eISSN: 2449-8238
ISSN: 2392-1099
Clinical and Experimental Hepatology
Current issue Archive Manuscripts accepted About the journal Editorial board Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
3/2017
vol. 3
 
Share:
Share:
Review paper

The interplay between Th17 and T-regulatory responses as well as adipokines in the progression of non-alcoholic fatty liver disease

Magdalena Świderska
,
Jerzy Jaroszewicz
,
Agnieszka Stawicka
,
Anna Parfieniuk-Kowerda
,
Adrian Chabowski
,
Robert Flisiak

Clin Exp HEPATOL 2017; 3, 3: 127-134
Online publish date: 2017/06/21
Article file
- The interplay.pdf  [0.11 MB]
Get citation
 
PlumX metrics:
 

Introduction

Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease, which affects 6.3% to 33% of worldwide population [1]. The prevalence of NAFLD industrialized countries is estimated for 1/3 of adult population and 25-30% in Europe [2, 3]. Non-alcoholic fatty liver disease is associated with an accumulation of fat in liver despite the lack of secondary causes. Non-alcoholic fatty liver disease includes different histologic and clinic types: non-alcoholic fatty liver (NAFL) and non-alcoholic steatohepatitis (NASH), which is usually associated with ongoing liver fibrosis [2]. Non-alcoholic fatty liver disease may lead to liver cirrhosis with portal hypertension and hepatocellular carcinoma (HCC) [2, 4]. Frequently, NAFLD is connected with higher risk of obesity, cardiovascular diseases, metabolic syndrome, type 2 diabetes, and insulin resistance [2, 4]. It is assumed that among patients with NAFLD, approximately 33% will develop NASH [5]. Cirrhosis may occur in 5-19% of patients diagnosed with NASH [4]. Understanding of the mechanisms, which are responsible for progression from simple steatosis to NASH, may be crucial in diagnostics and planning therapy for patients with progressive disease.
Pathogenesis of NASH is complex with a variety of different processes that may lead to liver damage [4]. The balance between pro- and anti-inflammatory processes plays a crucial role, mediated by both, innate and adaptive immunity. Aforementioned mechanisms are a part of induction and progression of metabolic liver damage [3]. Accumulation of lipid deposits in hepatocytes leads to synthesis of proinflammatory cytokines, and finally results in development of hepatitis [6, 7]. Cytokines and chemokines are important mediators of steatosis. The damage of the organ is connected with an increase of tumor necrosis factor (TNF-α) and interleukin 1 beta (IL-1β) synthesis, activating the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) inflammatory signal as well as IL-12 and IL-23, which induce Th1 and Th17 response [6]. What is more, in NAFLD pathogenesis, hormones called adipokines, are of vast importance. Secretory activity of fatty tissue changes in steatosis, causing the release of adipokines, which participate in insulin resistance development [8]. In patients with NAFLD, an increased concentration TNF-α, IL-6 and resisting is observed as well as decreased concentration of adiponectin. It is possible that retinol binding protein 4 (RBP-4) and hyaluronic acid participate in development of the disease [9]. Nevertheless, the crosslink between immunity and adiponectin as well as profibrogenic pathways leads to progression of disease.

T helper cells (Th)

It is a subgroup of lymphocytes, which participate in adaptive immunity. They release cytokines that activate the immune cells, including cytotoxic T lymphocytes and macrophages. T helper cells (CD4+) may adopt a proinflammatory (Th1) phenotype, which is important for synthesis of interferon (INF-α). Their function is protection from viral, bacterial, and parasites infections. Th1 response is also beneficial for antineoplastic immunity [4, 10]. Furthermore, CD4+ may develop anti-inflammatory phenotype – Th2 characterized by synthesis of IL-4, IL-5, and IL-13 [4, 11]. Differentiation of CD4+ T cells into the Th2 subset is controlled by GATA-3-transcription factor into Th1 subset monitored by T-bet [12]. The balance between Th1 and Th2 is important, and lack or excess of proinflammatory cytokines in relation to deficiency of anti-inflammatory cytokines have been observed in the course of NASH in liver and visceral adipose tissue (Fig. 1) [4, 13, 14]. Impaired balance of Th1/Th2 may result in rapid growth of cytotoxic cells T (CD8+), which may release proinflammatory mediators, participating in recruiting and activation of macrophages into adipose tissue (Fig. 1) [11, 14]. A disruption of Th1/Th2 balance towards the Th1 polarization was demonstrated in diabetic patients. Interestingly, the predominant Th2 response was common in patients with diabetes and NASH [4]. Furthermore, it was shown in mouse model that lipid metabolism alterations observed in NAFLD result in a selective loss of intrahepatic CD4+, but not CD8+ T cells, which can lead to hepatocarcinogenesis. The researchers conclude that there is an association between obesity-induced lipid accumulation and selective loss of CD4+ T cells, which suggested a critical role for the CD4+ T lymphocytes in the progression NAFLD to develop HCC [15].

Th17 interleukin-17 (IL-17)-producing effector CD4+ T (Th17) cells

Lymphocytes T, among others subpopulations, include IL-17 releasing cells that are classified as Th17 type of response. For immunology hemostasis it is important to preserve proper Th17 function, as its lack was observed in many autoimmune infectious and metabolic diseases [11, 10]. Th17 cells release interleukins IL-17A, IL-17F, IL-21, and IL-22 (Fig. 1) [10, 14, 15]. Th17 cells are also proinflammatory cells [16, 17] that participate in bacterial and fungal infections, but may also contribute to nonspecific damage of organs in the course of autoimmune or metabolic diseases [7, 10]. Furthermore, Th17 lymphocytes stimulate the synthesis of proinflammatory cytokines, which in turn stimulate the secretion of chemokines by local cells that leads to the recruitment of neutrophils and macrophages to the area of inflammation. Permanent stimulation of Th17 cells promotes chronic inflammatory activity associated with damage of tissues in autoimmune diseases. Only few reports exist evaluating the significance of Th17 cells in course of NASH in humans [7, 17-19]. It is proved that activation of Th17 cells and increase of IL-17 production promote the progression of the disease from the steatosis to steatosis with inflammation. However, particular influence of Th17 on the pathogenesis of NASH as well as potential application of determination of this immune response markers in diagnostics and prognoses of NASH have not been examined. Among main cytokines associated with type Th17 response are counterbalancing each other IL-17A, IL-17F, and interleukin 21 (IL-21) [16, 19, 20].
Th17 cells play an important role in chronic inflammatory diseases. The increased number of circulating and intrahepatic Th1 cells was found in chronic hepatitis C (CHC) and correlated with the severity of hepatitis [21]. Interleukin 17 probably participates in pathogenesis of liver fibrosis [16, 20]. The highest synthesis among Th17 cells was revealed with regard to IL-17A. It has also higher biologic activity compared to IL-17F. IL-17A is produced mainly by CD4+ Th17 cells, but also by different cells including CD8+ T, natural killer T-cells (NKT), natural killer (NK), eosinophils, neutrophils, and monocytes. IL17A and IL17F transmit the signal through receptors IL-17RA and IL-17RC5 [17]. Animal studies revealed that IL-17 stimulates production of inflammatory cytokines IL-6, IL-1, TNF-α, and TGF-β1. What is more important, IL-17 stimulates hepatic stellate cells (HSC) by increasing the production of type I collagen through signal transducer and activator of transcription 3 (STAT3). Interleukin 17 may serve as attractive target for antifibrotic therapies [17]. The development of Th17 cells is mutually connected with T regulatory (Treg) cells [13].
Th17 cells promote the autoimmunization and tissue damage, while Treg cells act antagonistically. The balance between two subgroups of T cells is crucial for immune homeostasis [13, 16]. The differentiation of Th17 cells may be regulated by nuclear receptors LXR (liver X receptor). Nuclear receptors act like modulators of lipids and cholesterol metabolism through regulation of genes, such as sterol regulating element binding protein 1c (SREBP-1c) and ATP binding cassette transporters-1 (ABCA-1) [16]. Increased values of LDL cholesterol were observed in patients with metabolic syndrome and NASH in comparison to those without steatosis [5]. That is why cholesterol along with LXR receptors may be important for differentiation of T lymphocytes into Th17 cells [5, 10]. Differentiation of Th17 with Th0 is regulated by TGF-β1, IL-6, IL-21, and IL-235. Th17 secretes IL-22 that participates in hepatocytes protection [17].
Concentration of IL-17A correlates with the inflammatory activity of different liver diseases including alcoholic liver disease [22] primary biliary cirrhosis, chronic hepatitis B and C, transplant rejection, and hepatocellular carcinoma [18].
IL-17RA signalization is crucial for progression of the disease from NALFD to NASH [18]. Results of animal studies indicate that not only IL-17A but also IL-17F are important in NAFLD progression [23]. Concentrations of IL-17 and IL-6 are strictly correlated with each other. Interleukin 17 causes significant increase of IL-6 concentration in liver, while IL-6 is necessary in differentiation of naive CD4 T cells into Th17 [19]. The study in mouse model revealed that IL-17 is connected with progression of NAFLD, and IL-17 blocking may be promising therapeutic option for NAFLD [19].
Interleukin 22 may play both protective or pathogenic role in chronic inflammatory diseases, depending on the type and location of the affected tissue [24]. It has been shown that IL-22 was involved the process of stimulation of hepatic fibrogenesis [24, 26]. For example, some studies in mice highlight IL-22 protective role in NASH, but only in the absence of IL-17 [25]. Treatment with recombinant IL-22 in animals mitigates the damage of the liver through activation of signaling tract of STAT3; it also leads to alleviation of oxidative stress. Furthermore, long term administration of recombinant IL-22 in mice with high fat diet and induced steatosis decreased the level of TNF- in liver [26]. Increased levels of IL-22 were found in serum and liver tissues in chronic hepatitis C and alcoholic liver disease [22, 24, 26]. Researchers Wu et al. [24] in the study of 32 CHC and 64 HCV associated liver cirrhosis patients, demonstrated significant increase (p < 0.001) of IL-22 concentration in patients with HCV, in comparison to the control group that was closely associated with the stages of liver fibrosis. Activation of HSCs to release extracellular matrix, a process implicated in liver fibrosis during chronic HCV infection, can be induced by elevated concentrations of IL-22 [24].

T regulatory cells

Another component of immune response with poorly recognized significance in NASH are T regulatory cells (Treg). Those cells play an important role in supporting immune homeostasis by inhibiting proliferation and functions of T effector cells [4, 10, 17]. Tregs consist 5-10% of total number of T CD4+ cells in a healthy human. It is a separate subpopulation of mature T lymphocytes produced in thymus and expressing transcription factor Foxp3 of forkhead family [27].
Tregs are divided into two groups: natural Treg (nTregs) and adaptive or induced Treg (iTreg) [28]. Treg lymphocytes are capable of producing of main anti-inflammatory cytokines, including IL-10 and TGF-1 [4, 28]. Importantly depletion of Tregs leads to severe systemic autoimmunity [28]. Deficiencies of Treg are frequently connected with mutation of FOXP3 gene on chromosome X [27]. IL-2, TGF- are important cytokines for Treg production. Participance of TGF- in Tregs development in thymus is under discussion, but TGF-β is crucial for Treg homeostasis [27]. Treg takes part in prevention of autoreactive cells proliferation, e.g. in autoimmune hepatitis, and also takes part in negative control of different immune responses, e.g. viral hepatitis and HCC, and in organs tolerance after transplantation [11].
What is more, the induction of regulatory T cells can alleviate insulin resistance, which contributes to reducing liver damage. It has been shown that administration of a monoclonal antibody OKT3 (inductive effect of Treg) in patients with NASH during 30 days therapy, resulted in improvement in the immune parameters and good biological effects in the liver of patients with NASH (increase in CD4(+) LAP(+) and CD4(+) CD25(+) LAP(+)), an increase in TGF-, and a decrease of AST activity in one of the treatment group patients with NASH. Further research on the role of Treg cells in disease development and their application in immunotherapy of patients with NASH is appropriate [29].

Transforming growth factor β

Transforming growth factor  is one of the most important factors for immune regulation [7]. It is produced by different cell types, including lymphocytes, monocytes, and macrophages [7, 28]. Transforming growth factor appears in at least three strictly connected isoforms TGF-1, TGF-2, and TGF-3 [20, 28]. TGF-1 is an important antiproliferatic and profibrogenic cytokine. The highest concentration in patients with liver fibrosis among three isoforms shows TGF-1. In a healthy liver, the main source of TGF-1 are Kupffer cells, HSCs, and endothelial cells. Transforming growth factor  is expressed exclusively by HSCs [20]. In a large study including 1,322 healthy persons, the frequency of occurrence of NAFLD after 4 years of follow-up was associated with TGF-3 serum concentration. It was proven that higher levels of TGF-3 in serum of healthy persons increase risk of steatosis development in further observation [20]. Importantly, anti-inflammatory activity of TGF- is exerted by stimulation of FOXP3 positive synthesis (forkhead box P3) and development of T regulator cells (Tregs) [7]. Many pathologies such as advanced neoplastic processes or diseases with ongoing fibrosis [20, 28] are connected with increased activity of TGF-. Abnormal TGF-1 signalization contributes to the development of PBC through inhibition of inflammatory responses, and an increase of fibrogenesis [30]. The disruption of transduction of TGF-1 signalization contributes to the development of autoimmune diseases, liver fibrosis, and cancer [31].
Transforming growth factor  regulates the synthesis of extracellular matrix, degradation, and reconstruction, which are important processes in chronic inflammatory liver disorders. Furthermore, TGF- mediates the transformation of hepatic HSCs into myofibroblasts. Its concentration is increased in patients with NASH in comparison to patients with simple steatosis and healthy persons, which suggests that this cytokine takes part in fibrosis in NASH [20, 28]. Additionally, TGF- inhibits the differentiation of Th1 and Th2 cells, and contributes to the development of Th17, Th9, and Treg’s phenotype [10, 28]. What is interesting, the combination of TGF- with proinflammatory cytokines, including IL-6 inhibits the production of Treg and stimulates the Th17 cells, acting proinflammatory. Low concentration of TGF- restrain the expression of IL-23 receptor, which is of a great importance for Th17 cells development, enhancing the expression of Foxp3, and determining the anti-inflammatory Tregs’ response [28]. High concentration of TGF-, along with IL-6 and IL-21, results in increase of IL-23 receptor expression, facilitating Th17 phenotype [28]. Mutual relationship between Treg lymphocytes and Th17 cells constitute the delicate balance between tolerance and activation of immune response [7]. The aforementioned results explain the importance of recognizing the relation between potentially cytotoxic response of Th17 and regulatory Treg in course of NASH.

Interleukin 10

Interleukin 10 (IL-10) is an anti-inflammatory cytokine [32]. It is mainly produced by macrophages, dendric cells, B and Treg lymphocytes, but also by different cells in the liver including hepatocytes, Kupffer cells, and HSCs [26]. Bacteria, viruses, and parasites may stimulate the production of IL-10 through host cells [26]. IL-10 signalization consists of IL10R1-ligand specific and second unit, which occurs also in other cytokines receptors. Binding of IL-10 to IL10R activates the transcriptive factor signal transducer and activator of transcription 3 STAT3 [26]. STAT3 plays significant role in balance of pro- and anti-inflammatory responses [11].
In liver damage induced by alcohol, viruses, autoimmune diseases, or steatosis, IL-10 acts as a protector [33]. Deficiencies of IL-10 expression or IL-10 receptor (IL10R) lead to inflammatory diseases [32]. In animal models of induced liver steatosis IL-10 inhibition, by administering/pre-treated with recombinant murine IL-10 (BD Biosciences, USA) caused the increase of proinflammatory cytokines and despair of insulin signalization [33]. In addition, IL-10 decreases the frequency of occurrence of liver damage after different traumas. It was proven that IL-10 may protect against ischemic liver injury after organ transplantation, thus significantly increase the number of useful livers [34].

Interleukin 6

Interleukin 6 is regarded as inflammatory cytokine. Its concentration increases in obesity and body mass loss leads to the reduction of IL-6 serum concentrations [35, 36]. Increased expression of IL-6 was observed in liver of patients with NASH and was connected to the severity of the disease. Scientists notice strong relation between NASH, insulin resistance, and type 2 diabetes [22]. Furthermore, IL-6 plays important role in inflammatory response and enhances neoplastic processes such as proliferation of cells and anti apoptic process [37].

Tumor necrosis factor α

Tumor necrosis factor is a proinflammatory cytokine participating in metabolic, inflammatory, proliferative, and also necrotic processes. In response to chronic inflammatory activity in organism, TNF-is secreted by macrophages, hepatocytes, Kupffer cells, and other types of cells [38]. TNF-participates in development of steatosis and liver fibrosis in NAFLD [38]. Body mass loss, similarly as in case of IL-6, is connected with the decrease of TNF-concentration, which in turn, results in a decrease of inflammatory response [36]. Increased level of TNF-leads to activation of NF-κB that regulates immune and inflammatory response, taking part in apoptosis inhibition [36].

Adipokines

Adipokines are biologically active substances, which participate in hemostasis and blood pressure regulation, lipids and carbohydrates metabolism, atherosclerosis and inflammatory diseases [35, 39]. The family of adipokines is very broad. Adipokines include inter alia: leptin, resistin, RBP-4, lipocalin 2, ANGPTL2, IL-18, CCL2, CXCL5, NAMPT, and adiponectin [35, 38]. They are produced in visceral fatty tissue and secreted into the system of portal vein, and along with blood are transported to the liver. Adipokines expression may differ depending on the location of an adipose tissue depot. What is more, adipocytes are localized in human organism in connection with many different organs including heart, kidneys, bone marrow, lungs, and tunica adventitia of large vessels [35]. An association between impaired secretory function of adipose tissue, adipokines, and immunological cells like Th17, Treg helps to understanding the pathomechanism NAFLD and can identify biomarkers for predicting of disease progression.
Important role of different adipokines in development of various diseases including obesity, diabetes, psoriasis, scleroderma, NAFLD has been already established [31, 35, 38, 39]. Adipopectin and leptin increase insulin sensitivity and act as anti-inflammatory mediators [40]. Adipopectin also inhibits proinflammatory cytokines (including TNF-α) and stimulates anti-inflammatory cytokines (IL-10), which, as consequence, leads to impediment of macrophages function [40]. Adipopectin also alleviates oxidative stress and fibrinogenesis. Decreased level of adipopectin is connected with metabolic syndrome and NAFLD [40].

Leptin

Leptin is a hormone derived from adipocytes, which reflects the nutritional status of organism [40, 41]. Leptin shows proinflammatory and profibrogenic properties of significant importance in pathogenesis of liver diseases including NASH [40]. Increased concentrations of leptin is correlated with obesity, which initiates proinflammatory mechanisms. Similarly to proinflammatory cytokines, leptin participates in differentiation of Th1 cells in adipose tissue, CD8+ T-cells, macrophages, and mast cells, which stimulate synthesis proinflammatory cytokines (such as TNF-, IL-6, and IL-12) [41]. Decreased concentrations of leptin in plasma suggests malnutrition and leads to impairment of immune system function [41]. Leptin participates in NASH development contributing to insulin resistance and steatosis, but also in regulation of hepatic HSCs [38]. Leptin level correlates with obesity and steatosis [38], however, further studies are needed in order to confirm the influence of leptin on NAFLD.

Retinol binding protein-4

Retinol binding protein-4 (RBP-4) is a protein responsible for transportation of retinol (vitamin A) stored and synthesized in hepatocytes and adipocytes [39]. Concentration of RBP-4 in serum is frequently increased in insulin resistance in obese patients and type 2 diabetes [35, 38, 42]. Serum concentration of RBP-4 may be associated with inflammatory response in obese patients and may increase in advanced stage of NAFLD [38, 42]. However, the role of RBP4 in pathogenesis of NAFLD is not completely understood.

Angiopoietin Like Protein 2 (ANGPTL)

Angiopoietin Like Protein 2 (ANGPTL) is regarded as adipokine promoting inflammation and insulin resistance. Deficiency of ANGPTL2 leads to alleviation of inflammatory reaction through decreasing the activity of proinflammatory cytokines in adipose tissue and reduction of insulin resistance in mice. Its increased amounts lead to intensification of adipose tissue inflammation and resistance for insulin. It activates the transition of signals through integrins, which induces inflammation via endothelial cells, monocytes, and macrophages [35].

Ghrelin

Ghrelin is a small peptide (28 amino acids), which participates in regulation of appetite, body mass, and energy balance [37]. Importantly, ghrelin influences insulin sensitivity [38]. In an animal study in rats in which liver steatosis was induced by high fat diet, treatment with ghrelin reduced damage caused by steatosis, oxidative stress, and inflammatory state. Authors concluded that ghrelin might have a protective activity in NAFLD [37].
The study by Zwirska-Korczala et al. demonstrated that there is no decrease of ghrelin in plasma after a meal in obese women, although it is observed in lean women. The researchers suggest that chronic administration of ghrelin increases weight gain. An increased ghrelin level in plasma and lack of decrease after a meal is characteristic feature of obesity. However, paying attention to the anti-inflammatory ghrelin in obese patients, could have an impact in the progression of metabolic diseases to NASH [43].

Resistin

Resistin is an adipokine produced mainly by nucleated peripheral blood cells [38] and it is composed of 108 amino acids. It is believed that resistin may be a mediator of hepatic insulin resistance [44]. There is a relationship between resistin concentration and obesity. However, its relationship with type 2 diabetes and insulin resistance is not clear [38]. It was found that serum resistin level was directly proportional to body fat mass. In a study of 40 patients with NAFLD it was noted significantly higher resistin-values in comparison with a group of 40 healthy volunteers [44].

Visfatin

Visfatin is mainly secreted by visceral adipose tissue, but also by hepatocytes, lymphocytes, monocytes, and neutrophils [31, 45]. Visfatin level in plasma is associated with the amount of fat tissue, type 2 diabetes, and metabolic syndrome [31]. It has been shown that the concentration of visfatin levels and mRNA levels in a visceral adipose tissue and the liver were increased in a rat model [46]. Authors have suggested strengthening role of visfatin inflammatory response in the formation of visceral adipose tissue rats [46]. Furthermore, visfatin can induce TNF-α, which is associated with obesity and insulin resistance [46, 47]. Insulin resistance is accompanied with increase of visfatin concentration in serum. It is supposed that visfatin resembles the activity of insulin, i.e. inhibits gluconeogenesis in liver and also increases demand for glucose in skeletal muscles and adipose tissue [45]. In different studies, the correlation between visfatin concentration and development of NAFLD has been searched, but not confirmed [31, 45]. In the study [48], serum visfatin levels were significantly associated with NAFLD/NASH. Importantly, the study involving 70 patients with NAFLD showed a relationship between visfatin levels and the degree of liver fibrosis. The researchers suggested the application of visfatin as a non-invasive method for advanced fibrosis allowing to avoid liver biopsy in numerous patients with NAFLD [49]. It has been demonstrated in human studies that visfatin level in the liver tissue depends on the severity of liver fibrosis. There is no evidence that visfatin expression connects with liver steatosis degree; therefore, the expression of visfatin does not distinguish NASH from NAFLD [47].

Conclusions

In conclusion, it has to be highlighted that immune pathways, especially Th17 and Treg, as well as adipokines are a link between insulin resistance, obesity, and NAFLD. However, the significance of Th17, Treg, and adipokines in the course of NAFLD and progression to NASH requires further extension of research with well characterize group of patients with NAFLD and NASH. It is important to recognize the crucial role of Th17/Treg balance in course of NASH not only in the pathogenesis of the disease, but also as potential target of new therapies.

Acknowledgement

This study was supported by funds from National Leading Research Centre in Bialystok.

Disclosure

Authors report no conflict of interest.

Reference

1. Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and management of non-alcoholic fatty liver disease: practice Guideline by the American Association for the Study of Liver Diseases, American College of Gastroenterology, and the American Gastroenterological Association. Hepatology 2012; 55: 2005-2023.
2. Firneisz G. Non-alcoholic fatty liver disease and type 2 diabetes mellitus: the liver disease of our age? World J Gastroenterol 2014; 20: 9072-9089.
3. Blachier M, Leleu H, Peck-Radosavljevic M, et al. The burden of liver disease in Europe: a review of available epidemiological data. J Hepatol 2013; 58: 593-608.
4. Vonghia L, Magrone T, Verrijken A, et al. Peripheral and Hepatic Vein Cytokine Levels in Correlation with Non-Alcoholic Fatty Liver Disease (NAFLD)-Related Metabolic, Histological, and Haemodynamic Features. PLoS One 2015; 10: e0143380.
5. Zimmermann A, Zimmermann T, Schattenberg J, et al. Alterations in lipid, carbohydrate and iron metabolism in patients with non-alcoholic steatohepatitis (NASH) and metabolic syndrome. Eur J Intern Med 2011; 22: 305-310.
6. Onyekwere CA, Ogbera AO, Samaila AA, et al. Nonalcoholic fatty liver disease: Synopsis of current developments. Niger J Clin Pract 2015; 18: 703-712.
7. Tang Y, Bian Z, Zhao L, et al. Interleukin-17 exacerbates hepatic steatosis and inflammation in non-alcoholic fatty liver disease. Clin Exp Immunol 2011; 166: 281-290.
8. Mohan SK, Veeraraghavan VP, Jainu M. Effect of pioglitazone, quercetin and hydroxy citric acid on extracellular matrix components in experimentally induced non-alcoholic steatohepatitis. Iran J Basic Med Sci 2015; 18: 832-836.
9. Orlik B, Handzlik G, Olszanecka-Glinianowicz M. Rola adipokin i insulinooporności w patogenezie niealkoholowej stłuszczeniowej choroby wątroby. Postepy Hig Med Dosw (online) 2010; 64: 212-219.
10. Barbi J, Pardoll D, Pan F. Metabolic control of the Treg/Th17 axis. Immunol Rev 2013; 252: 52-77.
11. Vonghia L, Michielsen P, Francque S. Immunological mechanisms in the Pathophysiology of Non-Alcoholic Steatohepatitis. Int J Mol Sci 2013; 14: 19867-19890.
12. Kanhere A, Hertweck A, Bhatia U, et al. T-bet and GATA3 orchestrate Th1 and Th2 differentiation through lineage-specific targeting of distal regulatory elements. Nat Commun 2012; 3: 1268.
13. Maher JJ, Leon P, Ryan JC. Beyond insulin resistance: Innate immunity in nonalcoholic steatohepatitis. Hepatology 2008; 48: 670-678.
14. de Souza AW, Mesquita Júnior D, Araújo JA, et al. Immune system: part III. The delicate balance of the immune system between tolerance and autoimmunity. Rev Bras Reumatol 2010; 50: 665-679.
15. Ma C, Kesarwala AH, Eggert T, et al. NAFLD causes selective CD4(+) T lymphocyte loss and promotes hepatocarcinogenesis. Nature 2016; 531: 253-257.
16. Maggio R, Viscomi C, Andreozzi P, et al. Normocaloric low cholesterol diet modulates Th17/Treg balance in patients with chronic hepatitis C virus infection. PLoS One 2014; 9: e112346.
17. Meng F, Wang K, Aoyama T, et al. Interleukin-17 signaling in inflammatory, Kupffer cells, and hepatic stellate cells exacerbates liver fibrosis in mice. Gastroenterology 2012; 143: 765-776.
18. Giles DA, Moreno-Fernandez ME, Stankiewicz TE, et al. Regulation of Inflammation by IL-17A and IL-17F Modulates Non- Alcoholic Fatty Liver Disease Pathogenesis. PLoS One 2016; 11: e0149783.
19. Xu R, Tao A, Zhang S, et al. Neutralization of interleukin-17 attenuates high fat diet-induced non-alcoholic fatty liver disease in mice. Acta Biochim Biophys Sin (Shanghai) 2013; 45: 726-733.
20. Wei Y, Tian Q, Zhao X, et al. Serum transforming growth factor beta 3 predicts future development of nonalcoholic fatty liver disease. Int J Clin Exp Med 2015; 8: 4545-4550.
21. Jaroszewicz J, Flisiak R. Metabolic syndrome and hepatitis C infection – brothers in arms. Liver Int 2013; 33: 1135-1137.
22. Parfieniuk-Kowerda A, Szulżyk T, Świderska M, et al. Interleukin 17F concentration is associated with degree of liver damage in alcoholic liver disease. J Hepatol 2013 ; 58 Suppl 1: S222 [Abstr. 543].
23. Wieckowska A, Papouchado BG, Li Z, et al. Increased hepatic and circulating interleukin-6 levels in human nonalcoholic steatohepatitis. Am J Gastroenterol 2008; 103: 1372-1379.
24. Wu LY, Liu S, Liu Y, et al. Up-regulation of interleukin-22 mediates liver fibrosis via activating hepatic stellate cells in patients with hepatitis C. Clin Immunol 2015; 158: 77-87.
25. Rolla S, Alchera E, Imarisio C, et al. The balance between IL-17 and IL-22 produced by liver-infiltrating T-helper cells critically controls NASH development in mice. Clin Sci (London) 2016; 130: 193-203.
26. Sziksz E, Pap D, Lippai R, et al. Fibrosis Related Inflammatory Mediators: Role of the IL-10 Cytokine Family. Mediators Inflamm 2015; 2015: 764641.
27. Loser K, Beissert S. Regulatory T cells: banned cells for decades. J Invest Dermatol 2012; 132 (3 Pt 2): 864-871.
28. Schon HT, Weiskirchen R. Immunomodulatory effects of transforming growth factor-β in the liver. Hepatobiliary Surg Nutr 2014; 3: 386-406.
29. Lalazar G, Mizrahi M, Turgeman I, et al. Oral Administration of OKT3 MAb to Patients with NASH, Promotes Regulatory T-cell Induction, and Alleviates Insulin Resistance: Results of a Phase IIa Blinded Placebo-Controlled Trial. J Clin Immunol 2015; 35: 399-407.
30. Liu B, Zhang X, Zhang FC, et al. Aberrant TGF-β1 signaling contributes to the development of primary biliary cirrhosis in murine model. World J Gastroenterol 2013; 19: 5828-5836.
31. Waluga M, Kukla M, Żorniak M, et al. Visfatin and TGF-Β1 in primary biliary cirrhosis and two other common liver diseases. Folia Med Cracov 2015; 55: 59-70.
32. Barry JC, Shakibakho S, Durrer C, et al. Hyporesponsiveness to the anti-inflammatory action of interleukin-10 in type 2 diabetes. Sci Rep 2016; 17: 21244.
33. Cintra DE, Pauli JR, Araújo EP, et al. Interleukin-10 is a protective factor against diet-induced insulin resistance in liver. J Hepatol 2008; 48: 628-637.
34. Sutter AG, Palanisamy AP, Ellet JD, et al. Intereukin-10 and Kupffer cells protect steatotic mice livers from ischemia-reperfusion injury. Eur Cytokine Netw 2014; 25: 69-76.
35. Ouchi N, Parker JL, Lugus JJ. Adipokines in inflammation and metabolic disease. Nat Rev Immunol 2011; 11: 85-97.
36. Perumpail RB, Liu A, Wong RJ, et al. Pathogenesis of hepatocarcinogenesis in non-cirrhotic nonalcoholic fatty liver disease: Potential mechanistic pathways. World J Hepatol 2015; 7: 2384-2388.
37. Li Y, Hai J, Li L, et al. Administration of ghrelin improves inflammation, oxidative stress, and apoptosis during and after non-alcoholic fatty liver disease development. Endocrine 2013; 43: 376-386.
38. Stojsavljević S, Gomerčić Palčić M, Virović Jukić L, et al. Adipokines and proinflammatory cytokines, the key mediators in the pathogenesis of nonalcoholic fatty liver disease. World J Gastroenterol 2014; 20: 18070-18091.
39. Baran A, Swiderska M, Flisiak I. The effect of topical treatment and psoriasis severity on serum retinol-binding protein-4 levels. J Dermatolog Treat 2016; 27: 114-119.
40. Vonghia L, Francque S. Cross talk of the immune system in the adipose tissue and the liver in non-alcoholic steatohepatitis: Pathology and beyond. World J Hepatol 2015; 7: 1905-1912.
41. Procaccini C, Jirillo E, Matarese G. Leptin as an immunomodulator. Mol Aspects Med 2012; 33: 35-45.
42. Terra X, Auguet T, Broch M, et al. Retinol binding protein-4 circulating levels were higher in nonalcoholic fatty liver disease vs. histologically normal liver from morbidly obese women. Obesity (Silver Spring) 2013; 21: 170-177.
43. Zwirska-Korczala K, Konturek SJ, Sodowski M, et al. Basal and postprandial plasma levels of PYY, ghrelin, cholecystokinin, gastrin and insulin in women with moderate and morbid obesity and metabolic syndrome. J Physiol Pharmacol 2007; 58 Suppl 1: 13-35.
44. Eminler AT, Aygun C, Konduk T, et al. The relationship between resistin and ghrelin levels with fibrosis in nonalcoholic fatty liver disease. J Res Med Sci 2014; 19: 1058-1061.
45. Piekarska A, Wójcik K. Rola adipokin w przewlekłych chorobach wątroby. Gastroenterologia Polska/Gastroenterology 2010; 17: 138-143.
46. Iwasa T, Matsuzaki T, Matsui S, et al. The sensitivity of adipose tissue visfatin mRNA expression to lipopolysaccharide-induced endotoxemia is increased by ovariectomy in female rats. Int Immunopharmacol 2016; 35: 243-247.
47. Kukla M, Ciupińska-Kajor M, Kajor M, et al. Liver visfatin expression in morbidly obese patients with nonalcoholic fatty liver disease undergoing bariatric surgery. Pol J Pathol 2010; 61: 147-153.
48. Jamali R, Arj A, Razavizade M, et al. Prediction of Nonalcoholic Fatty Liver Disease Via a Novel Panel of Serum Adipokines. Medicine (Baltimore) 2016; 95: e2630.
49. Chwist A, Hartleb M, Lekstan A, et al. A composite model including visfatin, tissue polypeptide-specific antigen, hyaluronic acid, and hematological variables for the diagnosis of moderate-to-severe fibrosis in nonalcoholic fatty liver disease: a preliminary study. Pol Arch Med Wewn 2014; 124: 704-712.
Copyright: © 2017 Clinical and Experimental Hepatology. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.