Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 4529 2022-09-13 09:09:09 |
2 format -1 word(s) 4528 2022-09-14 03:47:12 | |
3 format -8 word(s) 4520 2022-09-16 08:28:18 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Francisco, V.;  Sanz, M.J.;  Real, J.T.;  Marques, P.;  Capuozzo, M.;  Eldjoudi, D.A.;  Gualillo, O. Adipokines in Non-Alcoholic Fatty Liver Disease. Encyclopedia. Available online: https://encyclopedia.pub/entry/27115 (accessed on 26 April 2024).
Francisco V,  Sanz MJ,  Real JT,  Marques P,  Capuozzo M,  Eldjoudi DA, et al. Adipokines in Non-Alcoholic Fatty Liver Disease. Encyclopedia. Available at: https://encyclopedia.pub/entry/27115. Accessed April 26, 2024.
Francisco, Vera, Maria Jesus Sanz, José T. Real, Patrice Marques, Maurizio Capuozzo, Djedjiga Ait Eldjoudi, Oreste Gualillo. "Adipokines in Non-Alcoholic Fatty Liver Disease" Encyclopedia, https://encyclopedia.pub/entry/27115 (accessed April 26, 2024).
Francisco, V.,  Sanz, M.J.,  Real, J.T.,  Marques, P.,  Capuozzo, M.,  Eldjoudi, D.A., & Gualillo, O. (2022, September 13). Adipokines in Non-Alcoholic Fatty Liver Disease. In Encyclopedia. https://encyclopedia.pub/entry/27115
Francisco, Vera, et al. "Adipokines in Non-Alcoholic Fatty Liver Disease." Encyclopedia. Web. 13 September, 2022.
Adipokines in Non-Alcoholic Fatty Liver Disease
Edit

Non-alcoholic fatty liver disease (NAFLD) has become the major cause of chronic hepatic illness and the leading indication for liver transplantation in the future decades. NAFLD is also commonly associated with other high-incident non-communicable diseases, such as cardiovascular complications, type 2 diabetes, and chronic kidney disease. Aggravating the socio-economic impact of this complex pathology, routinely feasible diagnostic methodologies and effective drugs for NAFLD management are unavailable. The pathophysiology of NAFLD, defined as metabolic associated fatty liver disease (MAFLD), is correlated with abnormal adipose tissue–liver axis communication because obesity-associated white adipose tissue (WAT) inflammation and metabolic dysfunction prompt hepatic insulin resistance (IR), lipid accumulation (steatosis), non-alcoholic steatohepatitis (NASH), and fibrosis. Accumulating evidence links adipokines, cytokine-like hormones secreted by adipose tissue that have immunometabolic activity, with NAFLD pathogenesis and progression.

adipokines NAFLD liver steatosis fibrosis inflammation NASH leptin biomarkers therapy

1. Introduction

Non-alcoholic fatty liver disease (NAFLD) encompasses a spectrum of liver abnormalities characterized by increased hepatic fat content (>5%)—steatosis—in the absence of secondary causes, namely excessive alcohol consumption (>20 g/day for women and >30 g/day in men), medications, viral hepatitis, or certain hereditable conditions. Histologically, NAFLD can be categorized into non-alcoholic fatty liver (NAFL), when there is only evidence of hepatic steatosis, and non-alcoholic steatohepatitis (NASH), when, apart from steatosis, lobular inflammation and hepatocyte ballooning with or without perisinusoidal fibrosis can be observed. NASH, especially in the fibrotic state, often presents worse prognoses and frequently progresses to more severe conditions, such as cirrhosis and hepatocellular carcinoma [1].
NAFLD has become the major cause of chronic hepatic illness in children and adults, as well as the leading indication for liver transplantation in the future decades, replacing chronic hepatitis C. This high-incident pathology is also associated with extra-hepatic complications, such as chronic kidney disease and cardiovascular disease, which is the leading cause of death among NAFLD patients [2]. Mainly asymptomatic, NAFLD is considered a major societal, clinical, and research challenge due to its increasingly high prevalence, difficulties in its diagnosis, the lack of approved therapies, and its complex pathophysiology.
The development and progression of NAFLD is induced by multiple factors in a “multiple parallel-hit” model, where numerous genetic and environmental determinants (“hits”) interplay on an individual basis. These factors encompass, but are not limited to, genetic alterations, inflammation, gut dysbiosis, and metabolic abnormalities. Indeed, almost 90% of NAFLD patients present at least one of the metabolic syndrome features (abdominal obesity, hypertriglyceridemia, low HDL-cholesterol, hypertension, and high fasting glucose), and about 33% fulfill the criteria for diagnosing metabolic syndrome [3]. Therefore, metabolic-associated fatty liver disease (MAFLD) was recently proposed as a more appropriate overarching term that recognizes the metabolic risk profile of patients as the master criterion for diagnosis and considers the disease activity grade to be a continuum, which better describes disease pathophysiology [4]. Therefore, it is not surprising that the worldwide prevalence of NAFLD is rising in parallel with obesity [2], the main driver of metabolic abnormalities. Stepping up the strict correlation between obesity and NAFLD, there is a dose-response between weight loss and histological disease improvement; a 7% weight loss reverts NASH in 65–90% of patients, and a ≥10% weight loss causes fibrosis relapse in 45% of patients [3].

2. Adipokines as Potential Therapeutic Targets for NAFLD

2.1. Leptin and Adiponectin

Leptin, the forerunner and best-characterized member of the adipokine family, plays a pivotal role in appetite and body-weight homeostasis by augmenting anorexigenic neuropeptides and suppressing orexigenic factors in the central nervous system [5]. Likewise, leptin has been described as modulating several physiological processes, such as lipid and glucose metabolism, as well as both innate and adaptive immunity [5]. Most of the current knowledge about leptin’s action arose from leptin-deficient ob/ob mice and leptin-receptor-deficient db/db mice. These murine models exhibited marked hepatic alterations, such as IR, accumulation of TG and lipids, and steatosis, which were partially reverted following leptin administration to ob/ob mice [6][7]. Although leptin acts as an anti-steatotic hormone preventing the accumulation of and promoting the mobilization of hepatic lipids, leptin resistance may cause the leptin inability to alleviate hepatic steatosis [6][7]. Furthermore, high leptin plasma levels derived from obese adipose tissue are associated with hepatic inflammatory and fibrogenic mechanisms and therefore, with NAFLD development [6][7].
On the contrary, adiponectin, an adipocyte-secreted hormone inversely correlated with obesity and with determining roles in insulin sensitivity, glucose levels, and lipid metabolism [8], has been reported to protect the liver from steatosis, inflammation, and fibrosis [9]. Adiponectin augments insulin’s capacity to suppress glucose production, prevents hepatic DNL, suppresses FA synthesis in hepatocytes, and enhances FA β-oxidation [9], overall protecting the liver from steatosis. Adiponectin was also described as decreasing the production of inflammatory cytokines, such as IL-6 and TNF-α, through the modulation of toll-like receptor 4 (TLR4) signaling. By boosting the beneficial effects of adiponectin in NAFLD development, this adipokine was described to possess anti-fibrotic effects by preventing leptin profibrogenic signaling [9]. Therefore, strategies aiming to rescue the leptin–adiponectin balance, i.e., reverting the obesity-associated increased leptin levels and reduced adipokines levels, are of relevance for NAFLD treatment [9].

2.2. Ghrelin

Ghrelin stands out as one of the few peripheral peptide hormones with an orexigenic effect. Initially identified as a stomach-derived hormone, ghrelin is an endogenous ligand for the growth hormone secretagogue receptor 1a (GHSR1a). It stimulates food intake and adiposity and acts as a regulator of glucose metabolism, reward behavior, gut motility, or even hepatic lipid metabolism and the immune system [10][11]. This hormone circulates in blood in two forms, an acylated (AG) form and an unacylated form (UAG, also named DAG from desacyl ghrelin). This post-transcriptional modification is catalyzed by ghrelin O-acyltransferase (GOAT), and the producing AG is the active form that triggers the signaling of the cognate receptor GHSR1a. Initially thought to be an inactive form, it has been suggested that UAG to antagonizes AG activity on glucose metabolism and lipolysis and reduces food consumption and body weight [11][12]. Given its regulatory activity on metabolism and immune system, there is a growing interest on the role of ghrelin-GOAT system in the development and progression of NAFLD.
In murine models, the administration of ghrelin during or after diet-induced NAFLD development counteracts dysregulated hepatic lipid metabolism, oxidative stress, apoptosis, and inflammation [13][14]. Furthermore, the action mechanisms involved in the beneficial effects of ghrelin on NAFLD were investigated. The amelioration of liver injury by ghrelin was accompanied by the reestablishment of phosphoinositide 3-kinase (PI3K)/Akt and liver kinase B1 (LKB1)/AMP-activated protein kinase (AMPK) pathways [13]. Ghrelin also attenuates lipotoxicity by upregulating autophagy via AMPK/mTOR restoration and inhibiting nuclear factor kappa B (NF-κB) [14]. Recently, ghrelin was demonstrated to block the progression of NASH induced by lipopolysaccharide (LPS) in mice fed with a high-fat diet through the reduction of Kupffer cells’ M1 polarization, which is mediated by GHSR1a [15]. In addition to these results demonstrating the beneficial effects of ghrelin, the genetic deletion of ghrelin in mice also significantly reduced age-associated hepatic steatosis, partly by downregulating diacylglycerol O-acyltransferase-1, one of the key enzymes of TG synthesis [16].
Concerning the effects of the ghrelin’s isoforms, it was verified that plasma UAG levels were reduced after a sleeve gastrectomy and a Roux-en-Y gastric bypass in Wistar rats, whereas the AG:UAG ratio was augmented. Concomitantly, both surgeries diminished obesity-associated hepatic steatosis, inflammation, mitochondrial dysfunction, and endoplasmic reticulum stress. Moreover, AG has a similar effect in palmitate-treated hepatocytes, which suggests that the increased AG:UAG ratio after bariatric surgery might ameliorate obesity-associated NAFLD [17]. Possible underlying mechanisms are the reduction of lipogenesis and stimulation of mitochondrial FA β-oxidation, as well as hepatic autophagy, by relatively increased AG levels [18]. Nevertheless, in lean rats, the administration of exogenous AG induced hepatic IR and lipid accumulation, while the co-administration of UAG prevented the AG-induced effects [19]. Thus, further evaluation of the ghrelin-GOAT system and the effects of AG and UAG isoforms on NAFLD development is needed.

2.3. Resistin

Resistin (named for its ability to induce “resistance to insulin”) is the founding member of resistin-like molecules (RELMs), a family of small, secreted cysteine-rich peptides with hormone-like and pro-inflammatory activities. It is mainly secreted by adipose tissue and inflammatory cells, and its action is thought to be mediated by the TLR4 receptor, although the receptors tyrosine kinase-like orphan receptor (ROR)-1, insulin-like growth factor type 1 receptor (IGF-1R), and adenylyl cyclase-associated protein 1 (CAP1) have also emerged as potential candidate receptors [20]. Resistin has been shown to have pleiotropic effects, including the regulation of blood–glucose levels and lipid metabolism, as well as the induction of pro-inflammatory cytokines secretion or monocyte differentiation into macrophages [20]. In fact, resistin administration to C57BL/6J mice increased blood glucose, i.e., impaired glucose tolerance, due to the decreased insulin sensitivity, which was rescued after the administration of an anti-resistin antibody [21]. Similarly, the high-fat diet (HFD)-fed mice presented increased plasma resistin levels and severe IR, which is completely reversed after treatment with a resistin antisense oligonucleotide [22]. More recently, the mechanisms of action of resistin in a HFD-induced NAFLD model were disclosed [23]. Acute elevated resistin altered mitochondrial morphology and content, increased lipid accumulation, and up-regulated pro-inflammatory mediators in HFD-fed mice and palmitate-treated HepG2 cells. Furthermore, steatosis aggravation induced by resistin in mice is mediated by the AMPK/peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) pathway [23]. It was also reported that resistin treatment augments the suppressor of cytokine signaling 3 (SOCS3) expression, a suppressor of insulin signaling, in adipocytes [24].
Resistin-deficient mice demonstrated reduced hepatic glucose production and, consequently, their blood–glucose levels after fasting were low [25]. In addition, ob/ob mice and diet-induced obese mice, both lacking resistin, had reduced hepatic steatosis, since the expression of genes involved in hepatic lipogenesis and the secretion of very-low-density lipoprotein (VLDL) were decreased [26].
At the cellular level, resistin hampered glycogen synthase kinase 3β (GSK3β) phosphorylation in primary rat hepatocytes under hyperinsulinemic and hyperglycemic conditions, with further reduction of glycogen synthesis and hepatic insulin action [27]. Resistin also exerts pro-inflammatory activity by inducing the expression of cell adhesion molecules and pro-inflammatory cytokines in macrophages and mononuclear cells, contributing to the recruitment of leukocytes to inflammation sites [20]. Moreover, the secretion of resistin by infiltrated monocytes/macrophages is enhanced by pro-inflammatory mediators [20]. Of note, hepatic myeloid cells and T-lymphocytes from NAFLD patients showed a decreased response to resistin, which is associated with a failure to maintain redox homeostasis, which would be a risk factor for NAFLD severity [28]. It was also verified that resistin increased hepatic inflammation through mitogen-activated protein kinase (MAPK) signaling and the activation of a coagulation cascade in animal models [29]. Finally, resistin demonstrated profibrogenic effects by activating HSCs, which release IL8/CXCL8 and monocyte chemoattractant protein (MCP)-1/CCL2 via NF-κB, and increasing the transforming growth-factor beta (TGFβ) and collagen type I production in Kupffer cells [30][31].

2.4. Retinol Binding Protein 4 (RBP4)

Distinct mouse models have been used to elucidate the RBP4 activity in metabolic diseases. In general, elevated circulating and adipose tissue RBP4 levels have been correlated with IR, dyslipidemia, and T2DM [32]. The possible RBP4-dependent mechanisms contributing to IR include impaired insulin signaling, the down-regulation of GLUT-4 translocation, and the induction of inflammatory and lipolytic pathways in adipose tissue, as well as the induction of phosphoenolpyruvate carboxykinase in the liver, thereby increasing glucose production [32].
In genetic and dietary mouse models of NAFLD, the results of hepatic expression of RBP4 are controversial. Liu et al. observed an abnormal hepatic RBP4 expression in apoE−/− mice fed with a high-fat and high-cholesterol (HFC) diet [33]. However, Saeed et al. described reduced hepatic RBP4 levels in C57BL/6J mice fed with a HFC diet and in ob/ob mice, while serum RBP4 levels were increased in both models [34]. Apart from that, transgenic mice overexpressing human RBP4 had increased hepatic lipid accumulation, cellular ballooning, and inflammation, which were exacerbated when mice were challenged with a high-fat diet, likely through RBP4-induced mitochondrial dysfunction [33][35]. Of note, recently, novel RBP4 antagonists were reported to reduce hepatic steatosis in transgenic mice with adipocyte-specific overexpression of RBP4 [36].

2.5. Visfatin

Nicotinamide phosphoribosiltransferase (NAMPT), also called pre-B cell colony-enhancing factor (PBEF) or visfatin, functions as an intracellular enzyme (iNAMPT) mediating the synthesis of nicotinamide adenine dinucleotide (NAD+) and as a cytokine-like soluble factor secreted into extracellular space (eNAMPT) [37]. Intracellular NAMPT regulates mitochondrial biogenesis, cellular metabolism, and survival, as well as the adaptive response to cell stress; it was described as modulating pancreatic β-cell function, likely regulating glucose homeostasis and IR [38]. On the other hand, extracellular NAMPT acts mainly as an inductor of pro-inflammatory cytokine production [37]. The NAMPT extracellular form has been associated with metabolic and inflammatory disorders, but its pathophysiological mechanisms are still ill-defined [37].
Administration of NAMPT to a methionine-choline-deficient (MCD)-diet-fed mouse model of NAFLD aggravated hepatic steatosis, increased inflammatory cell infiltration and inflammatory cytokines levels and exacerbated the expression of fibrotic markers in the liver, together with the induction of endoplasmic reticulum and oxidative stress [39]. In hepatocytes, NAMPT also induced the expression of inflammatory cytokines and diminished insulin signaling through a signal transducer and activator of transcription 3 (STAT3) and NF-κB activation [40]. These results support the adverse effects of NAMPT in hepatic steatosis, inflammation, and fibrosis. However, the pharmacologic inhibition or genetic ablation of NAMPT also showed deleterious effects. The intracellular NAMPT inhibitor FK866 promoted liver steatosis in HFD-fed mice and hepatic lipid accumulation in vitro via the sirtuin 1 (SIRT1)/sterol regulatory element-binding protein 1 (SREBP1)/fatty acid synthase (FASN) pathway [41]. Similarly, the knockdown of NAMPT in hepatocyte cells led to TG accumulation through the regulation of DNL via SIRT1/AMPK pathway [42]. Accordingly, the overexpression of NAMPT in hepatocyte cell lines mitigated lipid accumulation [41]. However, hepatocyte-specific NAMPT knockout mice on low-methionine and choline-free high-fat diet showed less TG accumulation than wild-type controls but had augmented histological scores for hepatic inflammation, necrosis, and fibrosis [43]. Of note, NAMPT KO mice on a control diet also showed liver injury, since they had decreased mitochondrial proteins and respiratory capacity and increased fibrosis due to low NAD+ levels [43]. Taken together, the present data suggest that the opposing activity of NAMPT in NAFLD pathophysiology could be derived from the different roles of extracellular and intracellular NAMPT, but further studies are needed.

2.6. Chemerin

Chemerin, also named tazarotene-induced gene 2 (TIG2) and retinoic acid responder 2 (RARRES2), is secreted as inactive precursor, which is activated by proteases of the coagulation cascade, neutrophil-derived proteases (elastase and cathepsin G), bacterial proteases, and mast cell products (tryptase) [44]. This chemotactic adipokine binds to the G protein-coupled receptor chemokine-like receptor 1 (CMKLR1), which is expressed in dendritic cells, macrophages, and natural killer (NK) cells and may serve as bridge between innate and adaptive immunity [45]. Although the C-C chemokine receptor-like 2 (CCRL2) and the G protein-coupled receptor 1 (GPR1)/CMKLR2 were also described as chemerin receptors, their physiological activity is still uncertain. Chemerin and its receptor CMKLR1 are both expressed in adipose tissue [46], and they have been reported to be augmented in obesity and IR states (T2DM), decreasing after weight loss [47]. This adipokine also seems to regulate adipocyte differentiation, glucose and lipid homeostasis, and insulin sensitivity [44].
In addition to its ability to regulate glucose metabolism, IR, and inflammation, the role of chemerin in NAFLD is still unclear. The administration of recombinant chemerin ameliorate HFD-induced NASH in mice, as well as IR, leptin resistance, and liver lesions, by alleviating oxidative stress and promoting autophagy, at least in part, due to chemerin/CMKLR1-dependent activation of janus kinase 2 (JAK2)-STAT3 pathway [48]. On the contrary, the administration of a chemerin-derived C15 peptide did not affect hepatic TG accumulation, inflammation, or fibrotic gene expression in atherogenic diet-induced murine NASH [49]. Moreover, PI3K inhibition mitigated liver steatosis and KC-mediated inflammation due to the down-regulation of the chemerin receptor CMKLR1 in the liver [49]. Nevertheless, whole-body Cmklr1-gene abrogation in mice did not affect either the hepatic lipid accumulation, inflammation, fibrotic gene expression, NAS, or the IR [50]. Inconsistencies in the current data could be related to the differential modulation of hepatic chemerin in distinct murine models of NAFLD [51].

2.7. Adipocyte Fatty Acid-Binding Protein (AFABP)

There is strong evidence correlating elevated AFABP with IR and adipose tissue lipolysis in obesity and metabolic syndrome [52]. Interestingly, a recent research pointed out that AFABP as a metabolic/functional marker regulating macrophage functions likely having a determining role in pathophysiology [53]. Concerning to NAFLD, AFABP expression was elevated in Kupffer cells in both LPS-induced acute liver injury and diet-induced NAFLD [54]. In these NAFLD mice models, the pharmacological inhibition of AFABP ameliorated hepatic steatosis, macrophage infiltration, and hepatocellular ballooning [54]. Genetic ablation and the pharmacological inhibition of AFABP also attenuated bile-duct-ligation- and carbon-tetrachloride-induced liver fibrosis in mice through the reduction of collagen accumulation, liver sinusoidal endothelial cells (LSEC) capillarization, and HSC activation [55]. Mechanistically, elevated AFABP promotes LSEC capillarization, an early event of NAFLD pathogenesis, and LSEC-derived AFABP activate HSCs that augments TGFβ production and further extracellular matrix accumulation and fibrosis [55]. Furthermore, AFABP could promote hepatic inflammation through Kupffer cell activation [54]. Altogether, these findings suggest pharmacological inhibition of AFABP as a promising therapeutic strategy for NAFLD.

3. Adipokines in NAFLD: Evidence from Clinical Studies

3.1. Ghrelin

Clinical studies have demonstrated that obese patients with IR or metabolic syndrome had lower UAG and total ghrelin levels, while the AG:UAG ratio was elevated [56][57]. Moreover, the AG:UAG ratio was positively correlated with IR in both obese children and adults [56][58]. After the proof about the influence of ghrelin on insulin sensitivity, some research focused on ghrelin’s role in NAFLD, but the evidence is scarce, and not all studies take into consideration the concentrations of total ghrelin and its isoforms, AG and UAG.
Ghrelin levels were associated negatively with body mass index (BMI) in obese NAFLD patients diagnosed by ultrasonography [59] or biopsy [60]. Compared to matched healthy controls, patients with NAFLD showed reduced ghrelin levels, which correlated with IR [61]. However, in a study with NAFLD biopsy-proven patients that underwent bariatric surgery, UAG levels were increased in NASH patients compared to non-NASH subjects, while similar levels of AG were observed. Additionally, higher levels of AG, but not UAG, were observed in higher stages of fibrosis [62]. Further evidencing the role of ghrelin in NAFLD, recent case-control retrospective studies of biopsy-proven NAFLD patients suggested that the Leu72Met (rs696217 G > T) polymorphism and the “GG” genotype of rs26802 variant in the ghrelin gene have a protective effect against NAFLD [63][64].

3.2. Resistin

As described above, the positive correlation between resistin and IR, steatosis, and inflammation is well stablished in murine and cellular models, but data in human NAFLD are conflicting. A systematic research of Bekaert et al. found 12 studies reporting resistin levels in biopsy-proven NAFLD patients, of which 6 reported statistically significant results [65]. Circulating resistin levels were positively correlated with hepatic steatosis, portal inflammation, and NAFLD ACTIVITY SCORES in non-diabetic NAFLD patients [66][67][68][69]. Of relevance, Aller et al. found that resistin association with the steatosis grade was lost when the homeostatic model assessment of insulin resistance (HOMA-IR) parameter was included in the multivariate logistic analysis, indicating that resistin is a surrogate marker of IR [67]. Supporting the relevance of resistin in NAFLD, a predicting diagnostic biomarker panel for histological NASH in obese subjects included the serum levels of resistin together with adiponectin and cytokeratin 18 (marker of cell death) [70]. However, resistin was not included in the predicting algorithm for NASH or NASH-related fibrosis in a more recent study by the same group [71]. In contrast, a study described a negative correlation between circulating resistin levels and the steatosis grade in severely obese NAFLD patients [72]. The remaining studies included in the cited meta-analysis did not demonstrate an association of resistin with liver histological parameters in obese and non-obese NAFLD patients [73][74][75][76]. It is worth mentioning that, in this meta-analysis, 4 out of the 12 studies did not adjust their results for confounding factors, and the potential association between resistin and IR was conflicting among the studies [65]. More recently, the determination of serum resistin levels in severe obese NAFLD patients found no correlation with steatohepatitis or fibrosis severity [77]. Similarly, resistin circulating levels did not associate with steatosis grade, NASH diagnosis, hepatic ballooning, or lobular inflammation grade, but they did correlate with fibrosis stage in obese NAFLD patients [78].
Although there are ambiguous data on circulating resistin levels in NAFLD patients, the existing results on its hepatic expression are more consistent. Augmented hepatic resistin mRNA levels were reported in patients with NASH compared to steatosis or control subjects and in steatosis patients compared to control individuals [79]. Moreover, a positive association between hepatic resistin mRNA levels and hepatic steatosis, inflammation, or fibrosis was reported in several studies [79][80]. Additionally, a positive correlation between hepatic resistin protein expression and NAS, aspartate aminotransferase (AST), alanine aminotransferase (ALT), BMI, glucose, insulin, HOMA-IR, gamma-glutamyl transferase (GGT), lactate dehydrogenase (LDH), TG, and glycated haemoglobin was verified in obese NAFLD patients [81]. The resistin mRNA expression in subcutaneous adipose tissue was also verified to be increased in non-obese NAFLD patients, but no correlation with liver histological parameters was observed [68]. Interestingly, the immuno–histological assessment of hepatic samples of NAFLD patients revealed a resistin distribution predominantly in perisinusoidal cells (Kupffer cells and HSCs) [79], histiocytes of inflammatory infiltrate, and histiocytes surrounding the hepatocytes with steatosis [81]. Finally, a significant association was observed between resistin rs1862513 polymorphism and NAFLD [82], which supports the significance of resistin in the development of NAFLD.

3.3. Retinol Binding Protein 4 (RBP4)

Several clinical studies showed that elevated circulating RBP4 levels were associated with insulin-resistance states, namely obesity and T2DM [32]. In addition, decreased levels of RBP4 were correlated with recovering insulin sensitivity after weight loss or lifestyle intervention in obese adult or children populations [83][84]. Given the close association of NAFLD pathogenesis and IR, NAFLD is assumed to be correlated with increased levels of serum RBP4; however, inconsistent findings were observed. In studies without histological confirmation, serum RBP4 levels seemed to be positively correlated with liver fat [85] and were found to be higher in NAFLD patients than controls, in adult and pediatric subjects [86][87]. Nevertheless, a systematic research reported that only three out of seven studies verified a positive correlation between serum RBP4 levels and liver histology among patients with biopsy-proven NAFLD [65]. Similarly, a meta-analysis research did not find any significant differences between NAFLD, NASH, or SS patients compared to controls, neither between NASH nor SS patients [88]. The researchers highlighted the heterogeneity across patient populations or the lack of adjustment for confounding factors in the analyzed research, which challenges comparisons between studies and limits the conclusions that can be drawn about the associations between adipokines levels and NAFLD. More recently, a 3-year follow-up study in a Chinese cohort of NAFLD patients diagnosed by abdominal ultrasonography verified that baseline serum RBP4 concentrations are positively associated with NAFLD development and inversely correlated with NAFLD regression [89]. Moreover, higher serum RBP4 levels were associated with an increased risk for prediabetes and metabolic syndrome in obese patients with NAFLD [90].

3.4. Visfatin

Several studies have evaluated the levels of visfatin in histologically confirmed NAFLD patients as well as the possible correlations with hepatic steatosis, inflammation, and fibrosis; but, current data are limited and inconclusive, as verified by two systematic research [65][91]. Most data reported similar serum visfatin levels in NAFLD [92], simple steatosis (SS) [93], or NASH patients [93][94] compared to control subjects, as well as in NASH compared to SS patients [93][95]. Likewise, similar hepatic visfatin expression was found in NASH and SS patients [96]. However, some studies also verified the augmented levels of serum visfatin in NAFLD compared to controls [97] or the decreased visfatin levels in NAFLD [98], SS, or NASH patients versus controls [70][95]. Of note, increased serum visfatin levels were associated with a reduced hepatic DNL index in women with ultrasound-diagnosed NAFLD, while in men it was correlated with augmented hepatic fat but not with DNL index, which suggests a sex-dependent interpretation for the serum visfatin levels in NAFLD prognosis [99].
Concerning histological parameters, most data did not report any correlation between serum visfatin and hepatic steatosis, inflammation, or fibrosis [92][93]. However, Aller et al. reported that circulating visfatin levels may predict portal inflammation, but not steatosis or fibrosis, in non-diabetic obese NAFLD patients [100]. In addition, Kukla et al. found a positive correlation between hepatic visfatin expression and the fibrosis stage but not hepatic steatosis and inflammation in morbidly obese NAFLD patients [96], while Gaddipati et al. reported a positive correlation between visfatin expression in visceral adipose tissue and steatosis degree in non-diabetic NAFLD patients [98].
Interestingly, visfatin was recently proposed as a potential serum biomarker related to the degree of hepatic steatosis and fibrosis among pediatric obese patients diagnosed by non-invasive methods (abdominal ultrasound and transient elastography with liver stiffness and controlled attenuation parameter) [101]. Moreover, a 10-year follow-up study verified no association between serum visfatin levels and leukocyte infiltration in fatty liver at the baseline, but visfatin serum levels were significantly increased during the follow-up, likely due to the combined effects of augmented BMI and diabetes prevalence [102]. However, this had certain limitations, in particular, the use of ultrasonography as the NAFLD diagnosis method, without confirmation by hepatic biopsy, and the measurement of visfatin levels in all samples (basal and 10-years after) at the end of the study, which likely affects the visfatin concentrations due to different storage times.
Thus, the different methodological strategies used to study visfatin levels in human NAFLD likely determine the inconsistencies among the current data, and future research is still needed.

3.5. Chemerin

Since chemerin regulates insulin signaling and chronic inflammation, it is reasonable to hypothesize that this adipokine may be related to NAFLD development. In fact, elevated serum chemerin levels were identified as a risk factor for NAFLD development in T2DM patients [103] and were pointed out as a novel non-invasive serum marker predicting liver steatosis in obese children [104]. A recent meta-analysis [105] further explored the correlations between serum chemerin levels and NAFLD (steatosis and/or NASH) and its specific hepatic histologic lesions (liver steatosis, lobular and portal inflammation, and fibrosis). Overall, circulating chemerin levels were consistently higher in patients with NAFLD and steatosis compared to controls, although no significant difference was verified between NASH patients and controls. Moreover, data on serum chemerin levels and specific liver lesions are inconsistent, and no correlations were verified [65][105].
It was found that chemerin expression in visceral adipose tissue was negatively correlated with the steatosis score and NAFLD ACTIVITY SCORES of obese NAFLD patients, likely through the modulation of IR and, thus, NAFLD [106]. Data on hepatic chemerin mRNA expression are contradictory; its levels were found to be negatively associated with inflammation, fibrosis, and NAS, but not with steatosis, in non-obese NAFLD patients [107], while other studies verified an increased hepatic chemerin mRNA expression, as well as hepatic CMKLR1 expression, that correlated with hepatic steatosis, hepatocyte ballooning, and the NAFLD activity score in obese NAFLD patients [108][109]. Of note, the hepatic expression of both chemerin and CMKLR1 was associated with obesity [109], which can partially explain the inconsistency of the results.

3.6. Adipocyte Fatty Acid-Binding Protein (AFABP)

In ultrasound-diagnosed NAFLD patients, serum AFABP levels were higher in NAFLD patients compared with non-NAFLD group [110][111]. The same was observed in biopsy-proven NAFLD patients, where serum AFABP had an independent positive correlation with lobular inflammation and hepatocyte ballooning, even after adjusting for confounding factors [112][113]. Milner et al. also reported higher serum AFABP levels in NASH patients compared with SS and correlated AFABP with IR, adiposity, and the fibrosis stage [112]. Nevertheless, other studies did not verify an association between AFABP and fibrosis, or that this adipokine was able to distinguish NASH from non-NASH patients [94]. In summary, serum AFABP levels are elevated in NAFLD, but its correlation with NASH, and particular fibrosis, is still unclear.

References

  1. Leoni, S.; Tovoli, F.; Napoli, L.; Serio, I.; Ferri, S.; Bolondi, L. Current guidelines for the management of non-alcoholic fatty liver disease: A systematic review with comparative analysis. World J. Gastroenterol. 2018, 24, 3361–3373.
  2. Younossi, Z.; Anstee, Q.M.; Marietti, M.; Hardy, T.; Henry, L.; Eslam, M.; George, J.; Bugianesi, E. Global burden of NAFLD and NASH: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 11–20.
  3. Godoy-Matos, A.F.; Silva Júnior, W.S.; Valerio, C.M. NAFLD as a continuum: From obesity to metabolic syndrome and diabetes. Diabetol. Metab. Syndr. 2020, 12, 60.
  4. Eslam, M.; Newsome, P.N.; Sarin, S.K.; Anstee, Q.M.; Targher, G.; Romero-Gomez, M.; Zelber-Sagi, S.; Wai-Sun Wong, V.; Dufour, J.F.; Schattenberg, J.M.; et al. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J. Hepatol. 2020, 73, 202–209.
  5. Francisco, V.; Pino, J.; Campos-Cabaleiro, V.; Ruiz-Fernández, C.; Mera, A.; Gonzalez-Gay, M.A.; Gómez, R.; Gualillo, O. Obesity, fat mass and immune system: Role for leptin. Front. Physiol. 2018, 9, 640.
  6. Martínez-Uña, M.; López-Mancheño, Y.; Diéguez, C.; Fernández-Rojo, M.A.; Novelle, M.G. Unraveling the role of leptin in liver function and its relationship with liver diseases. Int. J. Mol. Sci. 2020, 21, 9368.
  7. Jiménez-Cortegana, C.; García-Galey, A.; Tami, M.; Del Pino, P.; Carmona, I.; López, S.; Alba, G.; Sánchez-Margalet, V. Role of leptin in non-alcoholic fatty liver disease. Biomedicines 2021, 9, 762.
  8. Nguyen, T.M.D. Adiponectin: Role in Physiology and Pathophysiology. Int. J. Prev. Med. 2020, 11, 136.
  9. Heydari, M.; Cornide-Petronio, M.E.; Jiménez-Castro, M.B.; Peralta, C. Data on adiponectin from 2010 to 2020: Therapeutic target and prognostic factor for liver diseases? Int. J. Mol. Sci. 2020, 21, 5242.
  10. Müller, T.D.; Nogueiras, R.; Andermann, M.L.; Andrews, Z.B.; Anker, S.D.; Argente, J.; Batterham, R.L.; Benoit, S.C.; Bowers, C.Y.; Broglio, F.; et al. Ghrelin. Mol. Metab. 2015, 4, 437–460.
  11. Quiñones, M.; Fernø, J.; Al-Massadi, O. Ghrelin and liver disease. Rev. Endocr. Metab. Disord. 2020, 21, 45–56.
  12. Micioni Di Bonaventura, E.; Botticelli, L.; Del Bello, F.; Giorgioni, G.; Piergentili, A.; Quaglia, W.; Cifani, C.; Micioni Di Bonaventura, M.V. Assessing the role of ghrelin and the enzyme ghrelin O-acyltransferase (GOAT) system in food reward, food motivation, and binge eating behavior. Pharmacol. Res. 2021, 172, 105847.
  13. Li, Y.; Hai, J.; Li, L.; Chen, X.; Peng, H.; Cao, M.; Zhang, Q. Administration of ghrelin improves inflammation, oxidative stress, and apoptosis during and after non-alcoholic fatty liver disease development. Endocrine 2013, 43, 376–386.
  14. Mao, Y.; Cheng, J.; Yu, F.; Li, H.; Guo, C.; Fan, X. Ghrelin Attenuated Lipotoxicity via Autophagy Induction and Nuclear Factor-κB Inhibition. Cell. Physiol. Biochem. 2015, 37, 563–576.
  15. Yin, Y.; Wang, Q.; Qi, M.; Zhang, C.; Li, Z.; Zhang, W. Ghrelin ameliorates nonalcoholic steatohepatitis induced by chronic low-grade inflammation via blockade of Kupffer cell M1 polarization. J. Cell. Physiol. 2021, 236, 5121–5133.
  16. Guillory, B.; Jawanmardi, N.; Iakova, P.; Anderson, B.; Zang, P.; Timchenko, N.A.; Garcia, J.M. Ghrelin deletion protects against age-associated hepatic steatosis by downregulating the C/EBPα-p300/DGAT1 pathway. Aging Cell 2018, 17, e12688.
  17. Ezquerro, S.; Becerril, S.; Tuero, C.; Méndez-Giménez, L.; Mocha, F.; Moncada, R.; Valentí, V.; Cienfuegos, J.A.; Catalán, V.; Gómez-Ambrosi, J.; et al. Role of ghrelin isoforms in the mitigation of hepatic inflammation, mitochondrial dysfunction, and endoplasmic reticulum stress after bariatric surgery in rats. Int. J. Obes. 2020, 44, 475–487.
  18. Ezquerro, S.; Méndez-Giménez, L.; Becerril, S.; Moncada, R.; Valentí, V.; Catalán, V.; Gómez-Ambrosi, J.; Frühbeck, G.; Rodríguez, A. Acylated and desacyl ghrelin are associated with hepatic lipogenesis, β-oxidation and autophagy: Role in NAFLD amelioration after sleeve gastrectomy in obese rats. Sci. Rep. 2016, 6, 39942.
  19. Dallak, M.A. Acylated ghrelin induces but deacylated ghrelin prevents hepatic steatosis and insulin resistance in lean rats: Effects on DAG/PKC/JNK pathway. Biomed. Pharmacother. 2018, 105, 299–311.
  20. Acquarone, E.; Monacelli, F.; Borghi, R.; Nencioni, A.; Odetti, P. Resistin: A reappraisal. Mech. Ageing Dev. 2019, 178, 46–63.
  21. Steppan, C.M.; Bailey, S.T.; Bhat, S.; Brown, E.J.; Banerjee, R.R.; Wright, C.M.; Patel, H.R.; Ahima, R.S.; Lazar, M.A. The hormone resistin links obesity to diabetes. Nature 2001, 409, 307–312.
  22. Muse, E.D.; Obici, S.; Bhanot, S.; Monia, B.P.; McKay, R.A.; Rajala, M.W.; Scherer, P.E.; Rossetti, L. Role of resistin in diet-induced hepatic insulin resistance. J. Clin. Investig. 2004, 114, 232–239.
  23. Wen, F.; Shi, Z.; Liu, X.; Tan, Y.; Wei, L.; Zhu, X.; Zhang, H.; Zhu, X.; Meng, X.; Ji, W.; et al. Acute Elevated Resistin Exacerbates Mitochondrial Damage and Aggravates Liver Steatosis through AMPK/PGC-1α Signaling Pathway in Male NAFLD Mice. Horm. Metab. Res. 2021, 53, 132–144.
  24. Steppan, C.M.; Wang, J.; Whiteman, E.L.; Birnbaum, M.J.; Lazar, M.A. Activation of SOCS-3 by resistin. Mol. Cell. Biol. 2005, 25, 1569–1575.
  25. Banerjee, R.R.; Rangwala, S.M.; Shapiro, J.S.; Rich, A.S.; Rhoades, B.; Qi, Y.; Wang, J.; Rajala, M.W.; Pocai, A.; Scherer, P.E.; et al. Regulation of fasted blood glucose by resistin. Science 2004, 303, 1195–1198.
  26. Singhal, N.S.; Patel, R.T.; Qi, Y.; Lee, Y.S.; Ahima, R.S. Loss of resistin ameliorates hyperlipidemia and hepatic steatosis in leptin-deficient mice. Am. J. Physiol. Endocrinol. Metab. 2008, 295, E331–E338.
  27. Song, R.; Wang, X.; Mao, Y.; Li, H.; Li, Z.; Xu, W.; Wang, R.; Guo, T.; Jin, L.; Zhang, X.; et al. Resistin disrupts glycogen synthesis under high insulin and high glucose levels by down-regulating the hepatic levels of GSK3β. Gene 2013, 529, 50–56.
  28. Garcia, C.C.; Piotrkowski, B.; Baz, P.; Poncino, D.; Benavides, J.; Colombato, L.; Toso, M.L.R.; Yantorno, S.; Descalzi, V.; Gondolesi, G.E.; et al. A Decreased Response to Resistin in Mononuclear Leukocytes Contributes to Oxidative Stress in Nonalcoholic Fatty Liver Disease. Dig. Dis. Sci. 2021, 1, 3006–3016.
  29. Beier, J.I.; Guo, L.; Von Montfort, C.; Kaiser, J.P.; Joshi-Barve, S.; Arteel, G.E. New role of resistin in lipopolysaccharide-induced liver damage in mice. J. Pharmacol. Exp. Ther. 2008, 325, 801–808.
  30. Saxena, N.K.; Anania, F.A. Adipocytokines and hepatic fibrosis. Trends Endocrinol. Metab. 2015, 26, 153–161.
  31. Dong, Z.X.; Su, L.; Brymora, J.; Bird, C.; Xie, Q.; George, J.; Wang, J.H. Resistin mediates the hepatic stellate cell phenotype. World J. Gastroenterol. 2013, 19, 4475–4485.
  32. Nono Nankam, P.A.; Blüher, M. Retinol-binding protein 4 in obesity and metabolic dysfunctions. Mol. Cell. Endocrinol. 2021, 531, 111312.
  33. Liu, Y.; Mu, D.; Chen, H.; Li, D.; Song, J.; Zhong, Y.; Xia, M. Retinol-Binding Protein 4 Induces Hepatic Mitochondrial Dysfunction and Promotes Hepatic Steatosis. J. Clin. Endocrinol. Metab. 2016, 101, 4338–4348.
  34. Saeed, A.; Bartuzi, P.; Heegsma, J.; Dekker, D.; Kloosterhuis, N.; de Bruin, A.; Jonker, J.W.; van de Sluis, B.; Faber, K.N. Impaired Hepatic Vitamin A Metabolism in NAFLD Mice Leading to Vitamin A Accumulation in Hepatocytes. Cell. Mol. Gastroenterol. Hepatol. 2021, 11, 309–325.e3.
  35. Lee, S.A.; Yuen, J.J.; Jiang, H.; Kahn, B.B.; Blaner, W.S. Adipocyte-specific overexpression of retinol-binding protein 4 causes hepatic steatosis in mice. Hepatology 2016, 64, 1534–1546.
  36. Cioffi, C.L.; Racz, B.; Varadi, A.; Freeman, E.E.; Conlon, M.P.; Chen, P.; Zhu, L.; Kitchen, D.B.; Barnes, K.D.; Martin, W.H.; et al. Design, Synthesis, and Preclinical Efficacy of Novel Nonretinoid Antagonists of Retinol-Binding Protein 4 in the Mouse Model of Hepatic Steatosis. J. Med. Chem. 2019, 62, 5470–5500.
  37. Carbone, F.; Liberale, L.; Bonaventura, A.; Vecchié, A.; Casula, M.; Cea, M.; Monacelli, F.; Caffa, I.; Bruzzone, S.; Montecucco, F.; et al. Regulation and Function of Extracellular Nicotinamide Phosphoribosyltransferase/Visfatin. Compr. Physiol. 2017, 7, 603–621.
  38. Revollo, J.R.; Körner, A.; Mills, K.F.; Satoh, A.; Wang, T.; Garten, A.; Dasgupta, B.; Sasaki, Y.; Wolberger, C.; Townsend, R.R.; et al. Nampt/PBEF/Visfatin regulates insulin secretion in beta cells as a systemic NAD biosynthetic enzyme. Cell Metab. 2007, 6, 363–375.
  39. Heo, Y.J.; Choi, S.E.; Lee, N.; Jeon, J.Y.; Han, S.J.; Kim, D.J.; Kang, Y.; Lee, K.W.; Kim, H.J. Visfatin exacerbates hepatic inflammation and fibrosis in a methionine-choline-deficient diet mouse model. J. Gastroenterol. Hepatol. 2021, 36, 2592–2600.
  40. Heo, Y.J.; Choi, S.E.; Jeon, J.Y.; Han, S.J.; Kim, D.J.; Kang, Y.; Lee, K.W.; Kim, H.J. Visfatin Induces Inflammation and Insulin Resistance via the NF- κ B and STAT3 Signaling Pathways in Hepatocytes. J. Diabetes Res. 2019, 2019, 4021623.
  41. Wang, L.F.; Wang, X.N.; Huang, C.C.; Hu, L.; Xiao, Y.F.; Guan, X.H.; Qian, Y.S.; Deng, K.Y.; Xin, H.B. Inhibition of NAMPT aggravates high fat diet-induced hepatic steatosis in mice through regulating Sirt1/AMPKα/SREBP1 signaling pathway. Lipids Health Dis. 2017, 16, 82.
  42. Ilbeigi, D.; Nourbakhsh, M.; Pasalar, P.; Meshkani, R.; Afra, H.S.; Panahi, G.; Borji, M.; Sharifi, R. Nicotinamide phosphoribosyltransferase knockdown leads to lipid accumulation in HepG2 cells through the SIRT1-AMPK pathway. Cell J. 2020, 22, 125–132.
  43. Dall, M.; Hassing, A.S.; Niu, L.; Nielsen, T.S.; Ingerslev, L.R.; Sulek, K.; Trammell, S.A.J.; Gillum, M.P.; Barrès, R.; Larsen, S.; et al. Hepatocyte-specific perturbation of NAD+ biosynthetic pathways in mice induces reversible nonalcoholic steatohepatitis-like phenotypes. J. Biol. Chem. 2021, 297, 101388.
  44. Helfer, G.; Wu, Q.F. Chemerin: A multifaceted adipokine involved in metabolic disorders. J. Endocrinol. 2018, 238, R79–R94.
  45. Bondue, B.; Wittamer, V.; Parmentier, M. Chemerin and its receptors in leukocyte trafficking, inflammation and metabolism. Cytokine Growth Factor Rev. 2011, 22, 331–338.
  46. Bozaoglu, K.; Bolton, K.; McMillan, J.; Zimmet, P.; Jowett, J.; Collier, G.; Walder, K.; Segal, D. Chemerin is a novel adipokine associated with obesity and metabolic syndrome. Endocrinology 2007, 148, 4687–4694.
  47. Sell, H.; Divoux, A.; Poitou, C.; Basdevant, A.; Bouillot, J.L.; Bedossa, P.; Tordjman, J.; Eckel, J.; Clément, K. Chemerin Correlates with Markers for Fatty Liver in Morbidly Obese Patients and Strongly Decreases after Weight Loss Induced by Bariatric Surgery. J. Clin. Endocrinol. Metab. 2010, 95, 2892–2896.
  48. An, X.; Liu, J.; Li, Y.; Dou, Z.; Li, N.; Suo, Y.; Ma, Y.; Sun, M.; Tian, Z.; Xu, L. Chemerin/CMKLR1 ameliorates nonalcoholic steatohepatitis by promoting autophagy and alleviating oxidative stress through the JAK2-STAT3 pathway. Peptides 2021, 135, 170422.
  49. Pohl, R.; Rein-Fischboeck, L.; Meier, E.M.; Eisinger, K.; Krautbauer, S.; Buechler, C. Resolvin E1 and chemerin C15 peptide do not improve rodent non-alcoholic steatohepatitis. Exp. Mol. Pathol. 2015, 98, 295–299.
  50. Gruben, N.; Vergara, M.A.; Kloosterhuis, N.J.; Van Der Molen, H.; Stoelwinder, S.; Youssef, S.; De Bruin, A.; Delsing, D.J.; Kuivenhoven, J.A.; Van De Sluis, B.; et al. Chemokine-like receptor 1 deficiency does not affect the development of insulin resistance and nonalcoholic fatty liver disease in mice. PLoS ONE 2014, 9, e96345.
  51. Krautbauer, S.; Wanninger, J.; Eisinger, K.; Hader, Y.; Beck, M.; Kopp, A.; Schmid, A.; Weiss, T.S.; Dorn, C.; Buechler, C. Chemerin is highly expressed in hepatocytes and is induced in non-alcoholic steatohepatitis liver. Exp. Mol. Pathol. 2013, 95, 199–205.
  52. Li, H.L.; Wu, X.; Xu, A.; Hoo, R.L.C. A-FABP in Metabolic Diseases and the Therapeutic Implications: An Update. Int. J. Mol. Sci. 2021, 22, 9386.
  53. Jin, R.; Hao, J.; Yi, Y.; Sauter, E.; Li, B. Regulation of macrophage functions by FABP-mediated inflammatory and metabolic pathways. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2021, 1866, 158964.
  54. Hoo, R.L.C.; Lee, I.P.C.; Zhou, M.; Wong, J.Y.L.; Hui, X.; Xu, A.; Lam, K.S.L. Pharmacological inhibition of adipocyte fatty acid binding protein alleviates both acute liver injury and non-alcoholic steatohepatitis in mice. J. Hepatol. 2013, 58, 358–364.
  55. Wu, X.; Shu, L.; Zhang, Z.; Li, J.; Zong, J.; Cheong, L.Y.; Ye, D.; Lam, K.S.L.; Song, E.; Wang, C.; et al. Adipocyte Fatty Acid Binding Protein Promotes the Onset and Progression of Liver Fibrosis via Mediating the Crosstalk between Liver Sinusoidal Endothelial Cells and Hepatic Stellate Cells. Adv. Sci. 2021, 8, 2003721.
  56. St-Pierre, D.H.; Karelis, A.D.; Coderre, L.; Malita, F.; Fontaine, J.; Mignault, D.; Brochu, M.; Bastard, J.P.; Cianflone, K.; Doucet, E.; et al. Association of acylated and nonacylated ghrelin with insulin sensitivity in overweight and obese postmenopausal women. J. Clin. Endocrinol. Metab. 2007, 92, 264–269.
  57. Barazzoni, R.; Zanetti, M.; Ferreira, C.; Vinci, P.; Pirulli, A.; Mucci, M.P.; Dore, F.; Fonda, M.; Ciocchi, B.; Cattin, L.; et al. Relationships between desacylated and acylated ghrelin and insulin sensitivity in the metabolic syndrome. J. Clin. Endocrinol. Metab. 2007, 92, 3935–3940.
  58. Pacifico, L.; Poggiogalle, E.; Costantino, F.; Anania, C.; Ferraro, F.; Chiarelli, F.; Chiesa, C. Acylated and nonacylated ghrelin levels and their associations with insulin resistance in obese and normal weight children with metabolic syndrome. Eur. J. Endocrinol. 2009, 161, 861–870.
  59. Arslan, N.; Sayin, O.; Tokgoz, Y. Evaluation of serum xenin and ghrelin levels and their relationship with nonalcoholic fatty liver disease and insulin resistance in obese adolescents. Acad. Psychiatry 2014, 37, 1091–1097.
  60. MacHado, M.V.; Coutinho, J.; Carepa, F.; Costa, A.; Proença, H.; Cortez-Pinto, H. How adiponectin, leptin, and ghrelin orchestrate together and correlate with the severity of nonalcoholic fatty liver disease. Eur. J. Gastroenterol. Hepatol. 2012, 24, 1166–1172.
  61. Marchesini, G.; Pagotto, U.; Bugianesi, E.; De Iasio, R.; Manini, R.; Vanni, E.; Pasquali, R.; Melchionda, N.; Rizzetto, M. Low Ghrelin Concentrations in Nonalcoholic Fatty Liver Disease Are Related to Insulin Resistance. J. Clin. Endocrinol. Metab. 2003, 88, 5674–5679.
  62. Estep, M.; Abawi, M.; Jarrar, M.; Wang, L.; Stepanova, M.; Elariny, H.; Moazez, A.; Goodman, Z.; Chandhoke, V.; Baranova, A.; et al. Association of obestatin, ghrelin, and inflammatory cytokines in obese patients with non-alcoholic fatty liver disease. Obes. Surg. 2011, 21, 1750–1757.
  63. Tabaeian, S.P.; Mahmoudi, T.; Sabzikarian, M.; Rezamand, G.; Dabiri, R.; Nobakht, H.; Asadi, A.; Farahani, H.; Mansour-Ghanaei, F.; Zali, M.R. The leu72met (Rs696217 g>t) polymorphism of the ghrelin gene might be a protective factor for nonalcoholic fatty liver disease. J. Gastrointest. Liver Dis. 2021, 30, 233–239.
  64. Rezamand, G.; Mahmoudi, T.; Tabaeian, S.P.; Farahani, H.; Shahinmehr, F.; Nobakht, H.; Dabiri, R.; Asadi, A.; Mansour-Ghanaei, F.; Zali, M.R. The “GG” genotype of rs26802 variant in the ghrelin gene is a potential protective factor against nonalcoholic fatty liver disease. Physiol. Int. 2021, 108, 342–352.
  65. Bekaert, M.; Verhelst, X.; Geerts, A.; Lapauw, B.; Calders, P. Association of recently described adipokines with liver histology in biopsy-proven non-alcoholic fatty liver disease: A systematic review. Obes. Rev. 2016, 17, 68–80.
  66. Cengiz, C.; Ardicoglu, Y.; Bulut, S.; Boyacioglu, S. Serum retinol-binding protein 4 in patients with nonalcoholic fatty liver disease: Does it have a significant impact on pathogenesis? Eur. J. Gastroenterol. Hepatol. 2010, 22, 813–819.
  67. Aller, R.; De Luis, D.A.; Fernandez, L.; Calle, F.; Velayos, B.; Olcoz, J.L.; Izaola, O.; Sagrado, M.G.; Conde, R.; Gonzalez, J.M. Influence of insulin resistance and adipokines in the grade of steatosis of nonalcoholic fatty liver disease. Dig. Dis. Sci. 2008, 53, 1088–1092.
  68. Pagano, C.; Soardo, G.; Pilon, C.; Milocco, C.; Basan, L.; Milan, G.; Donnini, D.; Faggian, D.; Mussap, M.; Plebani, M.; et al. Increased serum resistin in nonalcoholic fatty liver disease is related to liver disease severity and not to insulin resistance. J. Clin. Endocrinol. Metab. 2006, 91, 1081–1086.
  69. Şenateş, E.; Çolak, Y.; Yeşil, A.; Coşkunpinar, E.; Şahin, Ö.; Kahraman, Ö.T.; Erkalma Şenateş, B.; Tuncer, I. Circulating resistin is elevated in patients with non-alcoholic fatty liver disease and is associated with steatosis, portal inflammation, insulin resistance and nonalcoholic steatohepatitis scores. Minerva Med. 2012, 103, 369–376.
  70. Younossi, Z.M.; Jarrar, M.; Nugent, C.; Randhawa, M.; Afendy, M.; Stepanova, M.; Rafiq, N.; Goodman, Z.; Chandhoke, V.; Baranova, A. A novel diagnostic biomarker panel for obesity-related nonalcoholic steatohepatitis (NASH). Obes. Surg. 2008, 18, 1430–1437.
  71. Younossi, Z.M.; Page, S.; Rafiq, N.; Birerdinc, A.; Stepanova, M.; Hossain, N.; Afendy, A.; Younoszai, Z.; Goodman, Z.; Baranova, A. A biomarker panel for non-alcoholic steatohepatitis (NASH) and NASH-related fibrosis. Obes. Surg. 2011, 21, 431–439.
  72. Argentou, M.; Tiniakos, D.G.; Karanikolas, M.; Melachrinou, M.; Makri, M.G.; Kittas, C.; Kalfarentzos, F. Adipokine serum levels are related to liver histology in severely obese patients undergoing bariatric surgery. Obes. Surg. 2009, 19, 1313–1323.
  73. Musso, G.; Gambino, R.; Durazzo, M.; Biroli, G.; Carello, M.; Fagà, E.; Pacini, G.; De Michieli, F.; Rabbione, L.; Premoli, A.; et al. Adipokines in NASH: Postprandial lipid metabolism as a link between adiponectin and liver disease. Hepatology 2005, 42, 1175–1183.
  74. Jarrar, M.H.; Baranova, A.; Collantes, R.; Ranard, B.; Stepanova, M.; Bennett, C.; Fang, Y.; Elariny, H.; Goodman, Z.; Chandhoke, V.; et al. Adipokines and cytokines in non-alcoholic fatty liver disease. Aliment. Pharmacol. Ther. 2008, 27, 412–421.
  75. Wong, V.W.S.; Hui, A.Y.; Tsang, S.W.C.; Chan, J.L.Y.; Tse, A.M.L.; Chan, K.F.; So, W.Y.; Cheng, A.Y.S.; Ng, W.F.; Wong, G.L.H.; et al. Metabolic and adipokine profile of Chinese patients with nonalcoholic fatty liver disease. Clin. Gastroenterol. Hepatol. 2006, 4, 1154–1161.
  76. Koehler, E.; Swain, J.; Sanderson, S.; Krishnan, A.; Watt, K.; Charlton, M. Growth hormone, dehydroepiandrosterone and adiponectin levels in non-alcoholic steatohepatitis: An endocrine signature for advanced fibrosis in obese patients. Liver Int. 2012, 32, 279–286.
  77. Baltieri, L.; Chaim, E.A.; Chaim, F.D.M.; Utrini, M.P.; Gestic, M.A.; Cazzo, E. Correlation between nonalcoholic fatty liver disease features and levels of adipokines and inflammatory cytokines among morbidly obese individuals. Arq. Gastroenterol. 2018, 55, 247–251.
  78. Ajmera, V.; Perito, E.R.; Bass, N.M.; Terrault, N.A.; Yates, K.P.; Gill, R.; Loomba, R.; Diehl, A.M.; Aouizerat, B.E. Novel plasma biomarkers associated with liver disease severity in adults with nonalcoholic fatty liver disease. Hepatology 2017, 65, 65–77.
  79. Shen, C.; Zhao, C.Y.; Wang, W.; Wang, Y.D.; Sun, H.; Cao, W.; Yu, W.Y.; Zhang, L.; Ji, R.; Li, M.; et al. The relationship between hepatic resistin overexpression and inflammation in patients with nonalcoholic steatohepatitis. BMC Gastroenterol. 2014, 14, 39.
  80. Nobili, V.; Carpino, G.; Alisi, A.; Franchitto, A.; Alpini, G.; De Vito, R.; Onori, P.; Alvaro, D.; Gaudio, E. Hepatic progenitor cells activation, fibrosis, and adipokines production in pediatric nonalcoholic fatty liver disease. Hepatology 2012, 56, 2142–2153.
  81. Gierej, P.; Gierej, B.; Kalinowski, P.; Wróblewski, T.; Paluszkiewicz, R.; Kobryń, K.; Ziarkiewicz-Wróblewska, B. Expression of resistin in the liver of patients with Non-Alcoholic fatty liver disease. Pol. J. Pathol. 2017, 68, 225–233.
  82. Kohan, L.; Safarpur, M.; Abdollahi, H. Omentin-1 rs2274907 and resistin rs1862513 polymorphisms influence genetic susceptibility to nonalcoholic fatty liver disease. Mol. Biol. Res. Commun. 2016, 5, 11.
  83. Balagopal, P.; Graham, T.E.; Kahn, B.B.; Altomare, A.; Funanage, V.; George, D. Reduction of elevated serum retinol binding protein in obese children by lifestyle intervention: Association with subclinical inflammation. J. Clin. Endocrinol. Metab. 2007, 92, 1971–1974.
  84. Haider, D.G.; Schindler, K.; Prager, G.; Bohdjalian, A.; Luger, A.; Wolzt, M.; Ludvik, B. Serum retinol-binding protein 4 is reduced after weight loss in morbidly obese subjects. J. Clin. Endocrinol. Metab. 2007, 92, 1168–1171.
  85. Stefan, N.; Hennige, A.M.; Staiger, H.; Machann, J.; Schick, F.; Schleicher, E.; Fritsche, A.; Häring, H.U. High circulating retinol-binding protein 4 is associated with elevated liver fat but not with total, subcutaneous, visceral, or intramyocellular fat in humans. Diabetes Care 2007, 30, 1173–1178.
  86. Seo, J.A.; Kim, N.H.; Park, S.Y.; Kim, H.Y.; Ryu, O.H.; Lee, K.W.; Lee, J.; Kim, D.L.; Choi, K.M.; Baik, S.H.; et al. Serum retinol-binding protein 4 levels are elevated in non-alcoholic fatty liver disease. Clin. Endocrinol. 2008, 68, 555–560.
  87. Huang, S.C.; Yang, Y.J. Serum retinol-binding protein 4 is independently associated with pediatric NAFLD and fasting triglyceride level. J. Pediatr. Gastroenterol. Nutr. 2013, 56, 145–150.
  88. Zhou, Z.; Chen, H.; Ju, H.; Sun, M. Circulating retinol binding protein 4 levels in nonalcoholic fatty liver disease: A systematic review and meta-analysis. Lipids Health Dis. 2017, 16, 180.
  89. Wang, X.; Chen, X.; Zhang, H.; Pang, J.; Lin, J.; Xu, X.; Yang, L.; Ma, J.; Ling, W.; Chen, Y. Circulating retinol-binding protein 4 is associated with the development and regression of non-alcoholic fatty liver disease. Diabetes Metab. 2020, 46, 119–128.
  90. Karamfilova, V.; Gateva, A.; Alexiev, A.; Zheleva, N.; Velikova, T.; Ivanova-Boyanova, R.; Ivanova, R.; Cherkezov, N.; Kamenov, Z.; Mateva, L. The association between retinol-binding protein 4 and prediabetes in obese patients with nonalcoholic fatty liver disease. Arch. Physiol. Biochem. 2022, 128, 217–222.
  91. Ismaiel, A.; Leucuta, D.C.; Popa, S.L.; Dumitrascu, D.L. Serum visfatin levels in nonalcoholic fatty liver disease and liver fibrosis: Systematic review and meta-analysis. J. Clin. Med. 2021, 10, 3029.
  92. Genc, H.; Dogru, T.; Kara, M.; Tapan, S.; Ercin, C.N.; Acikel, C.; Karslioglu, Y.; Bagci, S. Association of plasma visfatin with hepatic and systemic inflammation in nonalcoholic fatty liver disease. Ann. Hepatol. 2013, 12, 380–387.
  93. Polyzos, S.A.; Kountouras, J.; Papatheodorou, A.; Katsiki, E.; Patsiaoura, K.; Zafeiriadou, E.; Zavos, C.; Papadopoulou, E.; Terpos, E. Adipocytokines and cytokeratin-18 in patients with nonalcoholic fatty liver disease: Introduction of CHA index. Ann. Hepatol. 2013, 12, 749–757.
  94. Yoon, M.Y.; Sung, J.M.; Song, C.S.; Lee, W.Y.; Rhee, E.J.; Shin, J.H.; Yoo, C.H.; Chae, S.W.; Kim, J.Y.; Jin, W.; et al. Enhanced A-FABP expression in visceral fat: Potential contributor to the progression of NASH. Clin. Mol. Hepatol. 2012, 18, 279–286.
  95. Dahl, T.B.; Haukeland, J.W.; Yndestad, A.; Ranheim, T.; Gladhaug, I.P.; Damås, J.K.; Haaland, T.; Løberg, E.M.; Arntsen, B.; Birkeland, K.; et al. Intracellular Nicotinamide Phosphoribosyltransferase Protects against Hepatocyte Apoptosis and Is Down-Regulated in Nonalcoholic Fatty Liver Disease. J. Clin. Endocrinol. Metab. 2010, 95, 3039–3047.
  96. Kukla, M.; Ciupińska-Kajor, M.; Kajor, M.; Wylezoł, M.; Zwirska-Korczala, K.; Hartleb, M.; Berdowska, A.; Mazur, W. Liver visfatin expression in morbidly obese patients with nonalcoholic fatty liver disease undergoing bariatric surgery. Polish J. Pathol. 2010, 61, 147–153.
  97. Auguet, T.; Terra, X.; Porras, J.A.; Orellana-Gavaldà, J.M.; Martinez, S.; Aguilar, C.; Lucas, A.; Pellitero, S.; Hernández, M.; Del Castillo, D.; et al. Plasma visfatin levels and gene expression in morbidly obese women with associated fatty liver disease. Clin. Biochem. 2013, 46, 202–208.
  98. Gaddipati, R.; Mitnala, S.; Padaki, N.; Mukherjee, R.M.; Sekaran, A.; Jayaraj-Mansard, M.; Rabella, P.; Rao-Guduru, V.; Reddy-Duwuru, N. Visceral adipose tissue visfatin in nonalcoholic fatty liver disease. Ann. Hepatol. 2010, 9, 266–270.
  99. Amirkalali, B.; Sohrabi, M.R.; Esrafily, A.; Jalali, M.; Gholami, A.; Hosseinzadeh, P.; Keyvani, H.; Shidfar, F.; Zamani, F. Association between Nicotinamide Phosphoribosyltransferase and de novo Lipogenesis in Nonalcoholic Fatty Liver Disease. Med. Princ. Pract. 2017, 26, 251–257.
  100. Aller, R.; De Luis, D.A.; Izaola, O.; Sagrado, M.G.; Conde, R.; Velasco, M.C.; Alvarez, T.; Pacheco, D.; González, J.M. Influence of visfatin on histopathological changes of non-alcoholic fatty liver disease. Dig. Dis. Sci. 2009, 54, 1772–1777.
  101. Elkabany, Z.A.; Hamza, R.T.; Ismail, E.A.R.; Elsharkawy, A.; Yosry, A.; Musa, S.; Khalaf, M.A.; Elgawesh, R.M.; Esmat, G. Serum visfatin level as a noninvasive marker for nonalcoholic fatty liver disease in children and adolescents with obesity: Relation to transient elastography with controlled attenuation parameter. Eur. J. Gastroenterol. Hepatol. 2020, 32, 1008–1016.
  102. Johannsen, K.; Flechtner-Mors, M.; Kratzer, W.; Koenig, W.; Boehm, B.O.; Schmidberger, J. Association Between Visfatin and Hepatic Steatosis in the General Population During Long-Term Follow-Up. Horm. Metab. Res. 2019, 51, 602–607.
  103. Zhang, Z.; Wang, J.; Wang, H. Correlation of blood glucose, serum chemerin and insulin resistance with NAFLD in patients with type 2 diabetes mellitus. Exp. Ther. Med. 2018, 15, 2936.
  104. Kłusek-Oksiuta, M.; Bialokoz-Kalinowska, I.; Tarasów, E.; Wojtkowska, M.; Werpachowska, I.; Lebensztejn, D.M. Chemerin as a novel non-invasive serum marker of intrahepatic lipid content in obese children. Ital. J. Pediatr. 2014, 40, 84.
  105. Ren, Q.; Wang, H.; Zeng, Y.; Fang, X.; Wang, M.; Li, D.; Huang, W.; Xu, Y. Circulating chemerin levels in metabolic-associated fatty liver disease: A systematic review and meta-analysis. Lipids Health Dis. 2022, 21, 27.
  106. Bekaert, M.; Ouwens, D.M.; Hörbelt, T.; Van de Velde, F.; Fahlbusch, P.; Herzfeld de Wiza, D.; Van Nieuwenhove, Y.; Calders, P.; Praet, M.; Hoorens, A.; et al. Reduced expression of chemerin in visceral adipose tissue associates with hepatic steatosis in patients with obesity. Obesity 2016, 24, 2544–2552.
  107. Pohl, R.; Haberl, E.M.; Rein-Fischboeck, L.; Zimny, S.; Neumann, M.; Aslanidis, C.; Schacherer, D.; Krautbauer, S.; Eisinger, K.; Weiss, T.S.; et al. Hepatic chemerin mRNA expression is reduced in human nonalcoholic steatohepatitis. Eur. J. Clin. Investig. 2017, 47, 7–18.
  108. Kajor, M.; Kukla, M.; Waluga, M.; Liszka, Ł.; Dyaczyński, M.; Kowalski, G.; Żądło, D.; Berdowska, A.; Chapuła, M.; Kostrząb-Zdebel, A.; et al. Hepatic chemerin mRNA in morbidly obese patients with nonalcoholic fatty liver disease. Pol. J. Pathol. 2017, 68, 117–127.
  109. Döcke, S.; Lock, J.F.; Birkenfeld, A.L.; Hoppe, S.; Lieske, S.; Rieger, A.; Raschzok, N.; Sauer, I.M.; Florian, S.; Osterhoff, M.A.; et al. Elevated hepatic chemerin mRNA expression in human non-alcoholic fatty liver disease. Eur. J. Endocrinol. 2013, 169, 547–557.
  110. Suh, J.B.; Kim, S.M.; Cho, G.J.; Choi, K.M. Serum AFBP levels are elevated in patients with nonalcoholic fatty liver disease. Scand. J. Gastroenterol. 2014, 49, 979–985.
  111. Xu, Y.; Ma, X.; Pan, X.; He, X.; Wang, Y.; Bao, Y. Serum adipocyte fatty acid-binding protein levels: An indicator of non-alcoholic fatty liver disease in Chinese individuals. Liver Int. 2019, 39, 568–574.
  112. Milner, K.L.; van der Poorten, D.; Xu, A.; Bugianesi, E.; Kench, J.G.; Lam, K.S.L.; Chisholm, D.J.; George, J. Adipocyte fatty acid binding protein levels relate to inflammation and fibrosis in nonalcoholic fatty liver disease. Hepatology 2009, 49, 1926–1934.
  113. Shen, J.; Chan, H.L.Y.; Wong, G.L.H.; Choi, P.C.L.; Chan, A.W.H.; Chan, H.Y.; Chim, A.M.L.; Yeung, D.K.W.; Chan, F.K.L.; Woo, J.; et al. Non-invasive diagnosis of non-alcoholic steatohepatitis by combined serum biomarkers. J. Hepatol. 2012, 56, 1363–1370.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , ,
View Times: 358
Revisions: 3 times (View History)
Update Date: 16 Sep 2022
1000/1000