Non-Alcoholic Fatty Liver Disease – A Brief Insight into … · 2015. 6. 20. · Non-alcoholic...

7
Central Journal of Liver and Clinical Research Cite this article: Suyavaran A, Mareeswaran R, Ramamurthy C, Subastri A, Rao PL, et al. (2015) Non-Alcoholic Fatty Liver Disease – A Brief Insight into Pathogenesis and Review of Recent Reports on Therapeutic Targets J Liver Clin Res 2(2): 1014. *Corresponding author C. Thirunavukkarasu, Visiting scientist, Department of Medicine – Gastroenterology and Liver disease, Albert Einstein College of medicine of Yeshiva University, Bronx, New York, USA-10461, Tel: 011-347-471-6477; Email: Submitted: 15 March 2015 Accepted: 13 June 2015 Published: 15 June 2015 Copyright © 2015 Thirunavukkarasu et al. OPEN ACCESS Keywords NAFLD NASH • Inflammation Steatosis Fibrosis Review Article Non-Alcoholic Fatty Liver Disease – A Brief Insight into Pathogenesis and Review of Recent Reports on Therapeutic Targets Arumugam Suyavaran 1 , Ramachandran Mareeswaran 1 , Chitteti Ramamurthy 1 , Ariraman Subastri 1 , Polaki Lokeswara Rao 1 , Preedia Babu 1 , Chinnasamy Thirunavukkarasu 1,2 * 1 Department of Biochemistry and Molecular Biology, Pondicherry University, India 2 Department of Medicine – Gastroenterology and Liver disease, Albert Einstein College of medicine of Yeshiva University, USA Abstract Non-alcoholic fatty liver disease (NAFLD) is a complication of global prevalence occurring due to defective regulation of hepatic lipid metabolism. Currently, NAFLD is being viewed as an emerging epidemic by virtue of increase in obesity cases. Accumulation of excess fatty acid in the liver, leads to activation of an array of inflammatory signals, resulting in hepatic steatosis. Inadequately treated steatosis would progress to non-alcoholic steatohepatitis (NASH) marked by severe hepatic inflammation and fibrosis. In the light of rising NAFLD cases and lack of a validated therapy, formulation of novel and efficient treatment strategies are in high demand. Several factors including inflammatory cytokines, chemokines and receptors are involved in execution of the inflammatory process in NASH progression. A clarified comprehension related to the mode of action and interplay between these factors is indispensible for an efficient therapeutic approach for NAFLD. This review is focused on the key factors involved in NAFLD progression and their potential as therapeutic targets. ABBREVIATIONS NAFLD: Non-Alcoholic Fatty Liver Disease; NASH: Non- Alcoholic Steatohepatitis; VLDL: Very Low Density Lipoproteins; TNF-Α: Tumor Necrosis Factor-Α; JNK1: C-Jun N-Terminal Kinase 1; TGF-Β: Transforming Growth Factor-Β; Iκb: Inhibitory Factor Kappa B; Ikkβ: Iκb Kinase Β ; NF-Κb: Nuclear Factor Kappa B ; MAPK: Mitogen Activated Protein Kinase; EGCG: Epigallocatechin Gallate; MCD: Methionine and Choline Deficient; IL: Interleukin; CRP: C- Reactive Protein; CT: Computerized Tomography; FXR: Farnesoid X Receptor; CCL/CXCL: Chemokine Ligand; CCR/CXCR: Chemokine Receptor; PPAR: Peroxisome Proliferator Activated Receptor; KLF: Kruppel Like Factor 6; HDL: High Density Lipoprotein INTRODUCTION Globally there has been an upsurge in health complications related to hepatic disorders by virtue of high fat diet consumption of the general population and predisposing genetic factors. Non-alcoholic fatty liver disease is one of the leading causes of chronic liver diseases affecting one third of the population in developed countries [1]. Non-alcoholic fatty liver disease (NAFLD) is a term blanketing wide spectrum of observed clinical hepatic complications ranging from mild steatosis to fibrotic and cirrhotic liver [1,2]. It is characterized by accumulation of excess fat (>5%) in the hepatocytes in the form of triglycerides, leading to detectable enlargement of liver and altered metabolic symptoms [3]. NAFLD is primarily associated with impaired lipid metabolism attributed to genetic and physiological causes, the exact origin of which is yet to be comprehended completely [4,5]. The ‘two hit’- theory is considered as the most acceptable version of NAFLD pathogenesis. The first hit is due to an imbalance between uptake of free fatty acids, de novo – lipogenesis and their usage through β-oxidation and secretion into circulation by very low density

Transcript of Non-Alcoholic Fatty Liver Disease – A Brief Insight into … · 2015. 6. 20. · Non-alcoholic...

Page 1: Non-Alcoholic Fatty Liver Disease – A Brief Insight into … · 2015. 6. 20. · Non-alcoholic fatty liver disease is one of the leading causes of chronic liver diseases affecting

Central Journal of Liver and Clinical Research

Cite this article: Suyavaran A, Mareeswaran R, Ramamurthy C, Subastri A, Rao PL, et al. (2015) Non-Alcoholic Fatty Liver Disease – A Brief Insight into Pathogenesis and Review of Recent Reports on Therapeutic Targets J Liver Clin Res 2(2): 1014.

*Corresponding authorC. Thirunavukkarasu, Visiting scientist, Department of Medicine – Gastroenterology and Liver disease, Albert Einstein College of medicine of Yeshiva University, Bronx, New York, USA-10461, Tel: 011-347-471-6477; Email:

Submitted: 15 March 2015

Accepted: 13 June 2015

Published: 15 June 2015

Copyright© 2015 Thirunavukkarasu et al.

OPEN ACCESS

Keywords•NAFLD•NASH•Inflammation•Steatosis•Fibrosis

Review Article

Non-Alcoholic Fatty Liver Disease – A Brief Insight into Pathogenesis and Review of Recent Reports on Therapeutic TargetsArumugam Suyavaran1, Ramachandran Mareeswaran1, Chitteti Ramamurthy1, Ariraman Subastri1, Polaki Lokeswara Rao1, Preedia Babu1, Chinnasamy Thirunavukkarasu1,2* 1Department of Biochemistry and Molecular Biology, Pondicherry University, India2Department of Medicine – Gastroenterology and Liver disease, Albert Einstein College of medicine of Yeshiva University, USA

Abstract

Non-alcoholic fatty liver disease (NAFLD) is a complication of global prevalence occurring due to defective regulation of hepatic lipid metabolism. Currently, NAFLD is being viewed as an emerging epidemic by virtue of increase in obesity cases. Accumulation of excess fatty acid in the liver, leads to activation of an array of inflammatory signals, resulting in hepatic steatosis. Inadequately treated steatosis would progress to non-alcoholic steatohepatitis (NASH) marked by severe hepatic inflammation and fibrosis. In the light of rising NAFLD cases and lack of a validated therapy, formulation of novel and efficient treatment strategies are in high demand. Several factors including inflammatory cytokines, chemokines and receptors are involved in execution of the inflammatory process in NASH progression. A clarified comprehension related to the mode of action and interplay between these factors is indispensible for an efficient therapeutic approach for NAFLD. This review is focused on the key factors involved in NAFLD progression and their potential as therapeutic targets.

ABBREVIATIONSNAFLD: Non-Alcoholic Fatty Liver Disease; NASH: Non-

Alcoholic Steatohepatitis; VLDL: Very Low Density Lipoproteins; TNF-Α: Tumor Necrosis Factor-Α; JNK1: C-Jun N-Terminal Kinase 1; TGF-Β: Transforming Growth Factor-Β; Iκb: Inhibitory Factor Kappa B; Ikkβ: Iκb Kinase Β ; NF-Κb: Nuclear Factor Kappa B ; MAPK: Mitogen Activated Protein Kinase; EGCG: Epigallocatechin Gallate; MCD: Methionine and Choline Deficient; IL: Interleukin; CRP: C- Reactive Protein; CT: Computerized Tomography; FXR: Farnesoid X Receptor; CCL/CXCL: Chemokine Ligand; CCR/CXCR: Chemokine Receptor; PPAR: Peroxisome Proliferator Activated Receptor; KLF: Kruppel Like Factor 6; HDL: High Density Lipoprotein

INTRODUCTIONGlobally there has been an upsurge in health complications

related to hepatic disorders by virtue of high fat diet consumption

of the general population and predisposing genetic factors. Non-alcoholic fatty liver disease is one of the leading causes of chronic liver diseases affecting one third of the population in developed countries [1]. Non-alcoholic fatty liver disease (NAFLD) is a term blanketing wide spectrum of observed clinical hepatic complications ranging from mild steatosis to fibrotic and cirrhotic liver [1,2]. It is characterized by accumulation of excess fat (>5%) in the hepatocytes in the form of triglycerides, leading to detectable enlargement of liver and altered metabolic symptoms [3].

NAFLD is primarily associated with impaired lipid metabolism attributed to genetic and physiological causes, the exact origin of which is yet to be comprehended completely [4,5]. The ‘two hit’- theory is considered as the most acceptable version of NAFLD pathogenesis. The first hit is due to an imbalance between uptake of free fatty acids, de novo – lipogenesis and their usage through β-oxidation and secretion into circulation by very low density

Page 2: Non-Alcoholic Fatty Liver Disease – A Brief Insight into … · 2015. 6. 20. · Non-alcoholic fatty liver disease is one of the leading causes of chronic liver diseases affecting

Central

Thirunavukkarasu et al. (2015)Email:

J Liver Clin Res 2(2): 1014 (2015) 2/7

lipoproteins (VLDL) which results in excess lipid accumulation in liver. The second hit is the consequence of excess hepatic lipid accumulation which leads to oxidative stress which further triggers the release of inflammatory cytokines, adipokines and mitochondrial dysfunction leading to steatohepatitis [6,7]. The changes in hepatocytes are observed as a spectrum ranging from simple lipid accumulation in vacuoles to aggressive fibrotic lesions [8].

The transition from NAFLD to non-alcoholic steatohepatitis (NASH) is the major hallmark in the progressive pathogenesis [8, 9]. The inflammatory signals in liver lead to activation of hepatic stellate cells, which leads to secretion of extracellular matrix resulting in fibrotic lesions [10]. Studies show that approximately 29% of patients suffering from NASH end up with fibrosis in 10 years [11]. The physiological steatosis stage resulting due to lipid deposition in hepatocytes is highly reversible. However, the continued stress to hepatocytes due to diminished efflux of triglycerides (TGs) triggers the oxidative stress [12]. Lipid peroxidation of liver cells is considered as a significant contributor to initiation of inflammatory signals [13].

Pathophysiological factors such as insulin resistance, obesity and genetic predisposition aid in progression of NASH [14]. Several studies, have reported the role of inflammatory mediators viz., cytokines, chemokines, adipokines and transcriptional factors [15, 16]. However, an overall understanding of these signaling cascades and molecular players is essential for better interpretation and designing of future therapy for NAFLD. This review would focus on some of the key players in the inflammatory signaling and therapeutic targets involved in NAFLD.

MOLECULAR TARGETS

Cytokines

Cytokines are messengers secreted by various cell types in the body in response to a wide variety of physiological stimulus [17]. Cytokines aid in normal physiological processes such as growth, differentiation, hematopoiesis, as well as several inflammatory and immune responses [18]. Their involvement in execution of inflammatory reactions in several pathologies such as cardiovascular disease, rheumatoid arthritis, NAFLD, etc., have been reported earlier [19,20]. The cytokines as present in minimal levels in hepatic circulation during normal physiological status and are necessary for hepatic homeostasis [17]. However, the cytokines have been observed to play an active role in mediating the inflammatory progression of NAFLD as characterized by apoptotic and necrotic lesion in liver leading to fibrosis [15]. The cytokines involved in hepatic inflammation, are categorized under several subfamilies – Tumor necrosis factor-α (TNF-α), Tumor growth factor β (TGF-β), Interleukins and chemokines.

Tumor Necrosis Factor α

TNF-α is a pro-inflammatory cytokine secreted by several cell types such as neutrophils, macrophages, T and B lymphocytes, endothelial cells, mast cells, fibroblasts etc., In liver, hepatocytes and Kupffer cells are the major contributors of TNF- α [21]. TNF-α plays a central role in the initiation of inflammatory cascade and its progression from steatosis to steatohepatitis [22]. Experiments with mice models for obesity have shown the

importance of TNF-α in NAFLD, where anti- TNF-α –drug therapy showed promising results [23, 24]. Increased free fatty acid level in obesity stimulates the hepatocytes to secrete TNF-α, which elicits the free fatty acid induced expression of inflammatory genes [23]. A positive correlation has been observed between TNF-α level in serum and degree of fibrosis in patients with NAFLD [25]. A study with pentoxifylline - an inhibitor of TNF-α has shown suppressive effect on elevation of serum transaminases and triglycerides in experimental NAFLD induced rats. The study also demonstrated that NAFLD induced TNF-α expression stimulates endoplasmic reticulum stress, which further mediate the progression of steatosis to fibrosis [26].

c-Jun N-terminal Kinase 1 (JNK1) a stress activated protein kinase is activated by TNF-α which leads to initiation of an autocrine/paracrine loop resulting in enhanced TNF- α production in liver . TNF-α activates inhibitory kappa b kinase β (IKKβ) which phosphorylates IKB resulting in the translocation of NF-κB into nucleus. TNF-α - NF-κB axis mediated proinflammatory cytokine activation has been implemented in the pathogenesis of NAFLD [27]. Study with NF-κB and mitogen activated protein kinase (MAPKs) inhibition has shown to suppress TNF-α secretion, supporting the establishment of TNF-α - NF-κB loop for inflammatory mediation [28].

Transforming growth factor β

TGF-β is a cytokine secreted by hepatocytes and Kupffer cells in response to degradation changes in liver. TGF-β activates the resting stellate cells by transforming them into active myofibroblasts, which secrete extracellular matrix protein to initiate the fibrosis process [29]. Earlier studies have shown up regulated expression of TGF-β following experimentally induced hepatic damage such as in CCl4 poisoning [30, 31]. Elevated TGF-β mRNA expression levels have been found in patients with liver fibrosis. Earlier studies have shown that TGF-β is an early marker for the progression of steatohepatitis [32]. Hence, detection of TGF-β level would be helpful in marking NASH stage of NAFLD. Studies have observed polymorphisms in TGF-β1 gene in obese NAFLD patients with advanced stages of hepatic fibrosis [33]. Xiao and Ho have recently reported that administration of Epigallocatechin gallate (EGCG) reduced hepatic severity in NAFLD by suppression of TGF/SMAD pathway [34]. Studies with TGF-β receptor II deficient mice showed protective effect against experimentally induced NAFLD with methionine and choline deficient (MCD) diet, which was mediated through smad2 activation [35].

Interleukins

Interleukin-6 is a proinflammatory cytokine which has been implicated in metabolic syndrome [36]. IL-6 also plays several other functions such as inducer of immune response, hematopoiesis and oncogenesis [37]. Certain studies have reported IL-6 to be hepatoprotective and mediate hepatic regeneration after partial hepatectomy in mice [38, 39]. IL-6 has also been considered to reduce hepatic oxidative stress and to curtail mitochondrial dysfunction. However, studies have reported the role of IL-6 as an acute phase inflammatory mediator leading to secretion of several inflammatory serum proteins [40]. Hence, the possibility of its role in pathogenesis of NAFLD cannot

Page 3: Non-Alcoholic Fatty Liver Disease – A Brief Insight into … · 2015. 6. 20. · Non-alcoholic fatty liver disease is one of the leading causes of chronic liver diseases affecting

Central

Thirunavukkarasu et al. (2015)Email:

J Liver Clin Res 2(2): 1014 (2015) 3/7

be ignored. A positive correlation has been found in patients with NASH and circulating IL-6 level [41]. Studies with IL-6 knockout mice models have shown reduced severity when subjected to experimental NAFLD [42]. Yamaguchi et al, has shown that inhibition of IL-6 receptor with Tocilizumab, enhanced hepatic steatosis but protected against extensive hepatic damage in MCD diet induced NASH [43]. Recently, Hamirani et al, have observed a positive correlation between C – reactive protein (CRP) and IL-6 levels and increased liver fat accumulation in NAFLD patients as verified with CT-scans [44].

Interleukin -10 is considered as an anti-inflammatory cytokine with regulatory role over inflammation and pathophysiology of liver and other organs [19]. IL-10 is produced by hepatocytes, Kupffer cells and stellate cells in liver. But the exact role of IL-10 is yet to be defined. IL-10 has been shown to inhibit cell mediated inflammatory changes in liver [19]. Mc Mahan et al, found that INT-767 an agonist of farnesoid X receptor (FXR) increases IL-10 expression, which promotes the alternative activation of macrophage population. These macrophage population activated by IL-10 are found to be anti-inflammatory and hepatoprotective [45].

Chemokines

Chemokines are cytokines which act as chemo-attractants for leukocyte trafficking in the inflammatory sites [46]. Chemokines are classified into four subfamilies (C, CC, CXC and CX3C) according to the arrangement of the N-terminal conserved

cysteine residues. They are secreted by variety of cell types such as hepatocytes, stellate cells, endothelial cells and smooth muscle cells [47]. Chemokines execute their action by binding to the G-protein coupled receptors on the target cells. Studies have shown up regulated expression of chemokines and chemokines receptors in liver of patients with advanced stages of NAFLD portraying severe steatosis [48].

CCL2 is a crucial chemokine secreted by macrophages, endothelial cells and hepatic stellate cells in response to inflammatory stimulus. The elevation in free fatty acid level stimulates NF-κB activation induced secretion of inflammatory cytokines including TNF-α and CCL2 [49, 50]. It executes the target cell activation by binding to its receptor CCR2 [51]. Inhibition of CCL2/CCR2 pathway in mice has shown improvement of hepatic steatosis and adipocytes inflammation [52, 53]. Obstfeld et al, elucidated the role of CCL2 in progression of hepatic steatosis. They found that CCL2 expression in obesity activates and recruits a population of myeloid cells which leads to steatohepatitis [54]. In a recent study, Cynis et al have observed that the inhibition of glutaminyl cyclases - the enzymes necessary for maturation of cytokines to active form, improved the hepatic steatosis condition in experimental NAFLD [55].

CCL5 is associated with chronic inflammatory conditions and its relation with NAFLD has recently been described [56]. CCL5 are mainly secreted by hepatocytes and their release is mediated by excess lipid deposition in liver [57]. CCL5 is

Figure 1 Schematic representation of inflammatory pathway signals. The figure illustrates the activation of key molecular players and the cross talk between different pathways involved in progression of NAFLD to NASH. The green arrows indicate activation and red arrows indicate inhibition/suppression.

Page 4: Non-Alcoholic Fatty Liver Disease – A Brief Insight into … · 2015. 6. 20. · Non-alcoholic fatty liver disease is one of the leading causes of chronic liver diseases affecting

Central

Thirunavukkarasu et al. (2015)Email:

J Liver Clin Res 2(2): 1014 (2015) 4/7

essential for progression of hepatic steatosis to fibrosis and the studies blocking CCR5 have shown to be efficient in reducing the severity of NAFLD [58]. Certain studies have reported association of increases CXCL8 levels in serum with NASH [59, 60]. Similarly, CXCL9 and CXCL10 interact with a common receptor CXCR3 [61]. In NASH their levels are higher in liver and aid the migration of inflammatory cell types into hepatic parenchyma, thus promoting hepatic fibrosis [62, 63]. Inhibition of CXCL9 –CXCR3 has proven to be an efficient therapeutic target [64].

Peroxisome proliferator activated receptors (PPARs)

PPARs belong to nucleic acid receptor superfamily which consists of PPARα, PPARβ/δ and PPARɣ - expressed in mammals [65]. PPARα is synthesized extensively in metabolically active sites such as liver, intestine, muscle and brown adipose tissue. It mainly controls the transport of fatty acids and their β-oxidation, thereby depleting the hepatic lipid storage [66, 67]. PPARα activation inhibits NF-κB translocation mediated inflammatory cascade and release of C-reactive protein [68]. Previous experiments with PPARα agonists in MCD diet fed mice have proven beneficial in prevention of steatosis progression [69, 70]. PPARα agonism has also been reported to revert fibrosis by suppression of inflammatory signals and reduced expression of fibrotic marker. PPARα -/- mice have demonstrated increased susceptibility to NASH [71]. Bechmann et al, have reported the role of Kruppel like factor 6 (KLF6) a transcription factor which elicits pathology of NAFLD through PPARα post-transcriptional activation. KLF6 deficient mice were protected from high fat diet induced hepatic damage compared to normal mice [72]. PPARα activation has a direct anti-inflammatory role irrespective of hepatic triglyceride levels [73].

PPARɣ is expressed in adipocytes where it regulates fatty acids uptake and increases insulin sensitivity [74]. Increased expression of PPARɣ has been found in steatotic liver, as elucidated by previous studies due to its activation of de novo lipogenesis [75]. Studies with PPARɣ deletion in mice have shown to protect liver against steatosis, suggesting that PPARɣ has a prosteatotic role [76]. Contrastingly, other studies have elucidated a protective effect of PPARɣ against NAFLD [77]. Adenovirus mediated overexpression of PPARɣ has shown to protect liver against steatosis and fibrosis [78]. PPARɣ expression has shown to possess anti-fibrotic activity by suppression of stellate cell proliferation [79]. Though the role of PPARβ/δ in NAFLD remains unclear, it has been implicated as a metabolic regulator with roles in fatty acid transport and glucose homeostasis [80]. Studies have exemplified its role in attenuation of macrophage inflammatory response and elevation of plasma HDL concentrations [81]. Treatment with PPARβ/δ agonist GW501516 improves the hepatic condition by suppression of steatosis and inflammatory signals by regulation of lipid metabolism in MCD diet fed mice [82]. CCL4 toxicity in liver triggers PPARβ/δ expression, attributing to the inducible hepatic expression of PPARβ/δ as a protective response [83]. Recently, Staels et al, have reported beneficial role of PPARβ/δ agonist GFT505 in western diet fed hApoE2 knock-in/PPAR-α knockout mice. GFT505 demonstrated protective effect on liver irrespective of high fat diet feeding or exposure to CCL4 [69].

CONCLUSIONThe progression of steatosis to steatohepatitis in NAFLD is

an intricate process organized by complex signaling cascades of metabolic and inflammatory factors. Current review provides an overview of crucial factors involved in mediating the inflammatory progression and also an update on the inhibitor/agonist studies targeting specific factors to achieve suppression/inhibition of steatosis progression. Though several studies have reported the beneficial effects of inhibitors/agonists of specific factors on prevention or suppression of NAFLD, further understanding of the relay of inflammatory signals and molecular crosstalk is essential to devise flawless therapeutic designs. Moreover, the adverse effects of such therapeutic agents shouldn’t be ignored to prevent complications arising post therapy. Future studies exploring multiple inflammatory targets are necessary for improved combinatorial therapies for better management of NAFLD.

ACKNOWLEDGEMENTSThe authors duly acknowledge the funding support from

Department of Biochemistry and Molecular Biology, Pondicherry University, India, in the form of DST-FIST, Government of India. Author R. Mareeswaran, acknowledges Department of Science and Technology (NO.SR/FT/LS-63/2011 & DST-FIST), New Delhi, India for funding support. Author A. Suyavaran acknowledges UGC, New Delhi, India, for financial support in the form of Junior Research Fellowship [CSIR-UGC-JRF; S. no. F.17-115/98 (SA-I)].

REFERENCES1. Sheedfar F, Di Biase S, Koonen D, Vinciguerra M. Liver diseases and

aging: friends or foes? Aging Cell. 2013; 12: 950-954.

2. Farrell GC, Hall P, George J, McCullough AJ. Fatty Liver disease: NASH and related disorders, Blackwell, Malden, Mass, USA. 2005.

3. Durazzo M, Belci P, Collo A, Grisoglio E, Bo S. Focus on therapeutic strategies of nonalcoholic Fatty liver disease. Int J Hepatol. 2012; 2012: 464706.

4. Machann J, Thamer C, Schnoedt B, Stefan N, Haring HU, Claussen CD, et al. Hepatic lipid accumulation in healthy subjects: a comparative study using spectral fat-selective MRI and volume-localized 1H-MR spectroscopy. Magn Reson Med. 2006; 55: 913-917.

5. Musso G, Gambino R, Cassader M. Recent insights into hepatic lipid metabolism in non-alcoholic fatty liver disease (NAFLD). Prog Lipid Res. 2009; 48: 1-26.

6. Chung MY, Yeung SF, Park HJ, Volek JS, Bruno RS. Dietary a- and?-tocopherol supplementation attenuates lipopolysaccharide-induced oxidative stress and inflammatory-related responses in an obese mouse model of nonalcoholic steatohepatitis. J Nutr Biochem. 2010; 21:1200-1206.

7. Pérez-Carreras M, Del Hoyo P, Martín MA, Rubio JC, Martín A, Castellano G, et al. Defective hepatic mitochondrial respiratory chain in patients with nonalcoholic steatohepatitis. Hepatology. 2003; 38: 999-1007.

8. Lefkowitch JH. Morphology of alcoholic liver disease. Clin Liver Dis. 2005; 9: 37-53.

9. Pais R, Pascale A, Fedchuck L, Charlotte F, Poynard T, Ratziu V. Progression from isolated steatosis to steatohepatitis and fibrosis in nonalcoholic fatty liver disease. Clin Res Hepatol Gastroenterol. 2011; 35: 23-28.

Page 5: Non-Alcoholic Fatty Liver Disease – A Brief Insight into … · 2015. 6. 20. · Non-alcoholic fatty liver disease is one of the leading causes of chronic liver diseases affecting

Central

Thirunavukkarasu et al. (2015)Email:

J Liver Clin Res 2(2): 1014 (2015) 5/7

10. Chen A, Tang Y, Davis V, Hsu FF, Kennedy SM, Song H, et al. Liver fatty acid binding protein (L-Fabp) modulated murine stellate cell activation and diet-induced nonalcoholic fatty liver disease. Hepatology. 2013; 57: 2202-2212.

11. Caldwell S, Argo C. The natural history of non-alcoholic fatty liver disease. Dig Dis. 2010; 28: 162-168.

12. Pessayre D. Role of mitochondria in non-alcoholic fatty liver disease. J Gastroenterol Hepatol. 2007; 22 Suppl 1: S20-27.

13. Jaeschke H. Reactive oxygen and mechanisms of inflammatory liver injury: Present concepts. J Gastroenterol Hepatol. 2011; 26 Suppl 1: 173-179.

14. Birkenfeld AL, Shulman GI. Nonalcoholic fatty liver disease, hepatic insulin resistance, and type 2 diabetes. Hepatology. 2014; 59: 713-723.

15. Braunersreuther V, Viviani GL, Mach F, Montecucco F. Role of cytokines and chemokines in non-alcoholic fatty liver disease. World J Gastroenterol. 2012; 18: 727-735.

16. Das SK, Balakrishnan V. Role of cytokines in the pathogenesis of non-alcoholic Fatty liver disease. Indian J Clin Biochem. 2011; 26: 202-209.

17. Gerner RR, Wieser V, Moschen AR, Tilg H. Metabolic inflammation: role of cytokines in the crosstalk between adipose tissue and liver. Can J Physiol Pharmacol. 2013; 91: 867-872.

18. Choy E, Ganeshalingam K, Semb AG, Szekanecz Z, Nurmohamed M. Cardiovascular risk in rheumatoid arthritis: recent advances in the understanding of the pivotal role of inflammation, risk predictors and the impact of treatment. Rheumatology (Oxford). 2014; 53: 2143-2154.

19. Ouyang W, Rutz S, Crellin NK, Valdez PA, Hymowitz SG. Regulation and functions of the IL-10 family of cytokines in inflammation and disease. Annu Rev Immunol. 2011; 29: 71-109.

20. Tiniakos DG, Vos MB, Brunt EM. Nonalcoholic fatty liver disease: pathology and pathogenesis. Annu Rev Pathol. 2010; 5: 145-171.

21. Montecucco F, Mach F. Does non-alcoholic fatty liver disease (NAFLD) increase cardiovascular risk? Endocr Metab Immune Disord Drug Targets. 2008; 8: 301-307.

22. Tosello-Trampont AC, Landes SG, Nguyen V, Nonobransteva TI, Hahn YS. Kuppfer cells trigger nonalcoholic steatohepatitis development in diet-induced mouse model through tumor necrosis factor-a production. J Biol Chem. 2012; 287: 40161-40172.

23. Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS. Protection from obesity-induced insulin resistance in mice lacking TNF-alpha function. Nature. 1997; 389: 610-614.

24. Pinto Lde F, Compri CM, Fornari JV, Bartchewsky W, Cintra DE, Trevisan M, et al. The immunosuppressant drug, thalidomide, improves hepatic alterations induced by a high-fat diet in mice. Liver Int. 2010; 30: 603-610.

25. Liew PL, Chen CL, Lee YC, Huang MT, Wang W, Lee WJ. Hepatic tumor necrosis factor-a, leptin and adiponectin expression in morbid obese patients: Clinicopathological correlations. Obes Res Clin Pract. 2012; 6: e1-e90.

26. Lee YM, Sutedja DS, Wai CT, Dan YY, Aung MO, Zhou L, et al. A randomized controlled pilot study of Pentoxifylline in patients with non-alcoholic steatohepatitis (NASH). Hepatol Int. 2008; 2: 196-201.

27. McNelis JC, Olefsky JM. Macrophages, immunity, and metabolic disease. Immunity. 2014; 41: 36-48.

28. Frasinariu OE, Ceccarelli S, Alisi A, Moraru E, Nobili V. Gut-liver axis and fibrosis in nonalcoholic fatty liver disease: an input for novel therapies. Dig Liver Dis. 2013; 45: 543-551.

29. Shek FW, Benyon RC. How can transforming growth factor beta be targeted usefully to combat liver fibrosis? Eur J Gastroenterol Hepatol. 2004; 16: 123-126.

30. Karkampouna S, Goumans MJ, Ten Dijke P, Dooley S, Kruithof-De Julio M. Inhibition of TGFβ type I receptor activity facilitates liver regeneration upon acute CCl4 intoxication in mice. Arch Toxicol. 2015;.

31. Perez-Vargas JE, Zarco N, Shibayama M, Segovia J, Tsutsumi V, Muriel P. Hesperidin prevents liver fibrosis in rats by decreasing the expression of nuclear factor-κB, transforming growth factor-β and connective tissue growth factor. Pharmacology. 2014; 94: 80-89.

32. Starkel P, Sempoux C, Leclercg I, Herin M, Deby C, Desager JP, et al. Oxidative stress, KLF6 and transforming growth factor beta up-regulation differentiate non-alcoholic steatohepatitis progressing to fibrosis from uncomplicated steatosis in rats. J Hepatol. 2003; 39: 538-546.

33. Dixon JB, Bhathal PS, Jonsson JR, Dixon AF, Powell EE, O’Brien PE. Pro-fibrotic polymorphisms predictive of advanced liver fibrosis in the severely obese. J Hepatol. 2003; 39: 967-971.

34. Xiao J, Ho CT, Liong EC, Nanji AA, Leung TM, Lau TY, et al. Epigallocatechin gallate attenuates fibrosis, oxidative stress, and inflammation in non-alcoholic fatty liver disease rat model through TGF/SMAD, PI3K/Akt/FOXO1, and NF-kappa B pathways. Eur J Nutr. 2014; 53:187-199.

35. Pathil A, Mueller J, Ludwig JM, Wang J, Warth A, Chamulitrat W, et al. Ursodeoxycholyl lysophosphatidylethanolamide attenuated hepatofibrogenesis by impairment of TGF-ß1/Smad2/3 signalling. Br J Pharmacol. 2014; 171:5113-5126.

36. Emanuela F, Grazia M, Marco de R, Maria Paola L, Giorgio F, Marco B. Inflammation as a Link between Obesity and Metabolic Syndrome. J Nutr Metab. 2012; 2012: 476380.

37. Kishimoto T. IL-6: from its discovery to clinical applications. Int Immunol. 2010; 22: 347-352.

38. Teoh N, Field J, Farrell G. Interleukin-6 is a key mediator of the hepatoprotective and pro-proliferative effects of ischaemic preconditioning in mice. J Hepatol. 2006; 45: 20-27.

39. Blindenbacher A, Wang X, Langer I, Savino R, Terracciano L, Heim MH. Interleukin 6 is important for survival after partial hepatectomy in mice. Hepatology. 2003; 38: 674-682.

40. Croker BA, Kiu H, Pellegrini M, Toe J, Preston S, Metcalf D, et al. IL-6 promotes acute and chronic inflammatory disease in the absence of SOCS3. Immunol Cell Biol. 2012; 90: 124-129.

41. Nakagawa H, Fujiwara N, Tateishi R, Arano T, Nakagomi R, Kondo M, et al. Impact of serum levels of interleukin-6 and adiponectin on all-cause, liver-related, and liver unrelated mortality in chronic hepatitis C patients. J Gastroenterol Hepatol. 2015; 30: 379-388.

42. Mas E, Danjoux M, Garcia V, Carpentier S, Ségui B, Levade T. IL-6 deficiency attenuates murine diet-induced non-alcoholic steatohepatitis. PLoS One. 2009; 4: e7929.

43. Yamaguchi K, Nishimura T, Ishiba H, Seko Y, Okajima A, Fujii H, et al. Blockade of interleukin 6 signalling ameliorates systemic insulin resistance through upregulation of glucose uptake in skeletal muscle and improves hepatic steatosis in high-fat diet fed mice. Liver Int. 2015; 35:550-561.

44. Hamirani YS, Katz R, Nasir K, Zeb I, Blaha MJ, Blumenthal RS, et al. Association between inflammatory markers and liver fat: The Multi-Ethnic Study of Atherosclerosis. J Clin Exp Cardiolog. 2014; 5.

45. McMahan RH, Wang XX, Cheng LL, Krisko T, Smith M, El Kasmi K, et

Page 6: Non-Alcoholic Fatty Liver Disease – A Brief Insight into … · 2015. 6. 20. · Non-alcoholic fatty liver disease is one of the leading causes of chronic liver diseases affecting

Central

Thirunavukkarasu et al. (2015)Email:

J Liver Clin Res 2(2): 1014 (2015) 6/7

al. Bile acid receptor activation modulates hepatic monocyte activity and improves nonalcoholic fatty liver disease. J Biol Chem. 2013; 288:11761-11770.

46. Gerard C, Rollins BJ. Chemokines and disease. Nat Immunol. 2001; 2: 108-115.

47. Saiman Y, Friedman SL. The role of chemokines in acute liver injury. Front Physiol. 2012; 3: 213.

48. Bertola A, Bonnafous S, Anty R, Patouraux S, Saint-Paul MC, Iannelli A, et al. Hepatic expression patterns of inflammatory and immune response genes associated with obesity and NASH in morbidly obese patients. PLoS One. 2010; 5: e13577.

49. Chen HJ, Kang SP, Lee IJ, Lin YL. Glycyrrhetinic acid suppressed NF-κB activation in TNF-α-induced hepatocytes. J Agric Food Chem. 2014; 62: 618-625.

50. Luedde T, Schwabe RF. NF-κB in the liver linking injury, fibrosis and hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2011; 8: 108-118.

51. Wobser H, Dorn C, Weiss TS, Amann T, Bollheimer C, Büttner R, et al. Lipid accumulation in hepatocytes induces fibrogenic activation of hepatic stellate cells. Cell Res. 2009; 19: 996-1005.

52. Tamura Y, Sugimoto M, Murayama T, Minami M, Nishikaze Y, Ariyasu H, et al. C-C chemokine receptor 2 inhibitor improves diet-induced development of insulin resistance and hepatic steatosis in mice. J Atheroscler Thromb. 2010; 17: 219-228.

53. Miura K, Yang L, Van Rooijen N, Ohnishi H, Seki E. Hepatic recruitment of macrophages promotes nonalcoholic steatohepatitis through CCR2. Am J Physiol Gastrointest Liver Physiol. 2012; 302: G1310-G1321.

54. Obstfeld AE, Suguru E, Thearle M, Francisco AM, Gayet C, Ginsberg HN, et al. C-C chemokine receptor 2 (CCR2) regulates the hepatic recruitment of myeloid cells that promote obesity-induced hepatic steatosis. Diabetes. 2010; 59: 916-925.

55. Cynis H, Kehlen A, Haegele M, Hoffmann T, Heiser U, Fujii M, et al. Inhibition of Glutaminyl Cyclases alleviates CCL2-mediated inflammation of non-alcoholic fatty liver disease in mice. Int J Exp Pathol. 2013; 94: 217-225.

56. Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature. 2012; 482:179-185.

57. Kirovski G, Gäbele E, Dorn C, Moleda L, Niessen C, Weiss TS, et al. Hepatic steatosis causes induction of the chemokine RANTES in the absence of significant hepatic inflammation. Int J Clin Exp Pathol. 2010; 3: 675-680.

58. Pérez-Martínez L, Pérez-Matute P, Aguilera-Lizarraga J, Rubio-Mediavilla S, Narro J, Recio E, et al. Maraviroc, a CCR5 antagonist, ameliorates the development of hepatic steatosis in a mouse model of non-alcoholic fatty liver disease (NAFLD). J Antimicrob Chemother. 2014; 69:1903-1910.

59. Eisinger K, Krautbauer S, Hebel T, Schmitz G, Aslanidis C, Liebisch G, et al. Lipidomic analysis of the liver from high-fat diet induced obese mice identifies changes in multiple lipid classes. Exp Mol Pathol. 2014; 97: 37-43.

60. Torer N, Ozenirler S, Yucel A, Bukan N, Erdem O. Importance of cytokines, oxidative stress and expression of BCL-2 in the pathogenesis of non-alcoholic steatohepatitis. Scand J Gastroenterol. 2007; 42: 1095-1101.

61. Müller M, Carter S, Hofer MJ, Campbell IL. Review: The chemokine receptor CXCR3 and its ligands CXCL9, CXCL10 and CXCL11 in neuroimmunity--a tale of conflict and conundrum. Neuropathol Appl Neurobiol. 2010; 36: 368-387.

62. Berres ML, Asmacher S, Lehmann J, Jansen C, Gortzen J, Klein S, et al. CXCL9 is a prognostic marker in patient with liver cirrhosis receiving tranjugular intrahepatic portosystemic shunt. J Hepatol. 2015; 62: 332-329.

63. Zhang X, Shen J, Man K, Chu ES, Yau TO, Sung JC, et al. CXCL10 plays a key role as an inflammatory mediator and a non-invasive biomarker of non-alcoholic steatohepatitis. J Hepatol. 2014; 61: 1365-1375.

64. Wasmuth HE, Lammert F, Zaldivar MM, Weiskirchen R, Hellerbrand C, Scholten D, et al. Antifibrotic effects of CXCL9 and its receptor CXCR3 in livers of mice and humans. Gastroenterology. 2009; 137: 309-319, 319.

65. Tailleux A, Wouters K, Staels B. Roles of PPARs in NAFLD: potential therapeutic targets. Biochim Biophys Acta. 2012; 1821: 809-818.

66. Staels B, Maes M, Zambon A. Fibrates and future PPARalpha agonists in the treatment of cardiovascular disease. Nat Clin Pract Cardiovasc Med. 2008; 5: 542-553.

67. Van Diepen JA, Jansen PA, Ballak DB, Hijmans A, Hooiveld GJ, Rommelaere S, et al. PPAR-alpha dependent regulation of vanin-1 mediates hepatic lipid metabolism. J Hepatol. 2014; 61: 366-372.

68. Kleemann R, Gervois PP, Verschuren L, Staels B, Princen HM, Kooistra T. Fibrates down-regulate IL-1-stimulated C-reactive protein gene expression in hepatocytes by reducing nuclear p50-NFkappa B-C/EBP-beta complex formation. Blood. 2003; 101: 545-551.

69. Staels B, Rubenstrunk A, Noel B, Rigou G, Delataille P, Millatt LJ, et al. Hepatoprotective effects of the dual peroxisome proliferator-activated receptor alpha/delta agonist, GFT505, in rodent models of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Hepatology. 2013; 58: 1941-1952.

70. Larter CZ, Yeh MM, Van Rooyen DM, Brooling J, Ghatora K, Farrell GC. Peroxisome proliferator-activated receptor-a agonist, Wy 14,643, improves metabolic indices, steatosis and ballooning in diabetic mice with non-alcoholic steatohepatitis. J Gastroenterol Hepatol. 2012; 27: 341-350.

71. Abdelmegeed MA, Yoo SH, Henderson LE, Gonzalez FJ, Woodcroft KJ, Song BJ. PPARalpha expression protects male mice from high fat-induced nonalcoholic fatty liver. J Nutr. 2011; 141: 603-610.

72. Bechmann LP, Vetter D, Ishida J, Hannivoort RA, Lang UE, Kocabayoglu P, et al. Post-transcriptional activation of PPAR alpha by KLF6 in hepatic steatosis. J Hepatol. 2013; 58: 1000-1006.

73. Stientstra R, Mandard S, Pastsouris D, Maass C, Kersten S, Muller M. Peroxisome proliferator-activated receptor alpha protects against obesity-induced hepatic inflammation. Endocrinology. 2007; 148: 2753-2763.

74. Lalloyer F, Staels B. Fibrates. Glitazones, and peroxisome proliferator-activated receptors. Arterioscler Thromb Vasc Biol. 2010; 30: 894-899.

75. Yu S, Matsusue K, Kashireddy P, Cao WQ, Yeldandi V, Yeldandi AV, et al. Adipocyte-specific gene expression and adipogenic steatosis in the mouse liver due to peroxisome proliferator-activated receptor gamma1 (PPAR gamma1) over expression. J Biol Chem. 2003; 278:498-505.

76. Moran-Salvador E, Lopez-Parra M, Garcia-Alonso V, Titos E, Martinez-Clemente M, Gonzalez-Periz A, et al. Role of PPAR? in obesity-induced heaptic steatosis as determined by hepatocyte- and macrophage-specific conditional knockouts. FASEB J. 2011; 25: 2538-2550.

77. Wang F, Mullican SE, DiSpirito JR, Peed LC, Lazar MA. Lipoatrophy and severe metabolic disturbance in mice with fat-specific deletion of PPARγ. Proc Natl Acad Sci U S A. 2013; 110: 18656-18661.

78. Nan YM, Han F, Kong LB, Zhao SX, Wang RQ, Wu WJ, et al. Adenovirus-

Page 7: Non-Alcoholic Fatty Liver Disease – A Brief Insight into … · 2015. 6. 20. · Non-alcoholic fatty liver disease is one of the leading causes of chronic liver diseases affecting

Central

Thirunavukkarasu et al. (2015)Email:

J Liver Clin Res 2(2): 1014 (2015) 7/7

Suyavaran A, Mareeswaran R, Ramamurthy C, Subastri A, Rao PL, et al. (2015) Non-Alcoholic Fatty Liver Disease – A Brief Insight into Pathogenesis and Review of Recent Reports on Therapeutic Targets J Liver Clin Res 2(2): 1014.

Cite this article

mediated peroxisome proliferator activated receptor gamma over expression prevent nutritional fibrotic steatohepatitis in mice. Scand J Gastroenterol. 2011; 46: 358-369.

79. Sharvit E, Abramovitch S, Reif S, Bruck R. Amplified inhibition of stellate cell activation pathways by PPAR-γ, RAR and RXR agonists. PLoS One. 2013; 8: e76541.

80. Wahli W, Michalik L. PPARs at the crossroads of lipid signaling and inflammation. Trends Endocrinol Metab. 2012; 23: 351-363.

81. Bojic LA, Sawyez CG, Telford DE, Edwards JY, Hegele RA, Huff MW. Activation of peroxisome proliferator-activated receptor d inhibits human macrophage foam cell formation and the inflammatory

response induced by very low-density lipoprotein. Arterioscler Thromb Vasc Biol. 2012; 32:2919-2928.

82. Nagasawa T, Inada Y, Nakano S, Tamura T, Takahashi T, Maruyama K, et al. Effects of bezafibrate, PPAR pan-agonist, and GW501516, PPARdelta agonist, on development of steatohepatis in mice fed a methionine- and choline-deficient diet. Eur J Pharmacol. 2006; 536:182-191.

83. Shan W, Nicol CJ, Ito S, Bility MT, Kennett MJ, Ward JM, et al. Peroxisome proliferator-activated receptor –beta/delta protects against chemically induced liver toxicity in mice. Hepatology. 2008; 47: 225-235.