Neuschwander-Tetri BA. Hepatic lipotoxicity and the pathogenesis of nonalcoholic steatohepatitis: the central role of nontriglyceride fatty acid metabolites. Hepatology. 2010;52:774-788.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 726]  [Cited by in F6Publishing: 764]  [Article Influence: 54.6]  [Reference Citation Analysis (0)]
20.  Postic C, Girard J. Contribution of de novo fatty acid synthesis to hepatic steatosis and insulin resistance: lessons from genetically engineered mice. J Clin Invest. 2008;118:829-838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 838]  [Cited by in F6Publishing: 877]  [Article Influence: 54.8]  [Reference Citation Analysis (0)]
21.  Feldstein AE, Werneburg NW, Canbay A, Guicciardi ME, Bronk SF, Rydzewski R, Burgart LJ, Gores GJ. Free fatty acids promote hepatic lipotoxicity by stimulating TNF-alpha expression via a lysosomal pathway. Hepatology. 2004;40:185-194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 586]  [Cited by in F6Publishing: 577]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
22.  Marí M, Caballero F, Colell A, Morales A, Caballeria J, Fernandez A, Enrich C, Fernandez-Checa JC, García-Ruiz C. Mitochondrial free cholesterol loading sensitizes to TNF- and Fas-mediated steatohepatitis. Cell Metab. 2006;4:185-198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 477]  [Cited by in F6Publishing: 466]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
23.  Chalasani N, Younossi Z, Lavine JE, Diehl AM, Brunt EM, Cusi K, Charlton M, Sanyal AJ. 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. 2012;55:2005-2023.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2413]  [Cited by in F6Publishing: 2453]  [Article Influence: 204.4]  [Reference Citation Analysis (0)]
24.  Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet. 2006;368:1696-1705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2787]  [Cited by in F6Publishing: 2681]  [Article Influence: 148.9]  [Reference Citation Analysis (1)]
25.  Jensterle M, Kocjan T, Kravos NA, Pfeifer M, Janez A. Short-term intervention with liraglutide improved eating behavior in obese women with polycystic ovary syndrome. Endocr Res. 2014;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
26.  Jensterle Sever M, Kocjan T, Pfeifer M, Kravos NA, Janez A. Short-term combined treatment with liraglutide and metformin leads to significant weight loss in obese women with polycystic ovary syndrome and previous poor response to metformin. Eur J Endocrinol. 2014;170:451-459.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 95]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
27.  Dushay J, Gao C, Gopalakrishnan GS, Crawley M, Mitten EK, Wilker E, Mullington J, Maratos-Flier E. Short-term exenatide treatment leads to significant weight loss in a subset of obese women without diabetes. Diabetes Care. 2012;35:4-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 90]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
28.  Bradley DP, Kulstad R, Racine N, Shenker Y, Meredith M, Schoeller DA. Alterations in energy balance following exenatide administration. Appl Physiol Nutr Metab. 2012;37:893-899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
29.  Hsieh J, Longuet C, Baker CL, Qin B, Federico LM, Drucker DJ, Adeli K. The glucagon-like peptide 1 receptor is essential for postprandial lipoprotein synthesis and secretion in hamsters and mice. Diabetologia. 2010;53:552-561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 173]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
30.  Schwartz EA, Koska J, Mullin MP, Syoufi I, Schwenke DC, Reaven PD. Exenatide suppresses postprandial elevations in lipids and lipoproteins in individuals with impaired glucose tolerance and recent onset type 2 diabetes mellitus. Atherosclerosis. 2010;212:217-222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 139]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
31.  Arakawa M, Mita T, Azuma K, Ebato C, Goto H, Nomiyama T, Fujitani Y, Hirose T, Kawamori R, Watada H. Inhibition of monocyte adhesion to endothelial cells and attenuation of atherosclerotic lesion by a glucagon-like peptide-1 receptor agonist, exendin-4. Diabetes. 2010;59:1030-1037.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 382]  [Cited by in F6Publishing: 395]  [Article Influence: 28.2]  [Reference Citation Analysis (0)]
32.  Blonde L, Klein EJ, Han J, Zhang B, Mac SM, Poon TH, Taylor KL, Trautmann ME, Kim DD, Kendall DM. Interim analysis of the effects of exenatide treatment on A1C, weight and cardiovascular risk factors over 82 weeks in 314 overweight patients with type 2 diabetes. Diabetes Obes Metab. 2006;8:436-447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 314]  [Cited by in F6Publishing: 329]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
33.  Nyström T, Gutniak MK, Zhang Q, Zhang F, Holst JJ, Ahrén B, Sjöholm A. Effects of glucagon-like peptide-1 on endothelial function in type 2 diabetes patients with stable coronary artery disease. Am J Physiol Endocrinol Metab. 2004;287:E1209-E1215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 488]  [Cited by in F6Publishing: 486]  [Article Influence: 24.3]  [Reference Citation Analysis (0)]
34.  Klonoff DC, Buse JB, Nielsen LL, Guan X, Bowlus CL, Holcombe JH, Wintle ME, Maggs DG. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin. 2008;24:275-286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 361]  [Cited by in F6Publishing: 396]  [Article Influence: 24.8]  [Reference Citation Analysis (0)]
35.  Samson SL, Sathyanarayana P, Jogi M, Gonzalez EV, Gutierrez A, Krishnamurthy R, Muthupillai R, Chan L, Bajaj M. Exenatide decreases hepatic fibroblast growth factor 21 resistance in non-alcoholic fatty liver disease in a mouse model of obesity and in a randomised controlled trial. Diabetologia. 2011;54:3093-3100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 84]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
36.  Wilkins JJ, Dubar M, Sébastien B, Laveille C. A drug and disease model for lixisenatide, a GLP-1 receptor agonist in type 2 diabetes. J Clin Pharmacol. 2014;54:267-278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
37.  Sebokova E, Bénardeau A, Sprecher U, Sewing S, Tobalina L, Migliorini C. Taspoglutide, a novel human once-weekly analogue of glucagon-like peptide-1, improves glucose homeostasis and body weight in the Zucker diabetic fatty rat. Diabetes Obes Metab. 2010;12:674-682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
38.  Aguilar RB. Evaluating treatment algorithms for the management of patients with type 2 diabetes mellitus: a perspective on the definition of treatment success. Clin Ther. 2011;33:408-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
39.  Wang XC, Gusdon AM, Liu H, Qu S. Effects of glucagon-like peptide-1 receptor agonists on non-alcoholic fatty liver disease and inflammation. World J Gastroenterol. 2014;20:14821-14830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 61]  [Cited by in F6Publishing: 72]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
40.  Gupta NA, Mells J, Dunham RM, Grakoui A, Handy J, Saxena NK, Anania FA. Glucagon-like peptide-1 receptor is present on human hepatocytes and has a direct role in decreasing hepatic steatosis in vitro by modulating elements of the insulin signaling pathway. Hepatology. 2010;51:1584-1592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 332]  [Cited by in F6Publishing: 355]  [Article Influence: 25.4]  [Reference Citation Analysis (0)]
41.  Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007;132:2131-2157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2461]  [Cited by in F6Publishing: 2465]  [Article Influence: 145.0]  [Reference Citation Analysis (0)]
42.  Kanazawa I, Tanaka K, Sugimoto T. DPP-4 inhibitors improve liver dysfunction in type 2 diabetes mellitus. Med Sci Monit. 2014;20:1662-1667.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 22]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
43.  Ratziu V, de Ledinghen V, Oberti F, Mathurin P, Wartelle-Bladou C, Renou C, Sogni P, Maynard M, Larrey D, Serfaty L. A randomized controlled trial of high-dose ursodesoxycholic acid for nonalcoholic steatohepatitis. J Hepatol. 2011;54:1011-1019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 211]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
44.  Neuschwander-Tetri BA, Loomba R, Sanyal AJ, Lavine JE, Van Natta ML, Abdelmalek MF, Chalasani N, Dasarathy S, Diehl AM, Hameed B, Kowdley KV, McCullough A, Terrault N, Clark JM, Tonascia J, Brunt EM, Kleiner DE, Doo E; NASH Clinical Research Network. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trial. Lancet. 2015;385:956-965.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1569]  [Cited by in F6Publishing: 1606]  [Article Influence: 178.4]  [Reference Citation Analysis (0)]
45.  Duseja A, Chawla YK. Obesity and NAFLD: the role of bacteria and microbiota. Clin Liver Dis. 2014;18:59-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 70]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
46.  Nardone G, Rocco A. Probiotics: a potential target for the prevention and treatment of steatohepatitis. J Clin Gastroenterol. 2004;38:S121-S122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
47.  Naito E, Yoshida Y, Makino K, Kounoshi Y, Kunihiro S, Takahashi R, Matsuzaki T, Miyazaki K, Ishikawa F. Beneficial effect of oral administration of Lactobacillus casei strain Shirota on insulin resistance in diet-induced obesity mice. J Appl Microbiol. 2011;110:650-657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 153]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
48.  Li Z, Yang S, Lin H, Huang J, Watkins PA, Moser AB, Desimone C, Song XY, Diehl AM. Probiotics and antibodies to TNF inhibit inflammatory activity and improve nonalcoholic fatty liver disease. Hepatology. 2003;37:343-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 658]  [Cited by in F6Publishing: 662]  [Article Influence: 31.5]  [Reference Citation Analysis (0)]
49.  Wagnerberger S, Spruss A, Kanuri G, Stahl C, Schröder M, Vetter W, Bischoff SC, Bergheim I. Lactobacillus casei Shirota protects from fructose-induced liver steatosis: a mouse model. J Nutr Biochem. 2013;24:531-538.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 86]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
50.  Velayudham A, Dolganiuc A, Ellis M, Petrasek J, Kodys K, Mandrekar P, Szabo G. VSL#3 probiotic treatment attenuates fibrosis without changes in steatohepatitis in a diet-induced nonalcoholic steatohepatitis model in mice. Hepatology. 2009;49:989-997.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 187]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
51.  Loguercio C, Federico A, Tuccillo C, Terracciano F, D’Auria MV, De Simone C, Del Vecchio Blanco C. Beneficial effects of a probiotic VSL#3 on parameters of liver dysfunction in chronic liver diseases. J Clin Gastroenterol. 2005;39:540-543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 316]  [Cited by in F6Publishing: 320]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]