Next Article in Journal
Vertebral Bone Marrow-Derived Mesenchymal Stromal Cells from Osteoporotic and Healthy Patients Possess Similar Differentiation Properties In Vitro
Next Article in Special Issue
Adipokines in the Skin and in Dermatological Diseases
Previous Article in Journal
The Potential Anticancer Activity of Phytoconstituents against Gastric Cancer—A Review on In Vitro, In Vivo, and Clinical Studies
Previous Article in Special Issue
Downregulation of CTRP-3 by Weight Loss In Vivo and by Bile Acids and Incretins in Adipocytes In Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Roles of Adipokines in Digestive Diseases: Markers of Inflammation, Metabolic Alteration and Disease Progression

1
Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
2
Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
3
Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
4
Liver Center, Cathay General Hospital Medical Center, Taipei 10630, Taiwan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(21), 8308; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21218308
Submission received: 13 October 2020 / Revised: 30 October 2020 / Accepted: 1 November 2020 / Published: 5 November 2020
(This article belongs to the Special Issue Adipokines 3.0)

Abstract

:
Adipose tissue is a highly dynamic endocrine tissue and constitutes a central node in the interorgan crosstalk network through adipokines, which cause pleiotropic effects, including the modulation of angiogenesis, metabolism, and inflammation. Specifically, digestive cancers grow anatomically near adipose tissue. During their interaction with cancer cells, adipocytes are reprogrammed into cancer-associated adipocytes and secrete adipokines to affect tumor cells. Moreover, the liver is the central metabolic hub. Adipose tissue and the liver cooperatively regulate whole-body energy homeostasis via adipokines. Obesity, the excessive accumulation of adipose tissue due to hyperplasia and hypertrophy, is currently considered a global epidemic and is related to low-grade systemic inflammation characterized by altered adipokine regulation. Obesity-related digestive diseases, including gastroesophageal reflux disease, Barrett’s esophagus, esophageal cancer, colon polyps and cancer, non-alcoholic fatty liver disease, viral hepatitis-related diseases, cholelithiasis, gallbladder cancer, cholangiocarcinoma, pancreatic cancer, and diabetes, might cause specific alterations in adipokine profiles. These patterns and associated bases potentially contribute to the identification of prognostic biomarkers and therapeutic approaches for the associated digestive diseases. This review highlights important findings about altered adipokine profiles relevant to digestive diseases, including hepatic, pancreatic, gastrointestinal, and biliary tract diseases, with a perspective on clinical implications and mechanistic explorations.

1. Introduction

Adipose tissue is recognized as a highly dynamic endocrine tissue exhibiting extensive physiological functions [1] and is composed of mature adipocytes and a stromal vascular fraction, where adipose-derived stem cells, blood cells, fibroblasts, and nerves reside [2]. Adipose tissue constitutes a central node in the interorgan crosstalk network and mediates the regulation of multiple organs and tissues through adipokines [3] (also called adipocytokines), biologically active molecules causing pleiotropic effects, including modulation of angiogenesis, metabolism, and inflammation [4]. The emerging functional characterization of adipokines suggests a close link between the endocrine and immune systems of adipose tissue. This link is emphasized by the altered expression pattern of adipokines in adipose tissue adjacent to sites of inflammation [5]. Obesity, the excessive accumulation of adipose tissue due to hyperplasia and hypertrophy [6], is currently considered a global epidemicand is related to low-grade systemic inflammation. This state of inflammation is characterized by alterations in adipokine regulation [7]. Interestingly, digestive cancers such as gastric and colon cancers grow anatomically near adipose tissue. During their interaction with cancer cells, adipocytes dedifferentiate into preadipocytes or are reprogrammed into cancer-associated adipocytes, which secrete adipokines to stimulate the adhesion, migration, and invasion of tumor cells [8]. In particular, the liver is the central metabolic hub for carbohydrate, lipid, and protein metabolism [9]. Adipose tissue and the liver play important roles in the regulation of whole-body energy homeostasis, and prolonged metabolic stress leads to adipose tissue dysfunction, inflammation, and adipokine release, causing increased lipid flux to the liver, resulting in fatty liver [10]. Moreover, adipokines are involved in modulating insulin resistance, which is at the heart of obesity-related digestive diseases [11], including gastroesophageal reflux disease (GERD), Barrett’s esophagus, esophageal cancer, colon polyps and cancer, non-alcoholic fatty liver disease (NAFLD), viral hepatitis, cholelithiasis, gallbladder cancer, cholangiocarcinoma, pancreatic cancer, and diabetes [12,13] (Figure 1).
However, whether dysregulated adipokines are merely the consequence of digestive disease or whether these altered adipokines promote disease progression is unknown, and the roles of adipokines in digestive diseases remain to be investigated.
Leptin was the first adipokine to be discovered in 1994 [14], and hundreds of adipokines have since been discovered [15]. For example, adiponectin is an anti-inflammatory and insulin-sensitizing adipokine and is secreted mainly by white adipose tissue; however, adiponectin is decreased in obesity [16], and low serum adiponectin is associated with many cancers and inflammatory diseases, such as colon cancer and colitis [17]. The specific alteration patterns of adipokine expression and the associated basis in the development of various digestive diseases might contribute to the identification of prognostic biomarkers as well as therapeutic and preventative approaches for the associated diseases. The current review thus systematically highlights important findings about altered adipokine profiles in the context of diseases of the digestive tract, including the liver, pancreas, esophagus, stomach, small intestine, and colon, with a perspective on the clinical implications and associated mechanistic approaches.

2. Adipokines and the Liver

2.1. NAFLD

NAFLD, characterized by the accumulation of fat in the liver [18] due to non-alcoholic causes, is the liver manifestation of metabolic syndrome and includes the spectrum of hepatic steatosis and non-alcoholic steatohepatitis (NASH) [19]. Systemic insulin resistance is a major driver of hepatic steatosis in NAFLD, while lipotoxicity of accumulated lipids along with activation of the innate immune system are major drivers of NASH [18]. Thus, many adipokines have evolved as crucial signals in NAFLD.

2.1.1. Leptin

Increased leptin levels act as a pro-inflammatory stimulus [20], and leptin increases susceptibility to hepatotoxicity by regulating cytokine production and T cell activation [21]. On the other hand, leptin augments the oxidation of fatty acids in the liver by activating peroxisome proliferator-activated receptor-alpha (PPAR-α) [22]. Higher levels of circulating leptin were found to be associated with increased severity of NAFLD [23]. Moreover, polymorphisms in the leptin receptor (ObR) gene have been reported to be related to NAFLD [24]. However, in contrast to patients with obesity-associated NAFLD, patients with lipodystrophy have low levels of adipokines, including leptin [25], and leptin therapy thus appears to be highly effective for NASH in hypoleptinemic lipodystrophic patients [26].

2.1.2. Adiponectin

Adiponectin enhances glucose and fatty acid oxidation, improves insulin sensitivity, attenuates plaque formation, and increases aldosterone production [27]. The hepatoprotective effects of adiponectin, including its antisteatotic, anti-inflammatory, and antifibrogenic effects, have been widely investigated. Adiponectin levels are reduced in individuals with NAFLD [28] and are inversely related to the severity of steatosis, necroinflammation [29], and fibrosis [28]. Hypoadiponectinemia may play an important pathophysiological role in the progression from non-alcoholic fatty liver to NASH [30]. The adiponectin signaling pathway in the liver acts through T-cadherin, adiponectin receptor 1 (AdipoR1), AdipoR2, AMP-activated protein kinase (AMPK), ceramidase activity, and an adaptor protein, phosphotyrosine interacting with a PH domain and leucine zipper 1, and the recently discovered suppressor of glucose from autophagy [31]. AdipoR1 is expressed abundantly in muscle, whereas AdipoR2 is predominantly expressed in the liver [32]. NAFLD is associated with decreased hepatic expression of the two adiponectin receptors (AdipoR1 and 2), thereby contributing to a state of hepatic adiponectin resistance [33]. Comprehensive crosstalk between adiponectin and its cognate receptors, specifically AdipoR2, in the liver attenuates hepatic lipoinflammation by interacting with hepatic PPARs [34]. In addition, adiponectin protects hepatocytes from tumor necrosis factor-alpha (TNF-α)-induced death [35]; specifically, adiponectin is a potent TNF-α-neutralizing adipokine [36]. Moreover, bile acid (BA) synthesis and serum BA levels are directly correlated with disease severity in NAFLD, while the adiponectin level is inversely correlated with this parameter [37]. Furthermore, the single-nucleotide polymorphism (SNP) rs1501299 in the adiponectin gene might be related to increased NAFLD susceptibility [38].

2.1.3. Other Adipokines

Interestingly, our previous case-control study showed that plasminogen activator inhibitor-1 (PAI-1) is independently associated with NAFLD after adjustment for leptin and adiponectin levels [39]. However, data regarding other adipokines, including resistin (RETN), visfatin (i.e., extracellular nicotinamide phosphoribosyltransferase (eNAMPT)), retinol-binding protein-4 (RBP-4), chemerin, adipsin, obestatin, omentin, and vaspin, in NAFLD are inconclusive or limited [40].

2.2. Viral Hepatitis

2.2.1. Hepatitis B

Chronic hepatitis B virus (HBV) infection represents a major global health issue, affecting an estimated 257–291 million persons worldwide and is associated with substantial morbidity and mortality because of complications, including hepatitis, cirrhosis, and hepatocellular carcinoma (HCC) [41]. HBV is a hepatotropic, noncytopathic member of the hepadnaviridaefamily, comprising a 3.2 kb partly double-stranded, relaxed circular DNA genome and viral DNA polymerase condensed into a nucleocapsid by hepatitis B core proteins. There are now known to be at least tengenotypes of HBV [42]. Current therapies for chronic hepatitis B (CHB) remain limited to pegylated-interferon-alpha (PegIFN-α), or any of the fiveapproved nucleos(t)ide analog (Nuc) treatments. While viral suppression can be achieved in the majority of patients with the high-barrier-to-resistance new-generation of Nuc, HBsAg loss is achieved by PegIFN-α and/or Nuc in only 10% of patients after a 5-year follow-up [43].

Leptin

Leptin levels may be related to fibrosis progression in nondiabetic patients with chronic HBV infection [44]. Consistent with this possibility, cirrhosis due to CHB is associated with high leptin levels, which constitute a negative prognostic factor for the response to lamivudine monotherapy in patients with CHB [45]. Additionally, increased baseline leptin levels were noted in CHB patients compared to controls, and leptin levels decreased during IFN-α treatment [46]. However, decreased serum leptin levels were ever found in patients with HBV-related cirrhosis and HCC [47].

Adiponectin

In HepG2-HBV-stable cells, HBV replication was found to be upregulated by adiponectin and downregulated by adiponectin-targeting small interfering RNAs [48]. Consistent with this finding, individuals with chronic HBV infection have high serum adiponectin levels. Particularly in overweight and obese HBV-infected patients, a high HBV load was found to be positively associated with serum adiponectin levels [49]. Intriguingly, in HBV-infected male subjects without diabetes, the serum HBV DNA level correlated inversely with the serum high-density lipoprotein cholesterol level, and patients with detectable HBV DNA had lower adiponectin levels than those without [50]. Regarding hepatic inflammation, alanine aminotransferase (ALT) levels were found to be inversely related to adiponectin levels, independent of metabolic factors and HBV status [51], andadiponectin levels tended to decrease in HBV responders following IFN-α therapy [52]. On the other hand, adiponectin levels were associated with an increased risk of HCC in HBV patients. Over time, participants with higher adiponectin levels were less likely to achieve seroclearance of HBV surface antigen (HBsAg) and more likely to have persistently higher HBV DNA levels. Eventually, they were also more likely to develop cirrhosis [53].

Resistin

HBV-infected patients were found to show increased levels of serum resistin, and high serum resistin levels were associated with intrahepatic inflammation and necrosis [54]. In addition, resistin levels decreased in HBV-infected patients after antiviral therapy, especially in the subgroup of responders [55].

Visfatin

Visfatin concentrations were found to be elevated and negatively correlated with haptoglobin and fibrinogen levels in patients with chronic HBV infection [56].

Chemerin

Although chemerin is protective in experimental models of HCC, chemerin was reported to be induced in tumor tissues of patients with HBV-related HCC [57].

Multiple Adipokines

CHB patients were found to have higher serum adiponectin and visfatin levels but lower leptin levels than healthy controls. Moreover, serum leptin, adiponectin, and visfatin levels were correlated with HBV viremia, HBsAg levels, and liver fibrosis stage [58].

2.2.2. Hepatitis C

Hepatitis C virus (HCV), a human pathogen responsible for acute and chronic liver disease, has variants classified into eight genotypes [59] and chronically infects an estimated 71.1 million individuals worldwide [60]. HCV is currently thought to cause metabolic alterations in addition to a simple hepatic viral infection, as it affects insulin signaling, and much of its life cycle is closely associated with lipid metabolism [61]. Because both HCV infection and alterations in adipokines are critical in metabolism, their potential relationship has attracted attention [62]. There are genotype-specific impacts on HCV-associated metabolic alterations [61]. The combination of PegIFN and ribavirin provided a “cure” for a considerable proportion of patients with HCV infection, particularly those with the favorable IFN λ3 (IFNL3) genotype [63]. These cure rates were further improved by replacing IFN-based therapy with potent direct-acting antiviral agents (DAAs) [64]. Thus, some cross-sectional studies, as well as many longitudinal studies of HCV-infected patients receiving IFN-based or DAA therapy, have provided a landscape in which to study metabolic alterations and the associated effects of HCV clearance by comparing adipokine profiles before and after anti-HCV treatment.

Leptin

In cross-sectional studies, increased [65,66] or unchanged [67,68] serum leptin levels in patients with chronic HCV infection compared with controls have been noted. Regarding genotype-specific characteristics, the connection between steatosis and leptin in patients infected with genotype (G) 1 or G2 HCV [69,70] has been reported. In addition, high baseline leptin levels have been reported to be negative predictors of a sustained virologic response (SVR) to IFN-based therapy [71,72]. Moreover, leptin levels were found to remain unchanged after IFN therapy in patients with chronic HCV infection who achieved SVRs; leptin and complement component 3 (C3) may maintain immune and metabolic homeostasis through association with C4 and total cholesterol [73].

Adiponectin

Increased adiponectin levels were noted in HCV-infected patients [74,75,76,77], especially those with severe fibrosis [78], compared with controls, suggesting a pattern of adiponectin resistance [67,76], although one study found similar adiponectin levels between HCV-infected patients and controls [67]. Studies involving various HCV genotypes have reported diverse findings regarding adiponectin alteration and its correlation with HCV viral load or disease progression. In cross-sectional studies, patients with G3 HCV infection were found to have lower adiponectin levels than patients infected with other genotypes of HCV [79]. High viral load and G2 HCV infection were found to be associated with low serum adiponectin levels [80], and adiponectin levels were found to increase with the progression of hepatic fibrosis but were not related to viral load in patients with G4 HCV infection [81]. In patients with G1 or G3 HCV infection, adiponectin levels were found to be linked with steatosis only in males and to increase with hepatic inflammation [82]. In addition, insulin resistance was found to be associated with a decrease in adiponectin levels in G3 HCV-infected patients but not in G1 HCV-infected patients [83]. However, adiponectin levels were found to be decreased in both G1 and G3 HCV-infected patients [84]. The lack of clarity regarding HCV infection and adiponectin alterations seems to stem from the heterogeneous hepatic pathologies, metabolic conditions, and immune reactions of the patients involved in various studies. In HCV-infected patients, hepatic fibrosis [76,81]/inflammation [85] and steatosis [79,84,86,87,88,89] are associated with hyperadiponectinemia and hypoadiponectinemia, respectively. Additionally, adiponectin was found to be negatively correlated with insulin resistance, hepatic steatosis, and metabolic syndrome [90]. Consistent with this finding, our previous study showed that HCV core-induced nonobese hepatic steatosis is associated with hypoadiponectinemia; however, these effects may be ameliorated by adiponectin treatment [91]. Moreover, an anti-HCV-specific immune response was found to be strongly associated with higher serum total adiponectin and high-molecular-weight (HMW) adiponectin levels [84]. Whether HCV viral clearance leads to hyper or hypoadiponectinemia remains unclear and may differ between G3 and G4 HCV infections [81,92]. However, a large cohort study of 747 consecutive patients with G1, G2, G3, and G6 HCV infection showed that the adiponectin level and the aminotransferase-to-platelet ratio index decreased 24 weeks post-therapy in patients with SVR. During HCV infection, adiponectin may affect insulin sensitivity through triglycerides. After viral clearance, adiponectin levels decrease; moreover, they are directly associated with insulin sensitivity and decrease upon the improvement of hepatic fibrosis [93]. Thus, after SVR, the decrease in adiponectin in G4 HCV-infected patients [81] may reflect the reversal of hepatic fibrosis and hypotriglyceridemia, whereas the increase in adiponectin in G3 HCV-infected patients [92] may indicate an improvement in hepatic steatosis, which is most evident in G3 HCV-infected patients [94]. Hepatic steatosis associated with infection with G3 but not other genotypes of HCV was improved after SVR [95]. Regarding HCC, in G1 HCV-associated HCC, baseline adiponectin levels were found to be positively associated with the occurrence of HCC, independent of the HCV replication status [96], and higher levels of plasma adiponectin may predict poor HCC survival in patients without liver transplantation [97]. However, serum adiponectin was found to be decreased in patients with HCC and to be inversely correlated with tumor size and number [98]. Moreover, in patients with HCV-related cirrhosis, serum adiponectin levels were significantly lower in patients who also had HCC, and the serum adiponectin level was significantly negatively correlated with both the overall tumor size and the number of tumor foci [99]. Lower serum total and HMW adiponectin levels were independent risk factors for the higher histological grade of HCC [100]. However, high serum levels of adiponectin were associated with higher all-cause, liver-unrelated, and liver-related mortality [101].

PAI-1

Although serum PAI-1 levels have been identified as positive predictors of the response to IFN-based therapy in G1 HCV-infected patients [102], another study of G1, G2, G3, and G6 HCV-infected patients showed no difference in pretherapy PAI-1 levels between patients with and without SVR. The study also demonstrated that the PAI-1-rs-1799889 and IFN-λ3-rs12979860 genotypes longitudinally affect the PAI-1 level and that patients with SVR showed increasing PAI-1 levels with escalating cardiovascular risk [103].

Visfatin

The serum visfatin concentration was found to increase significantly in patients with chronic HCV infection compared with controls [104,105] and was closely related to the low-density lipoprotein cholesterol level [106] and fibrosis score [107]. In patients with different stages of HCV infection, the plasma visfatin level was associated with the presence of HCC [108]. Consistent with this finding, the serum levels of visfatin differed significantly among HCC, HCV, and normal control groups, and the visfatin level was associated with liver cirrhosis in HCV-infected patients [109]. On the other hand, no correlation between visfatin and HCV genotype, viral load, or treatment response to IFN-based therapy has been shown [107].

RBP4

In a cross-sectional study, patients with chronic HCV infection had lower RBP4 levels than did control subjects, and higher RBP4 levels were linked to lower ALT levels, hyperlipidemia, and high HOMA-IR scores [110]. Moreover, a significant decrease in serum RBP4 levels in patients with advanced stages of disease due to HCV infection was reported [111]. Consistent with this finding, an inverse association between the serum RBP4 concentration and the fibrosis stage was found in patients with HCV infection [112]. However, in the JFH1 infectious cell culture system, HCV core protein-enhanced RBP4 levels, and partial knockdown of RBP4 had a positive impact on HCV replication [113]. Only patients with SVR after IFN-based therapy had higher RBP4 levels post-therapy than at baseline [114].

Resistin

Hyperresistinemia in patients with chronic HCV infection has been consistently reported [115,116,117,118,119]. This condition is reversed after viral clearance [55,120,121] and determines moderate to severe fibrosis [117]. Our previous study showed that resistin originates primarily from intrahepatic lymphocytes, stellate cells, Kupffer cells, hepatic progenitor cells, and hepatocytes in HCV-infected patients [120]. Although the baseline resistin level was reported to be unassociated with therapeutic response [55], fine-tuned by resisin SNPs including RETN-rs34861192, RETN-rs3219175, RETN-rs3745367, and RETN-rs1423096, the intrahepatic, multicellular resistin reinforced IFNL3 in eliminating HCV via immunomodulation [120]. Moreover, high serum resistin levels might allow early identification of patients with cirrhosis who are at substantially increased risk of HCC [121].

Chemerin

Serum chemerin levels were significantly higher in patients with HCV infection than in controls, although chemerin levels were negatively associated with the necroinflammatory stage [122]. On the other hand, there was a negative association between serum chemerin and hepatic chemerin expression, which was not associated with necroinflammatory activity, steatosis grade, fibrosis stage, or metabolic abnormalities in HCV-infected patients [123].

Multiple Adipokines

Adiponectin, leptin, and visfatin have been found to be associated with liver cirrhosis in HCV-infected patients [109]. Sex was associated with leptin and adiponectin levels, and body mass index (BMI) was associated with leptin and PAI-1 levels in HCV-infected patients at baseline. Among patients achieving SVR, at 24 weeks post-IFN-based therapy, sex and BMI were associated with leptin, adiponectin, and PAI-1 levels; hepatic steatosis and the aspartate aminotransferase-to-platelet ratio index with adiponectin levels; and the HOMA-IR score and HCV genotype with PAI-1 levels [62]. Serum leptin levels were higher in G1 HCV-infected patients than in G3 HCV-infected patients, and serum resistin levels were higher in G3 HCV-infected patients [116]. In patients with compensated HCV-associated cirrhosis, insulin resistance but not the serum levels of adiponectin and leptin predicted the occurrence of HCC and of liver-related death or transplantation [124]. The serum levels of leptin and resistin and the leptin-to-adiponectin ratio were significantly higher in patients with chronic HCV infection than in controls, and low serum levels of resistin were associated with the presence of fibrosis independent of potential confounders [115]. In nonobese HCV core transgenic mice, hepatic steatosis is associated with downregulated leptin gene and hypoadiponectinemia, and these effects may be ameliorated by adiponectin treatment [91].

2.3. Autoimmune Liver Disease

2.3.1. Primary Biliary Cholangitis (PBC)

PBC predominantly affects middle-aged women and is a rare, chronic progressive cholestatic liver disease characterized by the autoimmune-mediated destruction of the small- and medium-sized intrahepatic bile ducts [125].

Leptin

Most studies of adipokines in PBC patients involving leptin have shown diverse results. Leptin levels have been reported to be either higher [126,127] or lower [128,129,130] in PBC patients than in controls. Leptin levels have been reported either to be associated with the histological stage of PBC [130] or to be unrelated to disease severity [128].

Adiponectin and Resistin

Adiponectin and resistin levels have been reported to be higher in PBC patients than in controls [127].

2.3.2. Autoimmune Hepatitis (AIH)

AIH is a rare, immune-mediated, inflammatory condition of the liver that is characterized by circulating autoantibodies, hypergammaglobulinaemia, and distinctive features on liver biopsy [131].

Adiponectin

A positive association between inflammation and adiponectin is usually reported in inflammatory/immune pathologies, in contrast with the negative correlation typical in metabolic diseases [132]. For example, patients with AIH showed significantly higher adiponectin concentrations than controls despite their higher HOMA-IR scores [133].

2.4. Alcoholic Liver Disease (ALD)

ALD, caused by excess and chronic alcohol intake [134], is a complex disorder with a disease spectrum ranging from steatosis to steatohepatitis, cirrhosis, and HCC [135]. Alcohol is primarily metabolized in the body via diverse pathways by the catalytic activity of three different enzymes–alcohol dehydrogenase, cytochrome P450 2E1 (CYP2E1), and catalase. Studies on alcoholic patients and rodent models have shown that chronic ethanol consumption reduces adipose tissue mass and causes CYP2E1-mediated oxidative stress and inflammation of adipose tissue [134].

2.4.1. Leptin

The effects of alcohol on circulating leptin are not consistent and may be related to changes in fat mass instead of the alcohol per se. Leptin has been reported to be increased, decreased, or unchanged across a range of rodent models of chronic alcohol administration. Likewise, the serum leptin levels from humans appear to be unrelated to alcohol intake, although exceptions do exist. In alcoholic patients, leptin levels have been reported to be increased, decreased, or unchanged, and serum leptin levels were not altered by either alcohol withdrawal or the severity of liver disease [136]. However, in alcohol-dependent patients with cirrhosis, leptin is significantly higher before liver transplantation and decreases significantly after transplantation. Moreover, alcohol-dependent patients on the waiting list had significantly higher leptin promoter methylation values than patients who underwent liver transplantation for other reasons [137].

2.4.2. Adiponectin

Alcohol exhibits a specific effect on serum adiponectin levels that is dose- and time-dependent and is correlated with the degree of hepatic damage. Moreover, alcohol does not seem to affect adiponectin expression in adipocytes directly but potentially affects it via mediators systemically released as a result of chronic alcohol intake [138]. The majority of data garnered from animal models of chronic alcohol consumption show circulating adiponectin levels to be decreased, although a few report no change. Conversely, serum adiponectin levels in humans were increased in relation to alcohol consumption, although two investigations did report a dose-dependent decrease [136]. In addition, a study of cirrhosis and control patients showed that transplant-free survival was significantly lower among patients with alcoholic liver disease and adiponectin ≥17 μg/mL. Adiponectin levels were associated with the intensity of liver dysfunction and worse prognosis in patients with alcoholic liver disease, suggesting its potential as a prognostic biomarker [139]. Emerging evidence has revealed that dysregulated adiponectin-fibroblast growth factor (FGF) 15 (human homolog, FGF19) axis and impaired hepatic adiponectin-FGF15/19 signaling are associated with alcoholic liver damage in rodents and humans [140].

2.4.3. Other Adipokines

The levels of chemerin decrease with the progression of liver damage during alcoholic liver cirrhosis [141]. In addition, a novel adipose tissue-derived cytokine, C1q TNF-related protein-3 (CTRP3), was shown to attenuate hepatic triglyceride accumulation in response to long-term chronic, but not short-term, alcohol consumption [142].

3. Adipokines and the Pancreas

3.1. Pancreatic Cancer

Human pancreatic adipocytes store lipids and release adipokines in response to the overall metabolic, humoral, and neuronal status [143]. Fatty pancreas is associated with age, BMI, and diabetes, which are risk factors for pancreatic cancer [144]. In particular, expansion and inflammation of visceral adipose tissue induce insulin resistance that fosters systemic secretion of insulin and insulin-like growth factor 1 [145].

3.1.1. Leptin

Elevated leptin may promote pancreatic tumor invasion and metastasis, activating the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) axis [145].

3.1.2. Adiponectin

Reduced release of adiponectin was found to decrease the tumor-suppressive effects of adiponectin in a manner mediated by JAK2/STAT3 inhibition and downregulation of intracellular β-catenin [145].

3.2. Insulin Resistance and Diabetes

Insulin resistance is characterized by a diminished response to insulin stimulation, resulting in the failure of target tissues to adequately dispose of blood glucose, inhibit lipolysis, stimulate glycogen synthesis, and inhibit hepatic glucose output and is a precursor event to type 2 diabetes [146].

3.2.1. Leptin

Adipokines preferentially affect islet vasculature [147]. Pancreatic hormones play a role in energy balance, exerting short-acting control, while insulin and leptin derived from adipose tissue are involved in long-acting adiposity signaling and regulate body weight [148]. Leptin receptors are widely expressed in peripheral tissues, including the beta (β) cells of the endocrine pancreas [149], and their activation directly inhibits insulin secretion from these endocrine cells. Additionally, β cell mass can be affected by leptin through changes in proliferation, apoptosis, or cell size [150]. Specifically, insulin is adipogenic, increases body adipose tissue mass, and stimulates the production and secretion of leptin, which acts centrally to reduce food intake and increase energy expenditure. Leptin, in turn, suppresses insulin secretion by both central actions and direct actions on β cells. Because leptin levels are directly proportional to body adipose tissue mass, an increase in adiposity increases plasma leptin, thereby curtailing insulin production and further increasing fat mass [151], thus establishing a hormonal regulatory feedback loop, the adipo-insular axis [151]. In addition, leptin exerts a tonic inhibitory effect on β cell excitability via its ability to increase the plasma membrane ATP-sensitive K+ (KATP) channel density and whole-cell KATP channel current [152]. In most overweight individuals, physiological regulation of body weight by leptin is likely disturbed, constituting leptin resistance. This leptin resistance at the pancreatic β cell level may contribute to dysregulation of the adipo-insular axis and accelerate the development of hyperinsulinemia and can manifest as diabetes mellitus in overweight patients [153]. On the other hand, leptin might be used as an adjunct to insulin therapy in patients with insulin-deficient diabetes, providing insight into its therapeutic properties as an antidiabetic agent [154]; moreover, leptin monotherapy has been reported to reverse type 1 diabetes independent of insulin [155]. Because ob/ob mice lack functional leptin, they develop severe insulin resistance with hyperglycemia and hyperinsulinemia and are described as a model for the prediabetic state. Although ob/ob mice have large pancreatic islets, their β cells respond adequately to most stimuli [156].

3.2.2. Other Adipokines

In addition to leptin, other adipokines, including adiponectin and visfatin (i.e., eNAMPT), apelin, resistin, RBP4, fibroblast growth factor 21, nesfatin-1, and fatty acid binding protein 4 directly regulate β cell function [157,158]. In particular, adiponectin has received considerable attention for its potential antidiabetic actions. By stimulating adipogenesis, opposing inflammation, and influencing rates of lipid oxidation and lipolysis, adiponectin critically governs lipid spillover into nonadipose tissues [159]. Moreover, adiponectin stimulates insulin secretion and has antiapoptotic properties in β cells [160]. Resistin antagonizes insulin action, and it is downregulated by rosiglitazone and peroxisome proliferator-activated receptor gamma agonists [161]. Interestingly, visfatin does not exert insulin-mimetic effects in vitro or in vivo but rather exhibits robust nicotinamide adenine dinucleotide (NAD) biosynthetic activity. NAMPT-mediated systemic NAD biosynthesis is critical for beta cell function, suggesting a vital framework for the regulation of glucose homeostasis [162].

4. Adipokines and the Alimentary Tract

4.1. Esophagus

Almost all cases of esophageal adenocarcinoma arise from underlying Barrett’s esophagus, a metaplastic change in the esophagus [163]. Moreover, central obesity is involved in the pathogenesis and progression of Barrett’s esophagus to esophageal adenocarcinoma [164,165] and GERD, a disorder due to the retrograde flow of refluxate into the esophagus [166]. Barrett’s esophagus, esophageal adenocarcinoma, and GERD thus might be associated with adipokine alterations.

4.1.1. Leptin

In obese patients with GERD, leptin, and ObR levels were found to be higher and lower, respectively, than in nonobese patients with GERD [167,168]. Consistent with this finding, leptin resistance in individuals with overweight and obesity is associated with features of GERD, and leptin levels are positively associated with frequent GERD symptoms [169] and with the clinical and endoscopic severity of GERD [170]. The multi-biomarker score derived from multiple parameters, including leptin levels and GERD frequency and duration, can identify patients with Barrett’s esophagus [171]. Moreover, leptin levels were found to be positively associated with Barrett’s esophagus; this association was stronger in men with GERD than in women with GERD [172], and serum leptin levels might be associated with an increased risk of Barrett’s esophagus among men but not women [165]. Through enhancing macrophage migration inhibitory factor-induced inflammatory signaling, leptin may contribute to the development of GERD [173]. In addition, leptin stimulates cell proliferation and inhibits apoptosis in OAC cells via extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K)/Akt, and JAK2-dependent activation of cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE2) production [174]. Leptin receptors are highly expressed on esophageal epithelial cells. The finding that patients with Barrett’s esophagus had higher fundic leptin levels than individuals with a normal esophagus indicates that ObR expression on esophageal epithelial cells provides a pathway for leptin-mediated signal transduction [175]. In particular, the oncogenic effect of leptin has been reported to modulate the cellular response to radiation [176], angiogenesis and lymphangiogenesis [177], and chemoresistance in gastroesophageal adenocarcinomas [178], as well as to stimulate the proliferation, invasion, and migration and inhibit the apoptosis of OE33 esophageal adenocarcinoma cells [179].

4.1.2. Adiponectin

The anti-inflammatory effects of adiponectin are specific to its individual multimers, with low-molecular-weight (LMW) adiponectin being the most anti-inflammatory. High levels of LMW adiponectin are associated with a decreased risk of Barrett’s esophagus among patients with GERD [180]. Consistent with this finding, serum adiponectin was found to be inversely associated with Barrett’s esophagus, particularly in men [181]; in patients with GERD, erosive esophagitis and Barrett’s esophagus were found to be associated with decreased adiponectin levels compared to those in patients without GERD [182]; and low serum adiponectin levels may be associated with an increased risk for erosive esophagitis [183]. However, in a study of 863 cases, adiponectin levels were positively associated with the risk of Barrett’s esophagus in patients with GERD and in smokers but not in a control population without GERD symptoms [184].

4.1.3. Leptin and Adiponectin

A systematic review showed that increased serum levels of leptin are associated with an increased risk of Barrett’s esophagus. In contrast, increased total serum levels of adiponectin do not seem to modify the risk of Barrett’s esophagus [185]. Similarly, the adjusted odds ratios for Barrett’s esophagus were 8.02 for the highest quintile vs. the lowest quintile of leptin level, while there were no differences in adiponectin levels between the cases and controls [186]. An increased level of leptin was associated with an increased risk for esophageal adenocarcinoma, whereas an increased level of HMW adiponectin was inversely associated with esophageal adenocarcinoma [187]. Interestingly, higher adiponectin levels were found in patients with esophageal squamous cell carcinoma (SCC) than in patients with esophageal adenocarcinoma [188], and resistin may be a biomarker for esophageal SCC [189].

4.2. Stomach

Leptin

Similar to adipose tissue, the stomach simultaneously expresses leptin and ObR. Leptin maintains energy homeostasis with the aid of its antagonistic hormone ghrelin [190]. Ghrelin is a gut-derived peptide hormone that was first isolated from the stomach [191]. Ghrelin stimulates appetite and controls gastric motility and acid secretion [192]. Collectively, leptin and ghrelin are known as “hunger hormones”. In addition, leptin signaling can affect the gastric mucosal milieu [193]. Adipose tissue secretes leptin in a slow constitutive endocrine manner, and the gastric mucosa releases leptin in a rapidly regulated exocrine manner into the gastric juice. Thus, adipocytes and gastric epithelial cells are two cell types in which metabolism is closely linked to food intake and energy storage [194]. Moreover, overexpression of leptin and phosphorylated ObR is implicated in gastric cancer, and diet-induced obesity causes precancerous lesions in the mouse stomach [193].

4.3. Small Intestine

Leptin

Creeping fat, characterized by hyperplasia of the mesenteric fat, which creeps around inflamed segments of the small intestine [195], can be distinguished from healthy adipose tissue by its distinctively small adipocytes with high levels of adipokines and dominant immune cell infiltration. In particular, leptin has been reported to enhance the maturation of the systemic and intestinal immune systems in preterm conditions [196].

4.4. Colon

4.4.1. Colitis

Inflammatory bowel diseases (IBDs) comprise chronic inflammatory disorders of the gastrointestinal tract, affecting millions worldwide [197]. The exact etiopathogenesis of IBD remains unknown, while potential factors involve genetic predisposition, environmental conditions, and immunological dysfunctions. The main IBDs are ulcerative colitis (UC) and Crohn’s disease (CD) [198]. Although transmural inflammation in CD may affect any part of the gastrointestinal tract, it occurs most frequently in the terminal ileum or the large intestine. In contrast, UC usually occurs only in the large intestine and is limited to the mucosal layer [199]. Obesity-induced chronic inflammation increases the risk of UC and CD [200]. Mesenteric adipose tissue (MAT) hyperplasia is a hallmark of CD. Mesenteric adipose-derived stromal cells (ADSCs) synthesize and release adipokines in a disease-dependent manner and alter colonic epithelial cell signaling [201].Transmural inflammation facilitates bacterial translocation into the creeping fat, which exerts a protective effect via a localized anti-inflammatory effect, thus preventing a systemic inflammatory response in CD [202].

Leptin

Leptin may regulate dendritic cell migration from the gut under homeostatic and inflammatory conditions, linking mesenteric obesity and inflammation in CD [203]. However, activation of ObR is an important pathogenic mechanism of UC, and ObR deficiency may confer resistance to 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis by inhibiting the nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) and Ras homolog gene family member A (RhoA) signaling pathways [204]. Moreover, luminal leptin is likely an intestinal chloride secretagogue, particularly when present at elevated concentrations or in the setting of inflammation [205]. Intraperitoneal administration of leptin to lean rats increased colonic epithelial permeability and altered zonula occludens-1 expression and organization [206], and the increased mucosal leptin may interact with mast cells and the nervous system to enhance diarrhea-predominant irritable bowel syndrome [207]. On the other hand, the protective mucosal immune function of leptin in Clostridium difficile colitis is partially mediated by a leptin-STAT3 inflammatory pathway that is defective as a result of the ObR Q223R mutation [208].

Adiponectin

In contrast to the proinflammatory role of leptin, adiponectin maintains intestinal homeostasis and protects against murine colitis through interactions with its receptor AdipoR1 and by modulating adaptive immunity [209]. For example, adiponectin injection alleviated colonic injury and rectal bleeding in mice, downregulated colonic interleukin 1β (IL-1β), and TNF-α expression, and regulated apoptosis-related gene expression to attenuate dextran sodium sulfate (DSS)-induced colonic inflammation [210,211]. In addition, adiponectin markedly reduced the serum lipopolysaccharide concentration, a biomarker for intestinal integrity, and enhanced colonic expression of tight junction proteins [211]. Adiponectin expression was significantly suppressed by induction of colitis [212], and intracolonic silencing of adipoR1 in mice exacerbated TNBS-induced colitis [213]. However, whether adiponectin aggravates [214] or attenuates [215] DSS-induced colitis in adiponectin knockout mice remains controversial.

Leptin and Adiponectin

Overall, colitis induces a decrease in the levels of the mRNAs encoding leptin and adiponectin in MAT but an increase in the levels of mRNAs encoding inflammatory markers. Specifically, MAT in patients with inflammatory bowel disease shows a loss of the adipose profile and a greatly enhanced inflammatory profile [216].

4.4.2. Diverticulosis

Creeping fat can be observed in CD. Interestingly, adipose tissue also frequently covers the basolateral site of inflamed diverticula, thus locally reflecting the phenomenon seen in CD. This finding suggests that each inflamed diverticulum mechanistically reflects CD on a miniature scale [217].

Leptin and Adiponectin

Leptin levels were found to be positively associated with diverticulosis, and LMW adiponectin levels were inversely related to the presence of diverticulosis in asymptomatic men [218].

4.4.3. Colon Polyps and Cancer

Colorectal cancer (CRC) is the third most common cancer in men and the second most common in women worldwide. Most CRCs arise from colonic polyps, particularly adenomatous polyps [219]. The polyp size, number, and pathological findings are crucial prognostic factors for CRC. Nonadvanced colonic polyps are defined as one to two adenomatous polyps each <10 mm in size, and advanced colonic polyps are defined as any adenomatous polyp ≥10 mm in size or with >25% villous histology or high-grade dysplasia [220]. Obesity is a risk factor for both adenomatous polyps and CRC development [219], which likely results in adipokine alteration.

Leptin

In asymptomatic men, serum leptin levels were found to be significantly associated with the presence of tubular adenoma [221]. Leptin expression was more frequently observed in colon adenomas, especially in larger adenomas and adenocarcinoma in situ, than in normal colon tissues, but blood leptin levels were not found to be related to tissue leptin expression [222]. Tissue microarray analysis showed that leptin was gradually expressed during the normal-adenoma-adenocarcinoma sequence, suggesting an association between leptin and colorectal carcinogenesis. Intriguingly, high leptin expression was an indicator of favorable tumor features and better survival in CRC patients [223]. ObR is overexpressed in CRC cells, which may influence patient outcomes [224]. Both leptin [225] and ObR [225,226] were found to be present at higher levels in cancerous tissues than in adjacent colon tissues. Moreover, high circulating levels of ObR were found in patients with advanced-stage colon cancer [227]. However, a study of 2258 cases showed that soluble ObR levels were strongly inversely associated with CRC, whereas leptin was not associated with the risk of CRC [228]. Moreover, ObR was significantly correlated with early-stage and well-differentiated primary CRCs [229]. ObR expression was found to be higher in CRCs than in the corresponding normal mucosa, and ObR expression in tumors might be involved in the adaptive immune response in sporadic CRCs, likely via a microsatellite instability-high phenotype orientation [230]. Intriguingly, patients with ObR-positive tumors were found to have significantly better overall survival than those with ObR-negative tumors, and Ob-R is a prognostic marker associated with more favorable survival [229]. In human colon cancer, upregulation of leptin pathway members was found, and a large network of dysregulated transcripts was linked to poorer overall survival [231]. For example, leptin might regulate the proliferation, apoptosis, or invasion of CRC cells through the PI3K/Akt/mammalian target of rapamycin (mTOR) [232,233], nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent Silent Information Regulator 2 Homolog 1 (SIRT1) [234], ERK1/2 [235,236,237], MAPK [236,237,238], JAK2, STAT3, activator protein 1 (AP-1) [239,240] and NF-κB [241] signaling pathways. In addition, leptin regulates proinflammatory genes such as interleukin 6 (IL-6), IL1β, and chemokine (C-X-C motif) ligand 1 (CXCL1) [242], and induces preneoplastic colon epithelial cells to orchestrate vascular endothelial growth factor (VEGF)-driven angiogenesis and vascular development [243]. In leptin-deficient ob/ob and ObR-deficient db/db mice, colon tumor growth was inhibited, although the animals exhibited severe obesity [226]; moreover, in leptin-deficient ob/ob mice, the presence of abnormally dense mucus-filled goblet cells suggested the possible involvement of leptin in tissue injury and/or mucosal defense mechanisms. Furthermore, in human colonic goblet-like HT29-MTX cells expressing ObR, leptin increased mucin secretion by activating protein kinase C (PKC)- and PI3K-dependent pathways [244].

Adiponectin

In contrast to leptin, adiponectin protects against chronic inflammation-induced colon cancer (CICC) [245] and demonstrates beneficial effects on colon cancer [209]. Adiponectin may be involved in reducing the severity of CICC by preventing goblet cell apoptosis and increasing epithelial-to-goblet cell differentiation [246]. Plasma adiponectin levels have been found to be inversely associated with colonic polyps, multiple colonic polyps, high-risk colonic polyps [247], early-stage CRC [248,249], and CRC stage [249]. Consistent with these findings, adiponectin negatively regulates colorectal cell survival and migration [250]. Both adiponectin and AdipoRon, a small molecule adiponectin receptor agonist, were found to suppress colon cancer risk in part by reducing the number of leucine-rich repeat-containing G protein-coupled receptor 5+ (Lgr5+) stem cells in mouse colonic organoids [251]. In a study of 2412 cases, non-HMW but not HMW adiponectin was associated with CRC risk [252]. The expression of AdipoR1 has consistently been reported to be higher in cancerous than in normal colonic tissues [253,254], while the expression of AdipoR2 has been reported to be lower [255] or higher [256] in cancerous tissues. Low plasma adiponectin levels were found to be associated with KRAS-mutant CRC risk but not with KRAS wild-type cancer risk [256]. Additionally, adiponectin might inhibit the growth of colon cancer cells by stimulating AMPK activity [257,258], thereby downregulating the mTOR pathway [259]. Additionally, adiponectin might regulate IL1β-induced colon carcinogenesis [260]. On the other hand, adiponectin signaling plays a role in modulating cellular cholesterol homeostasis, plasma membrane biophysical properties, and Wnt-driven signaling [261]. Adiponectin treatment suppresses angiogenesis in colon cancers. In vitro studies showed that adiponectin directly controls the malignant potential (cell proliferation, adhesion, invasion, and colony formation) and regulates metabolic (AMPK/70-kDa ribosomal protein S6), inflammatory (STAT3/VEGF), and cell cycle (p21/p27/p53/cyclins) signaling pathways in a liver kinase B1 (LKB)-dependent manner [261]. However, in another study, adiponectin levels were not correlated with visceral fat in the CRC and adenoma groups [262]. The responsiveness of colonic stem cells to adiponectin in diet-induced obesity is impaired and may contribute to the accumulation of stem cells observed in obesity [263]. Moreover, adiponectin was found to suppress colonic epithelial proliferation via inhibition of the mTOR pathway under high-fat diet but not basal diet feeding conditions [264]. A significant inverse correlation was found between the number of dysplastic aberrant crypt foci (ACF) and the plasma adiponectin level. Consistent with this finding, enhanced formation of ACF and tumors was observed in adiponectin-deficient mice [265], whichdevelop more intestinal tumors than wild-type mice [258], and adiponectin administration suppressed the growth of implanted tumors, causing larger central necrotic areas in the mice [261]. However, elevated levels of circulating adiponectin in adiponectin transgenic mice did not confer protection against colon tumor development [266]. The rs12733285C/T genotype and the A allele of rs1342387 (A/G or A/A) of ADIPOR1 are protective factors for CRC, while the rs266729G/C genotype and the G allele of ADIPOQ are risk factors for colon cancer [267]. Together, given that the concentration of adiponectin is high in serum, these findings indicate that the main role of adiponectin is likely homeostasis regulation rather than action as an anticancer adipokine. However, as the above epidemiological evidence shows, a low adiponectin level may be a basic risk factor for CRC. It is likely that the colonic epithelium is stimulated by specific carcinogens and that cancer development is then facilitated underhypoadiponectinemia [268].

Leptin and Adiponectin

Serum leptin and AdipoR1 and AdipoR2 expression levels were found to be associated with lymph node involvement, and AdipoR1 expression was correlated with tumor size in colon cancer patients [269]. Ionizing radiation can persistently decrease the levels of AdipoR1 and AdipoR2 but increase those of leptin and ObR and activate downstream proliferative pathways, for example, upregulating PI3K/Akt and JAK2 signaling, which may contribute to carcinogenesis [270]. Regarding sexual dimorphism, plasma adiponectin levels were found to be associated with a reduced risk of CRC among men but not among women [271].

Other Adipokines

Serum resistin levels in patients with colon cancer are elevated and correlated with tumor grade. Resistin binds to Toll-like receptor 4 (TLR4) on the colon cancer cell membrane and initiates TLR4-myeloid differentiation primary response 88 (MYD88)-dependent activation of ERK [272,273]. In addition, the resistin C-420G and G+299A polymorphisms have potential roles in the genetic predisposition to colon cancer [274]. High serum levels of YKL-40 (also called Chitinase 3-like 1) are associated with CRC in subjects without comorbidities [275] and are correlated with poor prognosis in patients with colon cancer [276]. Colon adenoma risk is associated with high circulating levels of RBP4 [277]. Finally, chemerin is thought to exert chemotactic, adipogenic, and angiogenic functions. Higher chemerin levels are associated with CRC risks [278,279]
A schematic summarizing colon disease-associated adipokine alterations and the basis is provided in Figure 2.

5. Adipokines and the Biliary Tract

5.1. Leptin

Although higher leptin concentrations in the hepatic vein were found in bile duct ligated-(BDL) rats than in lean sham-operated rats, and colocalization of leptin and α-smooth muscle actin in activated hepatic stellate cells (HSCs) was observed by immunohistochemistry [280], the TNF-α-associated upregulation of leptin in dimethylnitrosamine (DMN)-induced but not in BDL-induced cirrhotic rats is consistent with a difference in the roles of TNF-α in rats with nonbiliary cirrhosis and those with biliary cirrhosis [281]. Regarding cholangiocarcinoma, leptin was found to increase the proliferation and metastatic potential of cholangiocarcinoma cells through STAT3-dependent activation of ERK 1/2. Moreover, loss of leptin function suppressed the development of cholangiocarcinoma [282]. Consistent with this finding, leptin increased the epithelial-mesenchymal transition and proangiogenic capability of cholangiocarcinoma cells, inhibited endogenous miR-122 expression, and upregulated pyruvate kinase muscle isozyme M2 [283].

5.2. Adiponectin

High levels of adiponectin were found in BDL rats [284], reflecting the antifibrotic role of adiponectin, as adiponectin overexpression in activated HSCs was found to reduce the proliferation but augment the apoptosis of HSCs [280]. Consistent with this finding, adiponectin protected the rat bile duct against early warm ischemia-reperfusion injury by suppressing the inflammatory response and hepatocyte apoptosis and NF-κB (p65) played an important role in this process [285].

5.3. Resistin

Hyperinsulinemia might upregulate the resistin gene in BDL-related cirrhosis [286].

6. Adipokines and the Gallbladder

Obesity, diabetes, and hyperlipidemia are known risk factors for the development of gallstones [287], and there is convincing evidence that excess body weight is associated with an increased risk for gallbladder cancer [288]. Gallbladder diseases, therefore, potentially lead to adipokine alteration.

6.1. Leptin

Prepregnancy obesity and the serum leptin concentration are strong risk factors for pregnancy-associated gallbladder disease [289], although a human study showed that the serum leptin concentration might not be a better predictor of gallbladder disease than anthropometry [290]. Leptin was found to promote cholesterol crystallization and gallstone formation [291] and, consistent with this finding, was reported to affect the components and secretion of bile in leptin-deficient mice. Furthermore, gallbladder diseases such as cholelithiasis are associated with serum leptin levels in humans [292] and dogs [293]. Leptin influences gallbladder bile volume, sodium, and pH, as well as numerous inflammatory cytokine genes and genes related to water, sodium, chloride, and bicarbonate transport [294]. Obese leptin-deficient (ob-ob) mice have large gallbladder volumes with decreased contraction and are predisposed to gallstone formation [295,296], and administration of leptin to these mice causes weight loss and restores gallbladder function [295]. Both leptin and ObR are localized throughout the cytoplasm of luminal and glandular epithelial cells in the canine gallbladder [292] and in human gallbladder cancer tissues and cell lines [297]. ObR-deficient (db-db) obese mice have an increased gallbladder volume due to abnormal gallbladder motility [298], decreased biliary cholesterol saturation despite elevated serum cholesterol, and hepatic steatosis, and decreased cholesterol crystal formation [299]. A large body of evidence demonstrates that high BMI, as an approximation for general adiposity, is a risk factor for the development of gallbladder cancers [300]. Consistent with this observation, leptin was found to promote the proliferation, migration, and invasion of gallbladder cancer cells by increasing ObR expression through the SOCS3/JAK2/p-STAT3 signaling pathway [297].

6.2. Adiponectin

Hypoadiponectinemia has been reported to be associated with cholesterol gallstone formation in humans and to promote gallstone formation in mice [301].
A summary of adipokine alterations in various clinical digestive diseases is provided in Table 1. The alteration patterns might act as diagnostic markers or therapeutic targets for specific digestive diseases.

7. Conclusive Remarks and Future Challenges

Considering the current review, almost all digestive diseases are associated with altered adipokine profiles; with few exceptions, the unfavorable and favorable implications of leptin and adiponectin, respectively, have been consistently reported. However, gaps remain in understanding the precise roles of adipokines in digestive diseases. For example, in addition topatients with lipodystrophy and those with insulin-deficient diabetes, which patients will benefit from leptin therapy? Is adiponectin therapy a promising approach for most patients with digestive diseases? In addition, many associated mechanisms have been explored in vitro or in animal studies. Future prospective studies in largeindependent cohorts with identifiable outcomes for specific digestive diseases and sophisticated molecular investigations are required to verify the proposed basis and to investigate the therapeutic targets in confirming the fundamental mechanisms underlying the findings described herein.

Author Contributions

Z.Y. and S.-S.Y. interpreted the data and wrote the manuscript. M.-L.C. designed and completed the study, drafted the article, and critically revised it for intellectual content. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by grants from the Chang Gung Medical Research Program (CMRPG3I0412 and CMRPG3K0721) and the National Science Council, Taiwan (MOST 108-2314-B-182-051-, 109-2314-B-182-024- and 109-2629-B-182-002-). The funders had no role in study design, data collection, and analysis, decision to publish, or preparation of the manuscript.

Acknowledgments

The authors thank Shu-Chun Chen, Chia-Hui Tsai, Chun-Kai Liang, and Shuen-Shian Shiau from the Division of Hepatology, Department of Gastroenterology, Chang Gung Memorial Hospital, Taiwan, for their assistance with data mining; and Chun-Ming Fan from the Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, and Shiang-Chi Chen from the Department of Nursing, Taipei Medical University, Taiwan for their generation of excellent figures.

Conflicts of Interest

The authors declare that they have no competing interest.

Abbreviations

NAFLDnonalcoholic fatty liver disease
PPAR-αproliferator-activated receptor-alpha
ObRleptin receptor
NASHnonalcoholic steatohepatitis
AdipoR1adiponectin receptor 1
TNF-αtumor necrosis factor-alpha
BAbile acid
SNPsingle nucleotide polymorphism
PAI-1plasminogen activator inhibitor-1
RBP-4retinol-binding protein-4
HBVhepatitis B virus
CHBchronic hepatitis B
IFN-αinterferon-alpha
HCChepatocellular carcinoma
ALTalanine aminotransferase
IFNL3interferon λ3
DAAdirect-acting antiviral agent
Ggenotype
SVRsustained virologic response
C3component 3
HMWhigh-molecular-weight
PBCprimary biliary cholangitis
AIHautoimmune hepatitis
BMIbody mass index
JAK2Janus kinase 2
STAT3signal transducer and activator of transcription 3
KATPATP-sensitive K+
GERDgastroesophageal reflux disease
ERKextracellular signal-regulated kinase
MAPKmitogen-activated protein kinase
PI3Kphosphoinositide 3-kinase
COX-2cyclooxygenase-2;
PGE2prostaglandin E2
LMWlow-molecular-weight
SCCsquamous cell carcinoma
UCulcerative colitis
CDCrohn’s disease
ADSCadipose-derived stromal cells
TNBS2,4,6-trinitrobenzene sulfonic acid
NF-κBnuclear factor κ-light-chain-enhancer of activated B cells
RhoARas homolog gene family, member A
IL-1βinterleukin 1β
DSSdextran sodium sulfate
MATmesenteric adipose tissue
CRCcolorectal cancer
mTORmammalian target of rapamycin
Nrf2nuclear factor erythroid 2-related factor 2
SIRT1silent information regulator 2 homologue 1
AP-1activator protein 1
IL-6interleukin 6
CXCL1chemokine (C-X-C motif) ligand 1
VEGFvascular endothelial growth factor
PKCprotein kinase C
CICCchronic inflammation-induced colon cancer
Lgr5+leucine-rich repeat-containing G-protein coupled receptor 5+
AMPKAMP-activated protein kinase
LKBliver kinase B1
ACFaberrant crypt foci
TLR4Toll-like receptor 4
MYD88myeloid differentiation primary response 88;
RETNresistin
BDLbile duct-ligated
DMNdimethylnitrosamine
HSChepatic stellate cell

References

  1. Kershaw, E.E.; Flier, J.S. Adipose Tissue as an Endocrine Organ. J. Clin. Endocrinol. Metab. 2004, 89, 2548–2556. [Google Scholar] [CrossRef] [PubMed]
  2. Han, S.; Sun, H.M.; Hwang, K.-C.; Kim, S.-W. Adipose-Derived Stromal Vascular Fraction Cells: Update on Clinical Utility and Efficacy. Crit. Rev. Eukaryot. Gene Expr. 2015, 25, 145–152. [Google Scholar] [CrossRef] [PubMed]
  3. Romacho, T.; Elsen, M.; Röhrborn, D.; Eckel, J. Adipose tissue and its role in organ crosstalk. Acta Physiol. 2014, 210, 733–753. [Google Scholar] [CrossRef] [PubMed]
  4. Szydło, B.; Kiczmer, P.; Świętochowska, E.; Ostrowska, Z. Role of omentin and chemerin in metabolic syndrome and tumor diseases. Postępy Hig. Med. Doświadczalnej 2016, 70, 844–849. [Google Scholar] [CrossRef] [PubMed]
  5. Batra, A.; Siegmund, B. The role of visceral fat. Dig. Dis. 2012, 30, 70–74. [Google Scholar] [CrossRef] [PubMed]
  6. Strong, A.L.; Burow, M.E.; Gimble, J.M.; Bunnell, B.A. Concise Review: The Obesity Cancer Paradigm: Exploration of the Interactions and Crosstalk with Adipose Stem Cells. Stem Cells 2015, 33, 318–326. [Google Scholar] [CrossRef] [Green Version]
  7. Yehuda-Shnaidman, E.; Schwartz, B. Mechanisms linking obesity, inflammation and altered metabolism to colon carcinogenesis. Obes. Rev. 2012, 13, 1083–1095. [Google Scholar] [CrossRef]
  8. Nieman, K.M.; Romero, I.L.; Van Houten, B.; Lengyel, E. Adipose tissue and adipocytes support tumorigenesis and metastasis. Biochim. Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2013, 1831, 1533–1541. [Google Scholar] [CrossRef] [Green Version]
  9. Sun, X.; Harris, E.N. New aspects of hepatic endothelial cells in physiology and nonalcoholic fatty liver disease. Am. J. Physiol. Physiol. 2020, 318, C1200–C1213. [Google Scholar] [CrossRef]
  10. Azzu, V.; Vacca, M.; Virtue, S.; Allison, M.; Vidal-Puig, A. Adipose Tissue-Liver Cross Talk in the Control of Whole-Body Metabolism: Implications in Nonalcoholic Fatty Liver Disease. Gastroenteroloy 2020, 158, 1899–1912. [Google Scholar] [CrossRef]
  11. Renehan, A.G.; Roberts, D.L.; Dive, C. Obesity and cancer: Pathophysiological and biological mechanisms. Arch. Physiol. Biochem. 2008, 114, 71–83. [Google Scholar] [CrossRef]
  12. Nam, S.Y. Obesity-Related Digestive Diseases and Their Pathophysiology. Gut Liver 2017, 11, 323–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Yamamoto, S.; Watabe, K.; Takehara, T. Is Obesity a New Risk Factor for Gastritis? Digestion 2012, 85, 108–110. [Google Scholar] [CrossRef] [PubMed]
  14. Conde, J.; Scotece, M.; Gómez, R.; López, V.; Gómez-Reino, J.J.; Lago, F.; Gualillo, O. Adipokines: BioFactors from white adipose tissue. A complex hub among inflammation, metabolism, and immunity. BioFactors 2001, 37, 413–420. [Google Scholar] [CrossRef] [PubMed]
  15. Lehr, S.; Hartwig, S.; Sell, H. Adipokines: A treasure trove for the discovery of biomarkers for metabolic disorders. Proteom. Clin. Appl. 2011, 6, 91–101. [Google Scholar] [CrossRef] [PubMed]
  16. Boddicker, R.L.; Whitley, E.; Birt, D.F.; Spurlock, M.E. Early Lesion Formation in Colorectal Carcinogenesis Is Associated With Adiponectin Status Whereas Neoplastic Lesions Are Associated With Diet and Sex in C57BL/6J Mice. Nutr. Cancer 2011, 63, 1297–1306. [Google Scholar] [CrossRef]
  17. Fenton, J.I.; Birmingham, J.M.; Hursting, S.D.; Hord, N.G. Adiponectin blocks multiple signaling cascades associated with leptin-induced cell proliferation inApcMin/+ colon epithelial cells. Int. J. Cancer 2008, 122, 2437–2445. [Google Scholar] [CrossRef]
  18. Parthasarathy, G.; Revelo, X.; Malhi, H. Pathogenesis of Nonalcoholic Steatohepatitis: An Overview. Hepatol. Commun. 2020, 4, 478–492. [Google Scholar] [CrossRef] [Green Version]
  19. Froehlich, S.J.; Lackerbauer, C.A.; Rudolph, G.; Rémi, J.; Noachtar, S.; Heppt, W.J.; Cryer, A.; Zenner, H.-P.; Niller, H.H.; Schwarzmann, F.; et al. Nonalcoholic Steatohepatitis. Encycl. Mol. Mech. Dis. 2009, 1487. [Google Scholar] [CrossRef]
  20. Boutari, C.; Perakakis, N.; Mantzoros, C.S. Association of Adipokines with Development and Progression of Nonalcoholic Fatty Liver Disease. Endocrinol. Metab. 2018, 33, 33–43. [Google Scholar] [CrossRef]
  21. Sennello, J.A.; Fayad, R.; Morris, A.M.; Eckel, R.H.; Asilmaz, E.; Montez, J.; Friedman, J.M.; Dinarello, C.A.; Fantuzzi, G. Regulation of T Cell-Mediated Hepatic Inflammation by Adiponectin and Leptin. Endocrinoloy 2005, 146, 2157–2164. [Google Scholar] [CrossRef] [Green Version]
  22. Giby, V.G.; Ajith, T.A. Role of adipokines and peroxisome proliferator-activated receptors in nonalcoholic fatty liver disease. World J. Hepatol. 2014, 6, 570–579. [Google Scholar] [CrossRef]
  23. Polyzos, S.A.; Aronis, K.N.; Kountouras, J.; Raptis, D.D.; Vasiloglou, M.F.; Mantzoros, C.S. Circulating leptin in non-alcoholic fatty liver disease: A systematic review and meta-analysis. Diabetoloy 2016, 59, 30–43. [Google Scholar] [CrossRef]
  24. Li, X.-L.; Sui, J.-Q.; Lu, L.-L.; Zhang, N.-N.; Xu, X.; Dong, Q.-Y.; Xin, Y.; Xuan, S. Gene polymorphisms associated with non-alcoholic fatty liver disease and coronary artery disease: A concise review. Lipids Health Dis. 2016, 15, 53. [Google Scholar] [CrossRef] [Green Version]
  25. Haque, W.A.; Shimomura, I.; Matsuzawa, Y.; Garg, A. Serum adiponectin and leptin levels in patients with lipodystrophies. J. Clin. Endocrinol. Metab. 2002, 87, 2395. [Google Scholar] [CrossRef]
  26. Zadeh, E.S.; Lungu, A.O.; Cochran, E.K.; Brown, R.J.; Ghany, M.G.; Heller, T.; Kleiner, D.E.; Gorden, P. The liver diseases of lipodystrophy: The long-term effect of leptin treatment. J. Hepatol. 2013, 59, 131–137. [Google Scholar] [CrossRef] [Green Version]
  27. Shabani, P.; Emamgholipour, S.; Doosti, M. CTRP1 in Liver Disease. Int. Rev. Cytol. 2017, 79, 1–23. [Google Scholar] [CrossRef]
  28. Balmer, M.L.; Joneli, J.; Schoepfer, A.; Stickel, F.; Thormann, W.; Dufour, J.-F. Significance of serum adiponectin levels in patients with chronic liver disease. Clin. Sci. 2010, 119, 431–436. [Google Scholar] [CrossRef] [Green Version]
  29. Silva, T.; Colombo, G.; Schiavon, L.L. Adiponectin: A multitasking player in the field of liver diseases. Diabetes Metab. 2014, 40, 95–107. [Google Scholar] [CrossRef]
  30. Polyzos, S.A.; Toulis, K.A.; Goulis, D.G.; Zavos, C.; Kountouras, J. Serum total adiponectin in nonalcoholic fatty liver disease: A systematic review and meta-analysis. Metabolism 2011, 60, 313–326. [Google Scholar] [CrossRef]
  31. Combs, T.P.; Marliss, E.B. Adiponectin signaling in the liver. Rev. Endocr. Metab. Disord. 2013, 15, 137–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Shehzad, A.; Iqbal, W.; Shehzad, O.; Lee, Y.S. Adiponectin: Regulation of its production and its role in human diseases. Hormones 2012, 11, 8–20. [Google Scholar] [CrossRef]
  33. Moschen, A.R.; Wieser, V.; Tilg, H. Adiponectin: Key Player in the Adipose Tissue-Liver Crosstalk. Curr. Med. Chem. 2012, 19, 5467–5473. [Google Scholar] [CrossRef] [PubMed]
  34. Ishtiaq, S.M.; Rashid, H.; Hussain, Z.; Arshad, M.I.; Khan, J.A. Adiponectin and PPAR: A setup for intricate crosstalk between obesity and non-alcoholic fatty liver disease. Rev. Endocr. Metab. Disord. 2019, 20, 253–261. [Google Scholar] [CrossRef]
  35. Duntas, L.H.; Popovic, V.; Panotopoulos, G. Adiponectin: Novelties in Metabolism and Hormonal Regulation. Nutr. Neurosci. 2004, 7, 195–200. [Google Scholar] [CrossRef]
  36. Tilg, H. The Role of Cytokines in Non-Alcoholic Fatty Liver Disease. Dig. Dis. 2010, 28, 179–185. [Google Scholar] [CrossRef]
  37. Bechmann, L.P.; Kocabayoglu, P.; Sowa, J.-P.; Sydor, S.; Best, J.; Schlattjan, M.; Beilfuss, A.; Schmitt, J.; Hannivoort, R.A.; Kilicarslan, A.; et al. Free fatty acids repress small heterodimer partner (SHP) activation and adiponectin counteracts bile acid-induced liver injury in superobese patients with nonalcoholic steatohepatitis. Hepatoloy 2013, 57, 1394–1406. [Google Scholar] [CrossRef]
  38. Liu, J.; Xing, J.; Wang, B.; Wei, C.; Yang, R.; Zhu, Y.; Qiu, H. Correlation Between Adiponectin Gene rs1501299 Polymorphism and Nonalcoholic Fatty Liver Disease Susceptibility: A Systematic Review and Meta-Analysis. Med. Sci. Monit. 2019, 25, 1078–1086. [Google Scholar] [CrossRef] [PubMed]
  39. Chang, M.-L.; Hsu, C.-M.; Tseng, J.-H.; Tsou, Y.-H.; Chen, S.-C.; Shiau, S.-S.; Yeh, C.-T.; Chiu, C.-T. Plasminogen activator inhibitor-1 is independently associated with non-alcoholic fatty liver disease whereas leptin and adiponectin vary between genders. J. Gastroenterol. Hepatol. 2015, 30, 329–336. [Google Scholar] [CrossRef]
  40. Polyzos, S.A.; Kountouras, J.; Mantzoros, C.S. Adipokines in nonalcoholic fatty liver disease. Metabolism 2016, 65, 1062–1079. [Google Scholar] [CrossRef] [PubMed]
  41. Lim, J.K.; Nguyen, M.H.; Kim, W.R.; Gish, R.; Perumalswami, P.; Jacobson, I.M. Prevalence of Chronic Hepatitis B Virus Infection in the United States. Am. J. Gastroenterol. 2020, 115, 1429–1438. [Google Scholar] [CrossRef]
  42. Duraisamy, G.S.; Bhosale, D.; Lipenská, I.; Huvarova, I.; Růžek, D.; Windisch, M.P.; Miller, A.D. Advanced Therapeutics, Vaccinations, and Precision Medicine in the Treatment and Management of Chronic Hepatitis B Viral Infections; Where Are We and Where Are We Going? Viruses 2020, 12, 998. [Google Scholar] [CrossRef]
  43. Durantel, D.; Zoulim, F. New antiviral targets for innovative treatment concepts for hepatitis B virus and hepatitis delta virus. J. Hepatol. 2016, 64, S117–S131. [Google Scholar] [CrossRef]
  44. Mousa, N.; Abdel-Razik, A.; Sheta, T.; Shabana, W.; Zakaria, S.; Awad, M.; Abdelsalam, M.; El-Wakeel, N.; Elkashef, W.; Effat, N.; et al. Serum leptin and homeostasis model assessment-IR as novel predictors of early liver fibrosis in chronic hepatitis B virus infection. Br. J. Biomed. Sci. 2018, 75, 192–196. [Google Scholar] [CrossRef] [PubMed]
  45. Manolakopoulos, S.; Bethanis, S.; Liapi, C.; Stripeli, F.; Sklavos, P.; Margeli, A.; Christidou, A.; Katsanika, A.; Vogiatzakis, E.; Tzourmakliotis, D.; et al. An assessment of serum leptin levels in patients with chronic viral hepatitis: A prospective study. BMC Gastroenterol. 2007, 7, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Zografos, T.; Rigopoulou, E.I.; Liaskos, C.; Togousidis, E.; Zachou, K.; Gatselis, N.; Germenis, A.; Dalekos, G.N. Alterations of leptin during IFN-α therapy in patients with chronic viral hepatitis. J. Hepatol. 2006, 44, 848–855. [Google Scholar] [CrossRef] [PubMed]
  47. Ataseven, H.; Bahcecioglu, I.H.; Kuzu, N.; Yalniz, M.; Celebi, S.; Erensoy, A.; Ustündağ, B. The Levels of Ghrelin, Leptin, TNF-α, and IL-6 in Liver Cirrhosis and Hepatocellular Carcinoma due to HBV and HDV Infection. Mediat. Inflamm. 2006, 2006, 078380. [Google Scholar] [CrossRef] [Green Version]
  48. Yoon, S.; Jung, J.; Kim, T.; Park, S.; Chwae, Y.-J.; Shin, H.-J.; Kim, K. Adiponectin, a downstream target gene of peroxisome proliferator-activated receptor γ, controls hepatitis B virus replication. Viroloy 2011, 409, 290–298. [Google Scholar] [CrossRef] [Green Version]
  49. Chiang, C.-H.; Lai, J.-S.; Hung, S.-H.; Lee, L.-T.; Sheu, J.-C.; Huang, K.-C. Serum adiponectin levels are associated with hepatitis B viral load in overweight to obese hepatitis B virus carriers. Obesity 2013, 21, 291–296. [Google Scholar] [CrossRef]
  50. Mohamadkhani, A.; Sayehmiri, K.; Ghanbari, R.; Elahi, E.; Poustchi, H.; Montazeri, G. The inverse association of serum HBV DNA level with HDL and adiponectin in chronic hepatitis B infection. Virol. J. 2010, 7, 228. [Google Scholar] [CrossRef] [Green Version]
  51. Lu, J.-Y.; Su, T.-C.; Liu, Y.-H.; Hsu, H.-J.; Chen, C.-L.; Yang, W.-S. Lower plasma adiponectin is correlated to higher alanine aminotransferase independent of metabolic factors and hepatitis B virus carrier status. Intern. Med. J. 2007, 37, 365–371. [Google Scholar] [CrossRef]
  52. Lu, J.-Y.; Chuang, L.-M.; Yang, W.-S.; Tai, T.-Y.; Lai, M.-Y.; Chen, P.-J.; Kao, J.-H.; Lee, C.-Z.; Lee, H.-S. Adiponectin levels among patients with chronic hepatitis B and C infections and in response to IFN-alpha therapy. Liver Int. 2005, 25, 752–759. [Google Scholar] [CrossRef]
  53. Chen, C.-L.; Yang, W.-S.; Yang, H.-I.; You, S.-L.; Wang, L.-Y.; Lu, S.-N.; Liu, C.-J.; Kao, J.-H.; Chen, P.-J.; Chen, D.-S.; et al. Plasma Adipokines and Risk of Hepatocellular Carcinoma in Chronic Hepatitis B Virus-Infected Carriers: A Prospective Study in Taiwan. Cancer Epidemiol. Biomarkers Prev. 2014, 23, 1659–1671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Meng, Z.; Zhang, Y.; Wei, Z.; Liu, P.; Kang, J.; Zhang, Y.; Ma, D.; Ke, C.; Chen, Y.; Luo, J.; et al. High serum resistin associates with intrahepatic inflammation and necrosis: An index of disease severity for patients with chronic HBV infection. BMC Gastroenterol. 2017, 17, 6. [Google Scholar] [CrossRef] [Green Version]
  55. Durazzo, M.; Belci, P.; Niro, G.A.; Collo, A.; Grisoglio, E.; Ambrogio, V.; Spandre, M.; Fontana, R.; Gambino, R.; Cassader, M.; et al. Variations of serum levels of adiponectin and resistin in chronic viral hepatitis. J. Endocrinol. Investig. 2013, 36, 600–605. [Google Scholar]
  56. Yuksel, E.; Akbal, E.; Koçak, E.; Akyürek, Ö.; Köklü, S.; Ekiz, F.; Yılmaz, B.; Yilmaz, B. The relationship between visfatin, liver inflammation, and acute phase reactants in chronic viral hepatitis B. Wien. Klin. Wochenschr. 2015, 128, 658–662. [Google Scholar] [CrossRef]
  57. Haberl, E.M.; Feder, S.; Pohl, R.; Rein-Fischboeck, L.; Dürholz, K.; Eichelberger, L.; Wanninger, J.; Weiss, T.S.; Buechler, C. Chemerin Is Induced in Non-Alcoholic Fatty Liver Disease and Hepatitis B-Related Hepatocellular Carcinoma. Cancers 2020, 12, 2967. [Google Scholar] [CrossRef]
  58. Hsu, C.-S.; Liu, W.-L.; Chao, Y.-C.; Lin, H.H.; Tseng, T.-C.; Wang, C.-C.; Chen, D.-S.; Kao, J.-H. Adipocytokines and liver fibrosis stages in patients with chronic hepatitis B virus infection. Hepatol. Int. 2015, 9, 231–242. [Google Scholar] [CrossRef]
  59. Borgia, S.M.; Hedskog, C.; Parhy, B.; Hyland, R.H.; Stamm, L.M.; Brainard, D.M.; Subramanian, G.M.; McHutchison, J.G.; Mo, H.; Svarovskaia, E.; et al. Identification of a Novel Hepatitis C Virus Genotype From Punjab, India: Expanding Classification of Hepatitis C Virus Into 8 Genotypes. J. Infect. Dis. 2018, 218, 1722–1729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Spearman, C.W.; Dusheiko, G.M.; Hellard, M. Hepatitis C. Lancet 2019, 394, 1451–1466. [Google Scholar] [CrossRef]
  61. Chang, M.-L. Metabolic alterations and hepatitis C: From bench to bedside. World J. Gastroenterol. 2016, 22, 1461–1476. [Google Scholar] [CrossRef]
  62. Chang, M.-L.; Chen, T.-H.; Hsu, C.-M.; Lin, C.-H.; Kuo, C.-J.; Huang, S.-W.; Chen, C.-W.; Cheng, H.-T.; Yeh, C.-T.; Chiu, C.-T. The Evolving Interplay among Abundant Adipokines in Patients with Hepatitis C during Viral Clearance. Nutrients 2017, 9, 570. [Google Scholar] [CrossRef]
  63. Thompson, A.J.; Muir, A.J.; Sulkowski, M.S.; Ge, D.; Fellay, J.; Shianna, K.V.; Urban, T.; Afdhal, N.H.; Jacobson, I.M.; Esteban, R.; et al. Interleukin-28B Polymorphism Improves Viral Kinetics and Is the Strongest Pretreatment Predictor of Sustained Virologic Response in Genotype 1 Hepatitis C Virus. Gastroenteroloy 2010, 139, 120–129.e18. [Google Scholar] [CrossRef] [Green Version]
  64. Pawlotsky, J.-M.; Feld, J.J.; Zeuzem, S.; Hoofnagle, J.H. From non-A, non-B hepatitis to hepatitis C virus cure. J. Hepatol. 2015, 62, S87–S99. [Google Scholar] [CrossRef] [Green Version]
  65. Liu, Z.; Zhang, N.; Han, Q.-Y.; Zeng, J.-T.; Chu, Y.-L.; Qiu, J.-M.; Wang, Y.-W.; Ma, L.-T.; Wang, X.-Q. Correlation of serum leptin levels with anthropometric and metabolic parameters and biochemical liver function in Chinese patients with chronic hepatitis C virus infection. World J. Gastroenterol. 2005, 11, 3357–3362. [Google Scholar] [CrossRef]
  66. El-Gindy, E.M.; Ali-Eldin, F.A.; Meguid, A.M. Serum leptin level and its association with fatigue in patients with chronic hepatitis C virus infection. Arab. J. Gastroenterol. 2012, 13, 54–57. [Google Scholar] [CrossRef]
  67. Cua, I.H.Y.; Hui, J.M.; Bandara, P.; Kench, J.G.; Farrell, G.C.; McCaughan, G.W.; George, J. Insulin resistance and liver injury in hepatitis C is not associated with virus-specific changes in adipocytokines. Hepatology 2007, 46, 66–73. [Google Scholar] [CrossRef]
  68. Giannini, E.; Ceppa, P.; Botta, F.; Mastracci, L.; Romagnoli, P.; Comino, I.; Pasini, A.; Risso, D.; BLantieri, P.; Icardi, G.; et al. Leptin has no role in determining severity of steatosis and fibrosis in patients with chronic hepatitis C. Am. J. Gastroenterol. 2000, 95, 3211–3217. [Google Scholar] [CrossRef]
  69. Hickman, I.J.; Powell, E.; Prins, J.; Clouston, A.D.; Ash, S.; Purdie, D.M.; Jonsson, J.R. In overweight patients with chronic hepatitis C circulating insulin is associated with hepatic fibrosis: Implications for therapy. J. Hepatol. 2003, 39, 1042–1048. [Google Scholar] [CrossRef]
  70. Romero-Gómez, M.; Castellano-Megias, V.M.; Grande, L. Serum leptin levels correlate with hepatic steatosis in chronic hepatitis C. Am. J. Gastroenterol. 2003, 98, 1135–1141. [Google Scholar] [CrossRef]
  71. Saad, Y.; Ahmed, A.; Saleh, D.A.; Doss, W. Adipokines and insulin resistance, predictors of response to therapy in Egyptian patients with chronic hepatitis C virus genotype 4. Eur. J. Gastroenterol. Hepatol. 2013, 25, 920–925. [Google Scholar] [CrossRef] [PubMed]
  72. Eguchi, Y.; Mizuta, T.; Yasutake, T.; Hisatomi, A.; Iwakiri, R.; Ozaki, I.; Fujimoto, K. High serum leptin is an independent risk factor for non-response patients with low viremia to antiviral treatment in chronic hepatitis C. World J. Gastroenterol. 2006, 12, 556–560. [Google Scholar] [CrossRef]
  73. Chang, M.-L.; Kuo, C.-J.; Huang, H.-C.; Chu, Y.-Y.; Chiu, C.-T. Association between Leptin and Complement in Hepatitis C Patients with Viral Clearance: Homeostasis of Metabolism and Immunity. PLoS ONE 2016, 11, e0166712. [Google Scholar] [CrossRef]
  74. Canavesi, E.; Porzio, M.; Ruscica, M.; Rametta, R.; Macchi, C.; Pelusi, S.; Fracanzani, A.L.; Dongiovanni, P.; Fargion, S.; Magni, P.; et al. Increased circulating adiponectin in males with chronic HCV hepatitis. Eur. J. Intern. Med. 2015, 26, 635–639. [Google Scholar] [CrossRef]
  75. Khattab, M.A.; Eslam, M.; Aly, M.M.; Shatat, M.; Hussen, A.; Moussa, Y.I.; Elsaghir, G.; Abdalhalim, H.; Aly, A.; Gaber, S.; et al. Association of Serum Adipocytokines With Insulin Resistance and Liver Injury in Patients With Chronic Hepatitis C Genotype 4. J. Clin. Gastroenterol. 2012, 46, 871–879. [Google Scholar] [CrossRef]
  76. Corbetta, S.; Redaelli, A.; Pozzi, M.; Bovo, G.; Ratti, L.; Redaelli, E.; Pellegrini, C.; Beck-Peccoz, P.; Spada, A. Fibrosis is associated with adiponectin resistance in chronic hepatitis C virus infection. Eur. J. Clin. Investig. 2011, 41, 898–905. [Google Scholar] [CrossRef] [PubMed]
  77. Hung, C.-H.; Lee, C.-M.; Chen, C.-H.; Hu, T.-H.; Jiang, S.-R.; Wang, J.-H.; Lu, S.-N.; Wang, P.-W. Association of inflammatory and anti-inflammatory cytokines with insulin resistance in chronic hepatitis C. Liver Int. 2009, 29, 1086–1093. [Google Scholar] [CrossRef] [PubMed]
  78. Wedemeyer, I.; Bechmann, L.P.; Odenthal, M.; Jochum, C.; Marquitan, G.; Drebber, U.; Gerken, G.; Gieseler, R.K.; Dienes, H.P.; Canbay, A. Adiponectin inhibits steatotic CD95/Fas up-regulation by hepatocytes: Therapeutic implications for hepatitis C. J. Hepatol. 2009, 50, 140–149. [Google Scholar] [CrossRef]
  79. Durante-Mangoni, E.; Zampino, R.; Marrone, A.; Tripodi, M.-F.; Rinaldi, L.; Restivo, L.; Cioffi, M.; Ruggiero, G.; Adinolfi, L.E. Hepatic steatosis and insulin resistance are associated with serum imbalance of adiponectin/tumour necrosis factor-? in chronic hepatitis C patients. Aliment. Pharmacol. Ther. 2006, 24, 1349–1357. [Google Scholar] [CrossRef]
  80. Liu, C.-J.; Chen, P.-J.; Jeng, Y.-M.; Huang, W.-L.; Yang, W.-S.; Lai, M.-Y.; Kao, J.-H.; Chen, D.-S. Serum adiponectin correlates with viral characteristics but not histologic features in patients with chronic hepatitis C. J. Hepatol. 2005, 43, 235–242. [Google Scholar] [CrossRef]
  81. Derbala, M.; Rizk, N.M.; Al-Kaabi, S.; Amer, A.; Shebl, F.; Al Marri, A.; Aigha, I.; Alyaesi, D.; Mohamed, H.; Aman, H.; et al. Adiponectin changes in HCV-Genotype 4: Relation to liver histology and response to treatment. J. Viral Hepat. 2009, 16, 689–696. [Google Scholar] [CrossRef]
  82. Shah, S.R.; Patel, K.; Marcellin, P.; Foster, G.R.; Manns, M.; Kottilil, S.; Healey, L.; Pulkstenis, E.; Subramanian, G.M.; McHutchison, J.G.; et al. Steatosis Is an Independent Predictor of Relapse Following Rapid Virologic Response in Patients With HCV Genotype 3. Clin. Gastroenterol. Hepatol. 2011, 9, 688–693. [Google Scholar] [CrossRef] [Green Version]
  83. Wang, A.Y.-H.; Hickman, I.J.; Richards, A.A.; Whitehead, J.P.; Prins, J.; Macdonald, G.A. High Molecular Weight Adiponectin Correlates with Insulin Sensitivity in Patients with Hepatitis C Genotype 3, But Not Genotype 1 Infection. Am. J. Gastroenterol. 2005, 100, 2717–2723. [Google Scholar] [CrossRef]
  84. Palmer, C.; Hampartzoumian, T.; Lloyd, A.R.; Zekry, A. A novel role for adiponectin in regulating the immune responses in chronic hepatitis C virus infection. Hepatology 2008, 48, 374–384. [Google Scholar] [CrossRef]
  85. Jonsson, J.R.; Moschen, A.R.; Hickman, I.J.; Richardson, M.M.; Kaser, S.; Clouston, A.D.; Powell, E.; Tilg, H. Adiponectin and its receptors in patients with chronic hepatitis C. J. Hepatol. 2005, 43, 929–936. [Google Scholar] [CrossRef]
  86. Korah, T.E.; El-Sayed, S.; Elshafie, M.K.; Hammoda, E.G.; Safan, A.M. Significance of serum leptin and adiponectin levels in Egyptian patients with chronic hepatitis C virus associated hepatic steatosis and fibrosis. World J. Hepatol. 2013, 5, 74–81. [Google Scholar] [CrossRef]
  87. Latif, H.A.; Assal, H.S.; Mahmoud, M.; Rasheed, W.I. Role of serum adiponectin level in the development of liver cirrhosis in patients with hepatitis C virus. Clin. Exp. Med. 2010, 11, 123–129. [Google Scholar] [CrossRef] [PubMed]
  88. Ashour, E.; Samy, N.; Sayed, M.; Imam, A. The relationship between serum adiponectin and steatosis in patients with chronic hepatitis C genotype-4. Clin. Lab. 2010, 56, 103. [Google Scholar]
  89. Petit, J.-M.; Minello, A.; Jooste, V.; Bour, J.B.; Galland, F.; Duvillard, L.; Verges, B.; Olsson, N.O.; Gambert, P.; Hillon, P. Decreased Plasma Adiponectin Concentrations Are Closely Related to Steatosis in Hepatitis C Virus-Infected Patients. J. Clin. Endocrinol. Metab. 2005, 90, 2240–2243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Lago, F.; Diéguez, C.; Gómez-Reino, J.; Gualillo, O. Adipokines as emerging mediators of immune response and inflammation. Nat. Clin. Pract. Rheumatol. 2007, 3, 716–724. [Google Scholar] [CrossRef] [PubMed]
  91. Chang, M.-L.; Yeh, H.-C.; Tsou, Y.-K.; Wang, C.-J.; Cheng, H.-Y.; Sung, C.-M.; Ho, Y.-P.; Chen, T.-H.; Yeh, C.-T. HCV Core-Induced Nonobese Hepatic Steatosis Is Associated With Hypoadiponectinemia and Is Ameliorated by Adiponectin Administration. Obesity 2012, 20, 1474–1480. [Google Scholar] [CrossRef]
  92. Zografos, T.; Liaskos, C.; Rigopoulou, E.I.; Togousidis, E.; Makaritsis, K.; Germenis, A.; Dalekos, G.N. Adiponectin: A New Independent Predictor of Liver Steatosis and Response to IFN-α Treatment in Chronic Hepatitis, C. Am. J. Gastroenterol. 2008, 103, 605–614. [Google Scholar] [CrossRef] [PubMed]
  93. Chang, M.-L.; Kuo, C.-J.; Pao, L.-H.; Hsu, C.-M.; Chiu, C.-T. The evolving relationship between adiponectin and insulin sensitivity in hepatitis C patients during viral clearance. Virulence 2017, 8, 1255–1264. [Google Scholar] [CrossRef] [Green Version]
  94. Fartoux, L.; Poujol-Robert, A.; Guéchot, J.; Wendum, D.; Poupon, R.; Serfaty, L. Insulin resistance is a cause of steatosis and fibrosis progression in chronic hepatitis C. Gut 2005, 54, 1003–1008. [Google Scholar] [CrossRef] [Green Version]
  95. Mihm, S. Hepatitis C Virus, Diabetes and Steatosis: Clinical Evidence in Favor of a Linkage and Role of Genotypes. Dig. Dis. 2010, 28, 280–284. [Google Scholar] [CrossRef]
  96. Bastard, J.-P.; Fellahi, S.; Audureau, E.; Layese, R.; Roudot-Thoraval, F.; Cagnot, C.; Mahuas-Bourcier, V.; Sutton, A.; Ziol, M.; Capeau, J.; et al. Elevated adiponectin and sTNFRII serum levels can predict progression to hepatocellular carcinoma in patients with compensated HCV1 cirrhosis. Eur. Cytokine Netw. 2018, 29, 112–120. [Google Scholar] [CrossRef]
  97. Shen, J.; Yeh, C.-C.; Wang, Q.; Gurvich, I.; Siegel, A.B.; Santella, R.M. Plasma Adiponectin and Hepatocellular Carcinoma Survival Among Patients Without Liver Transplantation. Anticancer Res. 2016, 36, 5307–5314. [Google Scholar] [CrossRef] [Green Version]
  98. Radwan, H.A.; Elsayed, E.H.; Saleh, O.M.; Hamed, E.H. Significance of Serum Adiponectin and Insulin Resistance Levels in Diagnosis of Egyptian Patients with Chronic Liver Disease and HCC. Asian Pac. J. Cancer Prev. 2019, 20, 1833–1839. [Google Scholar] [CrossRef] [Green Version]
  99. Hamdy, K.; Al Swaff, R.; Hussein, H.A.; Gamal, M. Assessment of serum adiponectin in Egyptian patients with HCV-related cirrhosis and hepatocellular carcinoma. J. Endocrinol. Investig. 2015, 38, 1225–1231. [Google Scholar] [CrossRef]
  100. Sumie, S.; Kawaguchi, T.; Kuromatsu, R.; Takata, A.; Nakano, M.; Satani, M.; Yamada, S.; Niizeki, T.; Torimura, T.; Sata, M. Total and High Molecular Weight Adiponectin and Hepatocellular Carcinoma with HCV Infection. PLoS ONE 2011, 6, e26840. [Google Scholar] [CrossRef] [Green Version]
  101. Nakagawa, H.; Fujiwara, N.; Tateishi, R.; Arano, T.; Nakagomi, R.; Kondo, M.; Minami, T.; Sato, M.; Uchino, K.; Enooku, K.; et al. Impact of serum levels of interleukin-6 and adiponectin on all-cause, liver-related, and liver-unrelated mortality in chronic hepatitis C patients. J. Gastroenterol. Hepatol. 2015, 30, 379–388. [Google Scholar] [CrossRef]
  102. Miki, D.; Ohishi, W.; Ochi, H.; Hayes, C.N.; Abe, H.; Tsuge, M.; Imamura, M.; Kamatani, N.; Nakamura, Y.; Chayama, K. Serum PAI-1 is a novel predictor for response to pegylated interferon-α-2b plus ribavirin therapy in chronic hepatitis C virus infection. J. Viral Hepat. 2011, 19, e126–e133. [Google Scholar] [CrossRef] [PubMed]
  103. Chang, M.-L.; Lin, Y.-S.; Pao, L.-H.; Huang, H.-C.; Chiu, C.-T. Link between plasminogen activator inhibitor-1 and cardiovascular risk in chronic hepatitis C after viral clearance. Sci. Rep. 2017, 7, 42503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Kukla, M.; Zwirska-Korczala, K.; Gabriel, A.; Waluga, M.; Warakomska, I.; Berdowska, A.; Rybus-Kalinowska, B.; Kalinowski, M.; Janczewska, E.; Wozniak-Grygiel, E.; et al. Visfatin serum levels in chronic hepatitis C patients. J. Viral Hepat. 2010, 17, 254–260. [Google Scholar] [CrossRef]
  105. Kukla, M.; Zalewska-Ziob, M.; Adamek, B.; Kasperczyk, J.; Bułdak, R.J.; Sawczyn, T.; Stygar, D.; Sobala-Szczygieł, B.; Stachowska, M.; Gabriel, A.; et al. Visfatin serum concentration and hepatic mRNA expression in chronic hepatitis C. Clin. Exp. Hepatol. 2019, 5, 147–154. [Google Scholar] [CrossRef] [PubMed]
  106. Chen, L.; Liu, W.; Lai, S.; Li, Y.; Wang, X.; Zhang, H. Insulin resistance, serum visfatin, and adiponectin levels are associated with metabolic disorders in chronic hepatitis C virus-infected patients. Eur. J. Gastroenterol. Hepatol. 2013, 25, 935–941. [Google Scholar] [CrossRef]
  107. Huang, J.; Huang, C.-F.; Yu, M.; Dai, C.; Huang, C.-I.; Yeh, M.-L.; Hsieh, M.-H.; Yang, J.-F.; Hsieh, M.-Y.; Lin, Z.-Y.; et al. Serum visfatin is correlated with disease severity and metabolic syndrome in chronic hepatitis C infection. J. Gastroenterol. Hepatol. 2010, 26, 530–535. [Google Scholar] [CrossRef]
  108. Tsai, I.-T.; Wang, C.-P.; Yu, T.-H.; Lu, Y.-C.; Lin, C.-W.; Lu, L.-F.; Wu, C.-C.; Chung, F.-M.; Lee, Y.-J.; Hung, W.-C.; et al. Circulating visfatin level is associated with hepatocellular carcinoma in chronic hepatitis B or C virus infection. Cytokine 2017, 90, 54–59. [Google Scholar] [CrossRef]
  109. El-Daly, U.M.; Saber, M.M.; Abdellateif, M.S.; Nassar, H.R.; Namour, E.A.; Ismail, Y.M.; Zekri, A.-R.N. The Possible Role of Adipokines in HCV Associated Hepatocellular Carcinoma. Asian Pac. J. Cancer Prev. 2020, 21, 599–609. [Google Scholar] [CrossRef]
  110. Huang, J.-F.; Dai, C.-Y.; Yu, M.-L.; Shin, S.; Hsieh, M.-Y.; Huang, C.-F.; Lee, L.-P.; Lin, K.-D.; Lin, Z.-Y.; Chen, S.-C.; et al. Serum retinol-binding protein 4 is inversely correlated with disease severity of chronic hepatitis C. J. Hepatol. 2009, 50, 471–478. [Google Scholar] [CrossRef]
  111. Qin, S.; Zhou, Y.; Lok, A.S.; Tsodikov, A.; Yan, X.; Gray, L.; Yuan, M.; Moritz, R.L.; Galas, D.; Omenn, G.S.; et al. SRM targeted proteomics in search for biomarkers of HCV-induced progression of fibrosis to cirrhosis in HALT-C patients. Proteomics 2012, 12, 1244–1252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Kataria, Y.; Deaton, R.J.; Enk, E.; Jin, M.; Petrauskaite, M.; Dong, L.; Goldenberg, J.R.; Cotler, S.J.; Jensen, D.M.; Van Breemen, R.B.; et al. Retinoid and carotenoid status in serum and liver among patients at high-risk for liver cancer. BMC Gastroenterol. 2016, 16, 1–12. [Google Scholar] [CrossRef] [Green Version]
  113. Gouthamchandra, K.; Kumar, A.; Shwetha, S.; Mukherjee, A.; Chandra, M.; Ravishankar, B.; Khaja, M.N.; Sadhukhan, P.C.; Das, S. Serum proteomics of hepatitis C virus infection reveals retinol-binding protein 4 as a novel regulator. J. Gen. Virol. 2014, 95, 1654–1667. [Google Scholar] [CrossRef]
  114. Iwasa, M.; Hara, N.; Miyachi, H.; Tanaka, H.; Takeo, M.; Fujita, N.; Kobayashi, Y.; Kojima, Y.; Kaito, M.; Takei, Y. Patients achieving clearance of HCV with interferon therapy recover from decreased retinol-binding protein 4 levels. J. Viral Hepat. 2009, 16, 716–723. [Google Scholar] [CrossRef]
  115. Tiftikçi, A.; Atug, O.; Yilmaz, Y.; Eren, F.; Ozdemir, F.T.; Yapali, S.; Özdoğan, O.; Celikel, C.A.; Imeryuz, N.; Tözün, N.; et al. Serum Levels of Adipokines in Patients with Chronic HCV Infection: Relationship with Steatosis and Fibrosis. Arch. Med. Res. 2009, 40, 294–298. [Google Scholar] [CrossRef] [PubMed]
  116. Baranova, A.; Jarrar, M.; Stepanova, M.; Johnson, A.; Rafiq, N.; Gramlich, T.; Chandhoke, V.; Younossi, Z.M. Association of Serum Adipocytokines with Hepatic Steatosis and Fibrosis in Patients with Chronic Hepatitis, C. Digestion 2010, 83, 32–40. [Google Scholar] [CrossRef]
  117. Marra, F.; Bertolani, C. Adipokines in liver diseases. Hepatoloy 2009, 50, 957–969. [Google Scholar] [CrossRef]
  118. Sjöwall, C.; Cardell, K.; Boström, E.A.; Bokarewa, M.; Enocsson, H.; Ekstedt, M.; Lindvall, L.; Frydén, A.; Almer, S. Highprevalence of autoantibodies to C-reactive protein in patients with chronic hepatitis C infection: Association with liver fibrosis and portal inflammation. Hum. Immunol. 2012, 73, 382–388. [Google Scholar] [CrossRef] [Green Version]
  119. Ibrahim, D.M.; Shaaban, E.S.E.; Fouad, T.A. Circulating Resistin Is Associated with Plasma Glucagon-Like Peptide-1 in Cirrhotic Patients with Hepatitis C Virus Genotype-4 Infection. Endocr. Res. 2019, 45, 17–23. [Google Scholar] [CrossRef]
  120. Chang, M.-L.; Liang, K.-H.; Ku, C.-L.; Lo, C.-C.; Cheng, Y.-T.; Hsu, C.-M.; Yeh, C.-T.; Chiu, C.-T. Resistin reinforces interferon λ-3 to eliminate hepatitis C virus with fine-tuning from RETN single-nucleotide polymorphisms. Sci. Rep. 2016, 6, 30799. [Google Scholar] [CrossRef] [Green Version]
  121. Elsayed, E.Y.; Mosalam, N.A.; Mohamed, N.R. Resistin and Insulin Resistance: A Link Between Inflammation and Hepatocarcinogenesis. Asian Pac. J. Cancer Prev. 2015, 16, 7139–7142. [Google Scholar] [CrossRef] [Green Version]
  122. Kukla, M.; Zwirska-Korczala, K.; Gabriel, A.; Waluga, M.; Warakomska, I.; Szczygiel, B.; Berdowska, A.; Mazur, W.; Wozniak-Grygiel, E.; Kryczka, W. Chemerin, vaspin and insulin resistance in chronic hepatitis C. J. Viral Hepat. 2009, 17, 661–667. [Google Scholar] [CrossRef]
  123. Kukla, M.; Adamek, B.; Waluga, M.; Zalewska-Ziob, M.; Kasperczyk, J.; Gabriel, A.; Mazur, W.; Sobala-Szczygieł, B.; Bułdak, R.J.; Zajecki, W.; et al. HepaticChemerinandChemokine-Like Receptor 1Expression in Patients with Chronic Hepatitis C. BioMed Res. Int. 2014, 2014, 1–12. [Google Scholar] [CrossRef] [Green Version]
  124. Nkontchou, G.; Bastard, J.-P.; Ziol, M.; Aout, M.; Cosson, E.; Ganne-Carrié, N.; Grando-Lemaire, V.; Roulot, D.; Capeau, J.; Trinchet, J.-C.; et al. Insulin resistance, serum leptin, and adiponectin levels and outcomes of viral hepatitis C cirrhosis. J. Hepatol. 2010, 53, 827–833. [Google Scholar] [CrossRef] [PubMed]
  125. Lin, C.-Y.; Cheng, Y.-T.; Chang, M.-L.; Chien, R.-N. The extrahepatic events of Asian patients with primary biliary cholangitis: A 30-year cohort study. Sci. Rep. 2019, 9, 7577. [Google Scholar] [CrossRef]
  126. Breidert, M.; Zimmermann, T.F.; Schneider, R.; Ehninger, G.; Brabant, G. Ghrelin/Leptin-Imbalance in Patients with Primary Biliary Cirrhosis. Exp. Clin. Endocrinol. Diabetes 2004, 112, 123–126. [Google Scholar] [CrossRef]
  127. Floreani, A.; Variola, A.; Niro, G.A.; Premoli, A.; Baldo, V.; Gambino, R.; Musso, G.; Cassader, M.; Bossa, F.; Ferrara, F.; et al. Plasma Adiponectin Levels in Primary Biliary Cirrhosis: A Novel Perspective for Link Between Hypercholesterolemia and Protection Against Atherosclerosis. Am. J. Gastroenterol. 2008, 103, 1959–1965. [Google Scholar] [CrossRef] [PubMed]
  128. Rieger, R.; Oertelt, S.; Selmi, C.; Invernizzi, P.; Podda, M.; Gershwin, M.E.; Oertelt-Prigione, S. Decreased Serum Leptin Levels in Primary Biliary Cirrhosis: A Link between Metabolism and Autoimmunity? Ann. N. Y. Acad. Sci. 2005, 1051, 211–217. [Google Scholar] [CrossRef]
  129. Szalay, F.; Folhoffer, A.; Horváth, A.; Csák, T.; Speer, G.; Nagy, Z.; Lakatos, P.L.; Horváth, C.; Habior, A.; Tornai, I. Serum leptin, soluble leptin receptor, free leptin index and bone mineral density in patients with primary biliary cirrhosis. Eur. J. Gastroenterol. Hepatol. 2005, 17, 923–928. [Google Scholar] [CrossRef]
  130. García-Suárez, C.; Crespo, J.; Fernández-Gil, P.L.; Amado, J.A.; García-Unzueta, M.T.; Pons Romero, F. Concentraciones plasmáticas de leptina en los pacientes con cirrosis biliar primaria y su relación con el grado de fibrosis [Plasma leptin levels in patients with primary biliary cirrhosis and their relationship with degree of fibrosis]. Gastroenterol. Hepatol. 2004, 27, 47–50. [Google Scholar] [CrossRef]
  131. Lohse, A.W.; Chazouillères, O.; Dalekos, G.; Drenth, J.; Heneghan, M.; Hofer, H. EASL clinical practice guidelines: Autoimmune hepatitis. J. Hepatol. 2015, 63, 971–1004. [Google Scholar]
  132. Fantuzzi, G. Adiponectin in inflammatory and immune-mediated diseases. Cytokine 2013, 64, 1–10. [Google Scholar] [CrossRef] [Green Version]
  133. Durazzo, M.; Niro, G.; Premoli, A.; Morello, E.; Rizzotto, E.R.; Gambino, R.; Bo, S.; Musso, G.; Cassader, M.; Pagano, G.; et al. Type 1 autoimmune hepatitis and adipokines: New markers for activity and disease progression? J. Gastroenterol. 2009, 44, 476–482. [Google Scholar] [CrossRef] [PubMed]
  134. Kema, V.H.; Mojerla, N.R.; Khan, I.; Mandal, P. Effect of alcohol on adipose tissue: A review on ethanol mediated adipose tissue injury. Adipocyte 2015, 4, 225–231. [Google Scholar] [CrossRef] [Green Version]
  135. Zhou, Y.; Yuan, G.; Zhong, F.; He, S. Roles of the complement system in alcohol-induced liver disease. Clin. Mol. Hepatol. 2020, 26, 677–685. [Google Scholar] [CrossRef]
  136. Steiner, J.L.; Lang, C.H. Alcohol, Adipose Tissue and Lipid Dysregulation. Biomolecules 2017, 7, 16. [Google Scholar] [CrossRef]
  137. Proskynitopoulos, P.J.; Rhein, M.; Jäckel, E.; Manns, M.P.; Frieling, H.; Bleich, S.; Thum, T.; Hillemacher, T.; Glahn, A. Corrigendum: Leptin Expression and Gene Methylation Patterns in Alcohol-Dependent Patients with Ethyltoxic Cirrhosis-Normalization After Liver Transplantation and Implications for Future Research. Alcohol. Alcohol. 2018, 53, 760. [Google Scholar] [CrossRef] [PubMed]
  138. Buechler, C.; Schäffler, A.; Johann, M.; Neumeier, M.; Kohl, P.; Weiss, T.S.; Wodarz, N.; Kiefer, P.; Hellerbrand, C. Elevated adiponectin serum levels in patients with chronic alcohol abuse rapidly decline during alcohol withdrawal. J. Gastroenterol. Hepatol. 2009, 24, 558–563. [Google Scholar] [CrossRef]
  139. Da Silva, T.E.; Costa-Silva, M.; Correa, C.G.; DeNardin, G.; Alencar, M.L.A.; Coelho, M.S.P.H.; Muraro-Wildner, L.; Luiza-Bazzo, M.; González-Chica, D.A.; Dantas-Correa, E.B.; et al. Clinical Significance of Serum Adiponectin and Resistin Levels in Liver Cirrhosis. Ann. Hepatol. 2018, 17, 286–299. [Google Scholar] [CrossRef]
  140. You, M.; Zhou, Z.; Daniels, M.; Jogasuria, A. Endocrine Adiponectin-FGF15/19 Axis in Ethanol-Induced Inflammation and Alcoholic Liver Injury. Gene Expr. 2018, 18, 103–113. [Google Scholar] [CrossRef]
  141. Prystupa, A.; Kiciński, P.; Luchowska-Kocot, D.; Sak, J.; Prystupa, T.; Tan, Y.-H.; Panasiuk, L.; Załuska, W. Factors influencing serum chemerin and kallistatin concentrations in patients with alcohol-induced liver cirrhosis. Ann. Agric. Environ. Med. 2019, 26, 143–147. [Google Scholar] [CrossRef]
  142. Trogen, G.; Bacon, J.; Li, Y.; Wright, G.L.; DeGroat, A.; Hagood, K.L.; Warren, Z.; Forsman, A.; Kilaru, A.; Clark, W.A.; et al. Transgenic overexpression of CTRP3 prevents alcohol-induced hepatic triglyceride accumulation. Am. J. Physiol. Metab. 2018, 315, E949–E960. [Google Scholar] [CrossRef]
  143. Gerst, F.; Wagner, R.; Oquendo, M.B.; Siegel-Axel, D.; Fritsche, A.; Heni, M.; Staiger, H.; Häring, H.-U.; Ullrich, S. What role do fat cells play in pancreatic tissue? Mol. Metab. 2019, 25, 1–10. [Google Scholar] [CrossRef]
  144. Takahashi, M.; Hori, M.; Ishigamori, R.; Mutoh, M.; Imai, T.; Nakagama, H. Fatty pancreas: A possible risk factor for pancreatic cancer in animals and humans. Cancer Sci. 2018, 109, 3013–3023. [Google Scholar] [CrossRef]
  145. Brocco, D.; Florio, R.; De Lellis, L.; Veschi, S.; Grassadonia, A.; Tinari, N.; Cama, A. The Role of Dysfunctional Adipose Tissue in Pancreatic Cancer: A Molecular Perspective. Cancers 2020, 12, 1849. [Google Scholar] [CrossRef]
  146. Rosa, S.C.D.S.; Nayak, N.; Caymo, A.M.; Gordon, J.W. Mechanisms of muscle insulin resistance and the cross-talk with liver and adipose tissue. Physiol. Rep. 2020, 8, e16407. [Google Scholar] [CrossRef]
  147. Jansson, L.; Carlsson, P. Pancreatic Blood Flow with Special Emphasis on Blood Perfusion of the Islets of Langerhans. Compr. Physiol. 2019, 9, 799–837. [Google Scholar] [CrossRef]
  148. Matafome, P.; Eickhoff, H.; Letra, L.; Seiça, R. Neuroendocrinology of Adipose Tissue and Gut–Brain Axis. Adv. Neurobiol. 2017, 19, 49–70. [Google Scholar] [CrossRef]
  149. Denroche, H.C.; Huynh, F.K.; Kieffer, T.J. The role of leptin in glucose homeostasis. J. Diabetes Investig. 2012, 3, 115–129. [Google Scholar] [CrossRef] [Green Version]
  150. Marroquí, L.; González, A.; Ñeco, P.; Caballero-Garrido, E.; Vieira, E.; Ripoll, C.; Nadal, A.; Quesada, I. Role of leptin in the pancreatic -cell: Effects and signaling pathways. J. Mol. Endocrinol. 2012, 49, R9–R17. [Google Scholar] [CrossRef] [Green Version]
  151. Kieffer, T.J.; Habener, J.F. The adipoinsular axis: Effects of leptin on pancreatic beta-cells. Am. J. Phys. Metab. 2000, 278, E1–E14. [Google Scholar] [CrossRef] [PubMed]
  152. Holz, G.G.; Chepurny, O.G.; Leech, C.A. Leptin-stimulated KATP channel trafficking: A new paradigm for β-cell stimulus-secretion coupling? Islets 2013, 5, 229–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Seufert, J. Leptin effects on pancreatic beta-cell gene expression and function. Diabetes 2004, 53, S152–S158. [Google Scholar] [CrossRef] [Green Version]
  154. Tucholski, K.; Otto-Buczkowska, E. The role of leptin in the regulation of carbohydrate metabolism. Endokrynol. Polska 2011, 62, 258–262. [Google Scholar]
  155. Gunawardana, S.C. Benefits of healthy adipose tissue in the treatment of diabetes. World J. Diabetes 2014, 5, 420. [Google Scholar] [CrossRef]
  156. Lindström, P. beta-cell function in obese-hyperglycemic mice [ob/ob Mice]. Adv. Exp. Med. Biol. 2010, 654, 463–477. [Google Scholar]
  157. Cantley, J. The control of insulin secretion by adipokines: Current evidence for adipocyte-beta cell endocrine signalling in metabolic homeostasis. Mamm. Genome 2014, 25, 442–454. [Google Scholar] [CrossRef]
  158. Arner, P. The adipocyte in insulin resistance: Key molecules and the impact of the thiazolidinediones. Trends Endocrinol. Metab. 2003, 14, 137–145. [Google Scholar] [CrossRef]
  159. Tao, C.; Sifuentes, A.; Holland, W.L. Regulation of glucose and lipid homeostasis by adiponectin: Effects on hepatocytes, pancreatic β cells and adipocytes. Best Pract. Res. Clin. Endocrinol. Metab. 2014, 28, 43–58. [Google Scholar] [CrossRef] [Green Version]
  160. Lee, Y.-H.; Magkos, F.; Mantzoros, C.S.; Kang, E.S. Effects of leptin and adiponectin on pancreatic β-cell function. Metabolism 2011, 60, 1664–1672. [Google Scholar] [CrossRef]
  161. Adeghate, E. An update on the biology and physiology of resistin. Cell. Mol. Life Sci. 2004, 61, 2485–2496. [Google Scholar] [CrossRef]
  162. 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. [Google Scholar] [CrossRef] [Green Version]
  163. Kendall, B.J.; Thrift, A.P. Unravelling the Riddle of Gastroesophageal Reflux Disease, Obesity, and Barrett’s Esophagus. Clin. Gastroenterol. Hepatol. 2015, 13, 2273–2275. [Google Scholar] [CrossRef]
  164. Chandar, A.K.; Iyer, P.G. Role of Obesity in the Pathogenesis and Progression of Barrett’s Esophagus. Gastroenterol. Clin. N. Am. 2015, 44, 249–264. [Google Scholar] [CrossRef]
  165. Kendall, B.J.; Macdonald, A.G.; Hayward, N.K.; Prins, J.B.; Brown, I.; Walker, N.; Pandeya, N.; Green, A.C.; Webb, P.M.; Whiteman, D.C.; et al. Leptin and the risk of Barrett’s oesophagus. Gut 2007, 57, 448–454. [Google Scholar] [CrossRef]
  166. Sharma, P.; Yadlapati, R. Pathophysiology and treatment options for gastroesophageal reflux disease: Looking beyond acid. Ann. N. Y. Acad. Sci. 2020. [Google Scholar] [CrossRef]
  167. Livzan, M.A.; Lapteva, I.V.; Krolevets, T.S.; Kiselev, I.E. Specific features of gastroesophageal reflux disease associated with obesity and overweight. Ter. Arkh. 2016, 88, 21–27. [Google Scholar] [CrossRef]
  168. Livzan, M.A.; Lapteva, I.V.; Krolevets, T.S. Gastroesophageal refluxed disease in persons with obesity and leptin resistance. Eksp. Klin. Gastroenterol. 2015, 3, 11–16. [Google Scholar]
  169. Thomas, S.J.; Almers, L.; Schneider, J.L.; Graham, J.L.; Havel, P.J.; Corley, D.A. Ghrelin and Leptin Have a Complex Relationship with Risk of Barrett’s Esophagus. Dig. Dis. Sci. 2015, 61, 70–79. [Google Scholar] [CrossRef] [Green Version]
  170. Abdelkader, N.A.; Montasser, I.F.; Bioumy, E.E.; Saad, W.E. Impact of anthropometric measures and serum leptin on severity of gastroesophageal reflux disease. Dis. Esophagus 2014, 28, 691–698. [Google Scholar] [CrossRef] [PubMed]
  171. Thrift, A.P.; Garcia, J.M.; El-Serag, H.B. A Multibiomarker Risk Score Helps Predict Risk for Barrett’s Esophagus. Clin. Gastroenterol. Hepatol. 2014, 12, 1267–1271. [Google Scholar] [CrossRef] [Green Version]
  172. Rubenstein, J.H.; Morgenstern, H.; McConell, D.; Scheiman, J.M.; Schoenfeld, P.; Appelman, H.; McMahon, L.F., Jr.; Kao, J.Y.; Metko, V.; Zhang, M.; et al. Associations of diabetes mellitus, insulin, leptin, and ghrelin with gastroesophageal reflux and Barrett’s esophagus. Gastroenterology 2013, 145, 1237–1244.e445. [Google Scholar] [CrossRef] [Green Version]
  173. Murata, T.; Asanuma, K.; Ara, N.; Iijima, K.; Hatta, W.; Hamada, S.; Asano, N.; Koike, T.; Imatani, A.; Masamune, A.; et al. Leptin Aggravates Reflux Esophagitis by Increasing Tissue Levels of Macrophage Migration Inhibitory Factor in Rats. Tohoku J. Exp. Med. 2018, 245, 45–53. [Google Scholar] [CrossRef] [Green Version]
  174. Ogunwobi, O.; Mutungi, G.; Beales, I.L.P. Leptin Stimulates Proliferation and Inhibits Apoptosis in Barrett’s Esophageal Adenocarcinoma Cells by Cyclooxygenase-2-Dependent, Prostaglandin-E2-Mediated Transactivation of the Epidermal Growth Factor Receptor and c-Jun NH2-Terminal Kinase Activation. Endocrinology 2006, 147, 4505–4516. [Google Scholar] [CrossRef]
  175. Beales, I.; Francois, F.; Roper, J.; Goodman, A.J.; Pei, Z.; Ghumman, M.; Mourad, M.; de Perez, A.Z.O.; Perez-Perez, G.I.; Tseng, C.-H.; et al. Faculty Opinions recommendation of The association of gastric leptin with oesophageal inflammation and metaplasia. Fac. Opin. Post Publ. Peer Rev. Biomed. Lit. 2008, 57, 16–24. [Google Scholar] [CrossRef]
  176. Mongan, A.M.; Lynam-Lennon, N.; Doyle, S.L.; Casey, R.; Carr, E.; Cannon, A.; Conroy, M.J.; Pidgeon, G.P.; Brennan, L.; Lysaght, J.; et al. Visceral Adipose Tissue Modulates Radiosensitivity in Oesophageal Adenocarcinoma. Int. J. Med. Sci. 2019, 16, 519–528. [Google Scholar] [CrossRef] [Green Version]
  177. Trevellin, E.; Scarpa, M.; Carraro, A.; Lunardi, F.; Kotsafti, A.; Porzionato, A.; Saadeh, L.; Cagol, M.; Alfieri, R.; Tedeschi, U.; et al. Esophageal adenocarcinoma and obesity: Peritumoral adipose tissue plays a role in lymph node invasion. Oncotarget 2015, 6, 11203–11215. [Google Scholar] [CrossRef]
  178. Bain, G.H.; Collie-Duguid, E.; Murray, I.G.; Gilbert, F.J.; Denison, A.; McKiddie, F.; Ahearn, T.; Fleming, I.; Leeds, J.; Phull, P.; et al. Tumour expression of leptin is associated with chemotherapy resistance and therapy-independent prognosis in gastro-oesophageal adenocarcinomas. Br. J. Cancer 2014, 110, 1525–1534. [Google Scholar] [CrossRef] [PubMed]
  179. Beales, I.L.; Garcia-Morales, C.; Ogunwobi, O.O.; Mutungi, G. Adiponectin inhibits leptin-induced oncogenic signalling in oesophageal cancer cells by activation of PTP1B. Mol. Cell. Endocrinol. 2014, 382, 150–158. [Google Scholar] [CrossRef]
  180. Rubenstein, J.H.; Kao, J.Y.; Madanick, R.D.; Zhang, M.; Wang, M.; Spacek, M.B.; Donovan, J.L.; Bright, S.D.; Shaheen, N.J. Association of adiponectin multimers with Barrett’s oesophagus. Gut 2009, 58, 1583–1589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Greer, K.B.; Falk, G.W.; Bednarchik, B.; Li, L.; Chak, A. Associations of Serum Adiponectin and Leptin with Barrett’s Esophagus. Clin. Gastroenterol. Hepatol. 2015, 13, 2265–2272. [Google Scholar] [CrossRef] [Green Version]
  182. Tseng, P.-H.; Yang, W.-S.; Liou, J.-M.; Lee, Y.-C.; Wang, H.-P.; Lin, J.-T.; Wu, M.-S. Associations of Circulating Gut Hormone and Adipocytokine Levels with the Spectrum of Gastroesophageal Reflux Disease. PLoS ONE 2015, 10, e0141410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Kato, M.; Watabe, K.; Hamasaki, T.; Umeda, M.; Furubayashi, A.; Kinoshita, K.; Kishida, O.; Fujimoto, T.; Yamada, A.; Tsukamoto, Y.; et al. Association of low serum adiponectin levels with erosive esophagitis in men: An analysis of 2405 subjects undergoing physical check-ups. J. Gastroenterol. 2011, 46, 1361–1367. [Google Scholar] [CrossRef]
  184. Almers, L.; Graham, J.L.; Havel, P.J.; Corley, D.A. Adiponectin May Modify the Risk of Barrett’s Esophagus in Patients with Gastroesophageal Reflux Disease. Clin. Gastroenterol. Hepatol. 2015, 13, 2256–2264.e1. [Google Scholar] [CrossRef] [Green Version]
  185. Chandar, A.K.; Devanna, S.; Lu, C.; Singh, S.; Greer, K.B.; Chak, A.; Iyer, P.G. Association of Serum Levels of Adipokines and Insulin With Risk of Barrett’s Esophagus: A Systematic Review and Meta-Analysis. Clin. Gastroenterol. Hepatol. 2015, 13, 2241–2255.e4. [Google Scholar] [CrossRef] [Green Version]
  186. Garcia, J.M.; Splenser, A.E.; Kramer, J.R.; Alsarraj, A.; Fitzgerald, S.; Ramsey, D.J.; El-Serag, H.B. Circulating Inflammatory Cytokines and Adipokines Are Associated With Increased Risk of Barrett’s Esophagus: A Case–Control Study. Clin. Gastroenterol. Hepatol. 2014, 12, 229–238.e3. [Google Scholar] [CrossRef] [Green Version]
  187. Duggan, C.; Onstad, L.; Hardikar, S.; Blount, P.L.; Reid, B.J.; Vaughan, T.L. Association between markers of obesity and progression from Barrett’s esophagus to esophageal adenocarcinoma. Clin. Gastroenterol. Hepatol. 2013, 11, 934–943. [Google Scholar] [CrossRef] [Green Version]
  188. Yildirim, A.; Bilici, M.; Cayir, K.; Yanmaz, V.; Yildirim, S.; Tekin, S.B.; Yıldırım, A.; Çayır, K. Serum Adiponectin Levels in Patients with Esophageal Cancer. Jpn. J. Clin. Oncol. 2008, 39, 92–96. [Google Scholar] [CrossRef] [Green Version]
  189. Nakajima, T.E.; Yamada, Y.; Hamano, T.; Furuta, K.; Oda, I.; Kato, H.; Kato, K.; Hamaguchi, T.; Shimada, Y. Adipocytokines and squamous cell carcinoma of the esophagus. J. Cancer Res. Clin. Oncol. 2009, 136, 261–266. [Google Scholar] [CrossRef] [PubMed]
  190. Rehman, K.; Akash, M.S.H.; Alina, Z. Leptin: A new therapeutic target for treatment of diabetes mellitus. J. Cell. Biochem. 2018, 119, 5016–5027. [Google Scholar] [CrossRef]
  191. Delhanty, P.J.; Van Der Eerden, B.C.; Van Leeuwen, J.P.T.M. Ghrelin and bone. BioFactors 2013, 40, 41–48. [Google Scholar] [CrossRef]
  192. Perboni, S.; Inui, A. Appetite and gastrointestinal motility: Role of ghrelin-family peptides. Clin. Nutr. 2010, 29, 227–234. [Google Scholar] [CrossRef]
  193. Inagaki-Ohara, K. Gastric Leptin and Tumorigenesis: Beyond Obesity. Int. J. Mol. Sci. 2019, 20, 2622. [Google Scholar] [CrossRef] [Green Version]
  194. Cammisotto, P.G.; Levy, E.; Bukowiecki, L.J.; Bendayan, M. Cross-talk between adipose and gastric leptins for the control of food intake and energy metabolism. Prog. Histochem. Cytochem. 2010, 45, 143–200. [Google Scholar] [CrossRef]
  195. Weidinger, C.; Ziegler, J.F.; Letizia, M.; Schmidt, F.; Siegmund, B. Adipokines and Their Role in Intestinal Inflammation. Front. Immunol. 2018, 9, 1974. [Google Scholar] [CrossRef] [Green Version]
  196. Grases-Pintó, B.; Torres-Castro, P.; Marín-Morote, L.; Abril-Gil, M.; Castell, M.; Rodriguez-Lagunas, M.J.; Pérez-Cano, F.J.; Franch, À. Leptin and EGF Supplementation Enhance the Immune System Maturation in Preterm Suckling Rats. Nutrients 2019, 11, 2380. [Google Scholar] [CrossRef] [Green Version]
  197. Kaplan, G.G. The global burden of IBD: From 2015 to 2025. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 720–727. [Google Scholar] [CrossRef] [PubMed]
  198. Barnes, E.L.; Loftus, E.V., Jr.; Kappelman, M.D. Effects of Race and Ethnicity on Diagnosis and Management of Inflammatory Bowel Diseases. Gastroenterology 2020. [Google Scholar] [CrossRef]
  199. Nijakowski, K.; Surdacka, A. Salivary Biomarkers for Diagnosis of Inflammatory Bowel Diseases: A Systematic Review. Int. J. Mol. Sci. 2020, 21, 7477. [Google Scholar] [CrossRef] [PubMed]
  200. Kim, S.-E.; Choo, J.; Yoon, J.; Chu, J.R.; Bae, Y.J.; Lee, S.; Park, T.; Sung, M.-K. Genome-wide analysis identifies colonic genes differentially associated with serum leptin and insulin concentrations in C57BL/6J mice fed a high-fat diet. PLoS ONE 2017, 12, e0171664. [Google Scholar] [CrossRef]
  201. Hoffman, J.M.; Sideri, A.; Ruiz, J.J.; Stavrakis, D.; Shih, D.Q.; Turner, J.R.; Pothoulakis, C.; Karagiannides, I. Mesenteric Adipose-derived Stromal Cells From Crohn’s Disease Patients Induce Protective Effects in Colonic Epithelial Cells and Mice With Colitis. Cell. Mol. Gastroenterol. Hepatol. 2018, 6, 1–16. [Google Scholar] [CrossRef] [Green Version]
  202. Kredel, L.; Batra, A.; Siegmund, B. Role of fat and adipokines in intestinal inflammation. Curr. Opin. Gastroenterol. 2014, 30, 559–565. [Google Scholar] [CrossRef] [PubMed]
  203. Al-Hassi, O.H.; Bernardo, D.; Murugananthan, A.U.; Mann, E.R.; English, N.R.; Jones, A.; Kamm, A.M.; Arebi, N.; Hart, A.L.; Blakemore, A.I.F.; et al. A mechanistic role for leptin in human dendritic cell migration: Differences between ileum and colon in health and Crohn’s disease. Mucosal Immunol. 2013, 6, 751–761. [Google Scholar] [CrossRef] [Green Version]
  204. Tian, Y.; Tian, S.; Wang, D.; Cui, F.; Zhang, X.; Zhang, Y. Elevated expression of the leptin receptor ob-R may contribute to inflammation in patients with ulcerative colitis. Mol. Med. Rep. 2019, 20, 4706–4712. [Google Scholar] [CrossRef]
  205. Hoda, M.R.; Scharl, M.; Keely, S.J.; McCole, D.F.; Barrett, K.E. Apical leptin induces chloride secretion by intestinal epithelial cells and in a rat model of acute chemotherapy-induced colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 298, G714–G721. [Google Scholar] [CrossRef]
  206. Le Dréan, G.; Haure-Mirande, V.; Ferrier, L.; Bonnet, C.; Hulin, P.; De Coppet, P.; Segain, J. Visceral adipose tissue and leptin increase colonic epithelial tight junction permeability via a RhoA-ROCK-dependent pathway. FASEB J. 2013, 28, 1059–1070. [Google Scholar] [CrossRef]
  207. Liu, D.-R.; Xu, X.-J.; Yao, S.-K. Increased intestinal mucosal leptin levels in patients with diarrhea-predominant irritable bowel syndrome. World J. Gastroenterol. 2018, 24, 46–57. [Google Scholar] [CrossRef]
  208. Madan, R.; Guo, X.; Naylor, C.; Buonomo, E.L.; Mackay, D.; Noor, Z.; Concannon, P.; Scully, K.W.; Pramoonjago, P.; Kolling, G.L.; et al. Role of Leptin-Mediated Colonic Inflammation in Defense against Clostridium difficile Colitis. Infect. Immun. 2013, 82, 341–349. [Google Scholar] [CrossRef] [Green Version]
  209. Obeid, S.; Wankell, M.; Charrez, B.; Sternberg, J.; Kreuter, R.; Esmaili, S.; Ramezani-Moghadam, M.; Devine, C.; Read, S.; Bhathal, P.; et al. Adiponectin confers protection from acute colitis and restricts a B cell immune response. J. Biol. Chem. 2017, 292, 6569–6582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Arsenescu, V.; Narasimhan, M.L.; Halide, T.; Bressan, R.A.; Barisione, C.; Cohen, N.A.; De Villiers, W.J.S.; Arsenescu, R. Adiponectin and Plant-Derived Mammalian Adiponectin Homolog Exert a Protective Effect in Murine Colitis. Dig. Dis. Sci. 2011, 56, 2818–2832. [Google Scholar] [CrossRef]
  211. Zhao, Q.; Liu, Y.; Tan, L.; Yan, L.; Zuo, X. Adiponectin administration alleviates DSS-induced colonic inflammation in Caco-2 cells and mice. Inflamm. Res. 2018, 67, 663–670. [Google Scholar] [CrossRef]
  212. Matsunaga, H.; Hokari, R.; Kurihara, C.; Okada, Y.; Takebayashi, K.; Okudaira, K.; Watanabe, C.; Komoto, S.; Nakamura, M.; Tsuzuki, Y.; et al. Omega-3 fatty acids exacerbate DSS-induced colitis through decreased adiponectin in colonic subepithelial myofibroblasts. Inflamm. Bowel Dis. 2008, 14, 1348–1357. [Google Scholar] [CrossRef]
  213. Sideri, A.; Stavrakis, D.; Bowe, C.; Shih, D.Q.; Fleshner, P.; Arsenescu, V.; Arsenescu, R.; Turner, J.R.; Pothoulakis, C.; Karagiannides, I. Effects of obesity on severity of colitis and cytokine expression in mouse mesenteric fat. Potential role of adiponectin receptor 1. Am. J. Physiol. Liver Physiol. 2015, 308, G591–G604. [Google Scholar] [CrossRef] [Green Version]
  214. Fayad, R.; Pini, M.; Sennello, J.A.; Cabay, R.J.; Chan, L.; Xu, A.; Fantuzzi, G. Adiponectin Deficiency Protects Mice From Chemically Induced Colonic Inflammation. Gastroenterology 2007, 132, 601–614. [Google Scholar] [CrossRef]
  215. Nishihara, T.; Matsuda, M.; Araki, H.; Oshima, K.; Kihara, S.; Funahashi, T.; Shimomura, I. Effect of Adiponectin on Murine Colitis Induced by Dextran Sulfate Sodium. Gastroenterology 2006, 131, 853–861. [Google Scholar] [CrossRef] [Green Version]
  216. Olivier, I.; Theodorou, V.; Valet, P.; Castan-Laurell, I.; Ferrier, L.; Eutamène, H. Modifications of mesenteric adipose tissue during moderate experimental colitis in mice. Life Sci. 2014, 94, 1–7. [Google Scholar] [CrossRef]
  217. Paeschke, A.; Erben, U.; Kredel, L.I.; Kühl, A.A.; Siegmund, B. Role of visceral fat in colonic inflammation. Curr. Opin. Gastroenterol. 2017, 33, 53–58. [Google Scholar] [CrossRef] [PubMed]
  218. Comstock, S.S.; Lewis, M.M.; Pathak, R.R.; Hortos, K.; Kovan, B.; Fenton, J.I. Cross-Sectional Analysis of Obesity and Serum Analytes in Males Identifies sRAGE as a Novel Biomarker Inversely Associated with Diverticulosis. PLoS ONE 2014, 9, e95232. [Google Scholar] [CrossRef] [Green Version]
  219. Chen, T.-H.; Hsu, C.-M.; Hsu, H.-C.; Chiu, C.-T.; Su, M.-Y.; Chu, Y.-Y.; Chang, M.-L. Plasminogen activator inhibitor-1 is associated with the metabolism and development of advanced colonic polyps. Transl. Res. 2018, 200, 43–53. [Google Scholar] [CrossRef]
  220. Lieberman, D.A.; Rex, D.K.; Winawer, S.J.; Giardiello, F.M.; Johnson, D.A.; Levin, T.R. Guidelines for Colonoscopy Surveillance After Screening and Polypectomy: A Consensus Update by the US Multi-Society Task Force on Colorectal Cancer. Gastroenterology 2012, 143, 844–857. [Google Scholar] [CrossRef] [Green Version]
  221. Comstock, S.S.; Hortos, K.; Kovan, B.; McCaskey, S.; Pathak, R.R.; Fenton, J.I. Adipokines and Obesity Are Associated with Colorectal Polyps in Adult Males: A Cross-Sectional Study. PLoS ONE 2014, 9, e85939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Oh, J.S.; Kim, H.H.; Hwang, H.S.; Yun, D.Y.; Kim, B.S.; Lee, C.H.; Han, J.; Kim, H.G.; Jung, J.T.; Kwon, J.G.; et al. Comparison of blood leptin concentration and colonic mucosa leptin expression in colon adenoma patients and healthy control. Korean J. Gastroenterol. 2014, 63, 354–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Paik, S.S.; Jang, S.-M.; Jang, K.; Lee, K.H.; Choi, D.; Jang, S.J. Leptin Expression Correlates with Favorable Clinicopathologic Phenotype and Better Prognosis in Colorectal Adenocarcinoma. Ann. Surg. Oncol. 2008, 16, 297–303. [Google Scholar] [CrossRef]
  224. Abolhassani, M.; Aloulou, N.; Chaumette, M.T.; Aparicio, T.; Martin-Garcia, N.; Mansour, H.; Le Gouvello, S.; Delchier, J.C.; Sobhani, I.; Rocks, N.; et al. Leptin Receptor-Related Immune Response in Colorectal Tumors: The Role of Colonocytes and Interleukin-8. Cancer Res. 2008, 68, 9423–9432. [Google Scholar] [CrossRef] [Green Version]
  225. Al-Shibli, S.M.; Harun, N.; Ashour, A.E.; Kasmuri, M.H.B.M.; Mizan, S. Expression of leptin and leptin receptors in colorectal cancer—an immunohistochemical study. PeerJ 2019, 7, e7624. [Google Scholar] [CrossRef]
  226. Endo, H.; Hosono, K.; Uchiyama, T.; Sakai, E.; Sugiyama, M.; Takahashi, H.; Nakajima, N.; Wada, K.; Takeda, K.; Nakagama, H.; et al. Leptin acts as a growth factor for colorectal tumours at stages subsequent to tumour initiation in murine colon carcinogenesis. Gut 2011, 60, 1363–1371. [Google Scholar] [CrossRef]
  227. Tutino, V.; Notarnicola, M.; Guerra, V.; Lorusso, D.; Caruso, M.G. Increased soluble leptin receptor levels are associated with advanced tumor stage in colorectal cancer patients. Anticancer Res. 2011, 31, 3381–3383. [Google Scholar]
  228. Aleksandrova, K.; Boeing, H.; Jenab, M.; Bueno-De-Mesquita, H.B.; Jansen, E.; Van Duijnhoven, F.J.B.; Rinaldi, S.; Fedirko, V.; Romieu, I.; Riboli, E.; et al. Leptin and Soluble Leptin Receptor in Risk of Colorectal Cancer in the European Prospective Investigation into Cancer and Nutrition Cohort. Cancer Res. 2012, 72, 5328–5337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Uddin, S.; Bavi, P.P.; Hussain, A.R.; Alsbeih, G.; Alsanea, N.; Abduljabbar, A.; Ashari, L.H.; Alhomoud, S.; Al-Dayel, F.; Ahmed, M.; et al. Leptin receptor expression in Middle Eastern colorectal cancer and its potential clinical implication. Carcinogenesis 2009, 30, 1832–1840. [Google Scholar] [CrossRef] [Green Version]
  230. Aloulou, N.; Bastuji-Garin, S.; Le Gouvello, S.; Abolhassani, M.; Chaumette, M.T.; Charachon, A.; Leroy, K.; Sobhani, I. Involvement of the Leptin Receptor in the Immune Response in Intestinal Cancer. Cancer Res. 2008, 68, 9413–9422. [Google Scholar] [CrossRef] [Green Version]
  231. Penrose, H.M.; Heller, S.; Cable, C.; Nakhoul, H.; Baddoo, M.; Flemington, E.; Crawford, S.E.; Savkovic, S.D. High-fat diet induced leptin and Wnt expression: RNA-sequencing and pathway analysis of mouse colonic tissue and tumors. Carcinogenesis 2017, 38, 302–311. [Google Scholar] [CrossRef] [Green Version]
  232. Wang, D.; Chen, J.; Chen, H.; Duan, Z.; Xu, Q.; Wei, M.; Wang, L.; Zhong, M. Leptin regulates proliferation and apoptosis of colorectal carcinoma through PI3K/Akt/mTOR signalling pathway. J. Biosci. 2011, 37, 91–101. [Google Scholar] [CrossRef]
  233. Fazolini, N.P.B.; Cruz, A.L.S.; Werneck, M.B.F.; Viola, J.P.B.; Maya-Monteiro, C.M.; Bozza, P.T. Leptin activation of mTOR pathway in intestinal epithelial cell triggers lipid droplet formation, cytokine production and increased cell proliferation. Cell Cycle 2015, 14, 2667–2676. [Google Scholar] [CrossRef]
  234. Dong, Z.; Lee, Y.-H.; Na, H.-K.; Baek, J.-H.; Dong, Z. Leptin induces SIRT1 expression through activation of NF-E2-related factor 2: Implications for obesity-associated colon carcinogenesis. Biochem. Pharmacol. 2018, 153, 282–291. [Google Scholar] [CrossRef]
  235. Bartucci, M.; Svensson, S.; Ricci-Vitiani, L.; Dattilo, R.; Biffoni, M.; Signore, M.; Ferla, R.; De Maria, R.; Surmacz, E. Obesity hormone leptin induces growth and interferes with the cytotoxic effects of 5-fluorouracil in colorectal tumor stem cells. Endocr. Relat. Cancer 2010, 17, 823–833. [Google Scholar] [CrossRef] [Green Version]
  236. Lin, M.-C.; Wang, F.-Y.; Ko, H.-H.; Tang, F.-Y. Cancer Chemopreventive Effects of Lycopene: Suppression of MMP-7 Expression and Cell Invasion in Human Colon Cancer Cells. J. Agric. Food Chem. 2011, 59, 11304–11318. [Google Scholar] [CrossRef] [PubMed]
  237. Jaffe, T.; Schwartz, B. Leptin promotes motility and invasiveness in human colon cancer cells by activating multiple signal-transduction pathways. Int. J. Cancer 2008, 123, 2543–2556. [Google Scholar] [CrossRef]
  238. Hoda, M.R.; Keely, S.J.; Bertelsen, L.S.; Junger, W.G.; Dharmasena, D.; Barrett, K.E. Leptin acts as a mitogenic and antiapoptotic factor for colonic cancer cells. BJS 2007, 94, 346–354. [Google Scholar] [CrossRef] [PubMed]
  239. Ogunwobi, O.O.; Beales, I.L. The anti-apoptotic and growth stimulatory actions of leptin in human colon cancer cells involves activation of JNK mitogen activated protein kinase, JAK2 and PI3 kinase/Akt. Int. J. Color. Dis. 2006, 22, 401–409. [Google Scholar] [CrossRef]
  240. Fenton, J.I.; Hursting, S.D.; Perkins, S.N.; Hord, N.G. Interleukin-6 production induced by leptin treatment promotes cell proliferation in an Apc (Min/+) colon epithelial cell line. Carcinogenesis 2006, 27, 1507–1515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Ogunwobi, O.; Beales, I.L.P. Cyclo-oxygenase-Independent Inhibition of Apoptosis and Stimulation of Proliferation by Leptin in Human Colon Cancer Cells. Dig. Dis. Sci. 2007, 52, 1934–1945. [Google Scholar] [CrossRef]
  242. Padidar, S.; Farquharson, A.J.; Williams, L.M.; Kelaiditi, E.; Hoggard, N.; Arthur, J.R.; Drew, J.E. Leptin up-regulates pro-inflammatory cytokines in discrete cells within mouse colon. J. Cell. Physiol. 2011, 226, 2123–2130. [Google Scholar] [CrossRef] [PubMed]
  243. Birmingham, J.M.; Busik, J.V.; Hansen-Smith, F.M.; Fenton, J.I. Novel mechanism for obesity-induced colon cancer progression. Carcinogenesis 2009, 30, 690–697. [Google Scholar] [CrossRef] [Green Version]
  244. Plaisancie, P.; Ducroc, R.; El Homsi, M.; Tsocas, A.; Guilmeau, S.; Zoghbi, S.; Thibaudeau, O.; Bado, A. Luminal leptin activates mucin-secreting goblet cells in the large bowel. Am. J. Physiol. Liver Physiol. 2006, 290, G805–G812. [Google Scholar] [CrossRef] [Green Version]
  245. Saxena, A.; Fayad, R.; Kaur, K.; Truman, S.; Greer, J.; Carson, J.A.; Chanda, A. Dietary selenium protects adiponectin knockout mice against chronic inflammation induced colon cancer. Cancer Biol. Ther. 2017, 18, 257–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Saxena, A.; Baliga, M.S.; Ponemone, V.; Kaur, K.; Larsen, B.; Fletcher, E.; Greene, J.; Fayad, R. Mucus and adiponectin deficiency: Role in chronic inflammation-induced colon cancer. Int. J. Color. Dis. 2013, 28, 1267–1279. [Google Scholar] [CrossRef] [PubMed]
  247. Deng, L.; Zhao, X.; Chen, M.; Ji, H.; Zhang, Q.; Chen, R.; Wang, Y. Plasma adiponectin, visfatin, leptin, and resistin levels and the onset of colonic polyps in patients with prediabetes. BMC Endocr. Disord. 2020, 20, 1–12. [Google Scholar] [CrossRef]
  248. Saetang, J.; Boonpipattanapong, T.; Palanusont, A.; Maneechay, W.; Sangkhathat, S. Alteration of Leptin and Adiponectin in Multistep Colorectal Tumorigenesis. Asian Pac. J. Cancer Prev. 2016, 17, 2119–2123. [Google Scholar] [CrossRef] [Green Version]
  249. Gonullu, G.; Kahraman, H.; Bedir, A.; Bektas, A.; Yücel, I. Association between adiponectin, resistin, insulin resistance, and colorectal tumors. Int. J. Color. Dis. 2009, 25, 205–212. [Google Scholar] [CrossRef]
  250. Nigro, E.; Schettino, P.; Polito, R.; Scudiero, O.; Monaco, M.L.; De Palma, G.D.; Daniele, A. Adiponectin and colon cancer: Evidence for inhibitory effects on viability and migration of human colorectal cell lines. Mol. Cell. Biochem. 2018, 448, 125–135. [Google Scholar] [CrossRef]
  251. Salinas, M.L.; Fuentes, N.R.; Choate, R.; Wright, R.C.; McMurray, D.N.; Chapkin, R.S. AdipoRon Attenuates Wnt Signaling by Reducing Cholesterol-Dependent Plasma Membrane Rigidity. Biophys. J. 2020, 118, 885–897. [Google Scholar] [CrossRef]
  252. Aleksandrova, K.; Boeing, H.; Jenab, M.; Bueno-De-Mesquita, H.B.; Jansen, E.; Van Duijnhoven, F.J.; Fedirko, V.; Rinaldi, S.; Romieu, I.; Riboli, E.; et al. Total and high-molecular weight adiponectin and risk of colorectal cancer: The European Prospective Investigation into Cancer and Nutrition Study. Carcinogenesis 2012, 33, 1211–1218. [Google Scholar] [CrossRef]
  253. Williams, C.J.; Mitsiades, N.; Sozopoulos, E.; His, A.; Wolk, A.; Nifli, A.P.; Tseleni-Balafouta, S.; Mantzoros, C.S. Adiponectin receptor expression is elevated in colorectal carcinomas but not in gastrointestinal stromal tumors. Endocr. Relat. Cancer 2008, 15, 289–299. [Google Scholar] [CrossRef] [Green Version]
  254. Vetvik, K.K.; Sonerud, T.; Lindeberg, M.; Lüders, T.; Størkson, R.H.; Jonsdottir, K.; Frengen, E.; Pietiläinen, K.H.; Bukholm, I. Globular adiponectin and its downstream target genes are up-regulated locally in human colorectal tumors: Ex vivo and in vitro studies. Metabolism 2014, 63, 672–681. [Google Scholar] [CrossRef]
  255. Polito, R.; Nigro, E.; Fei, L.; De Magistris, L.; Monaco, M.L.; D’Amico, R.; Naviglio, S.; Signoriello, G.; Daniele, A. Adiponectin Is Inversely Associated With Tumour Grade in Colorectal Cancer Patients. Anticancer Res. 2020, 40, 3751–3757. [Google Scholar] [CrossRef]
  256. Inamura, K.; Song, M.; Jung, S.; Nishihara, R.; Yamauchi, M.; Lochhead, P.; Qian, Z.R.; Kim, S.A.; Mima, K.; Sukawa, Y.; et al. Prediagnosis Plasma Adiponectin in Relation to Colorectal Cancer Risk According toKRASMutation Status. J. Natl. Cancer Inst. 2015, 108, djv363. [Google Scholar] [CrossRef] [Green Version]
  257. Kim, A.Y.; Lee, Y.S.; Kim, K.H. Adiponectin represses colon cancer cell proliferation via AdipoR1- and -R2-mediated AMPK activation. Mol. Endocrinol. 2010, 24, 1441–1452. [Google Scholar] [CrossRef]
  258. Mutoh, M.; Teraoka, N.; Takasu, S.; Takahashi, M.; Onuma, K.; Yamamoto, M.; Kubota, N.; Iseki, T.; Kadowaki, T.; Sugimura, T.; et al. Loss of Adiponectin Promotes Intestinal Carcinogenesis in Min and Wild-type Mice. Gastroenterology 2011, 140, 2000–2008.e2. [Google Scholar] [CrossRef]
  259. Sugiyama, M.; Takahashi, H.; Hosono, K.; Endo, H.; Kato, S.; Yoneda, K.; Nozaki, Y.; Fujita, K.; Yoneda, M.; Wada, K.; et al. Adiponectin inhibits colorectal cancer cell growth through the AMPK/mTOR pathway. Int. J. Oncol. 2009, 34, 339–344. [Google Scholar]
  260. Moon, H.-S.; Mantzoros, C.S. Adiponectin and metformin additively attenuate IL1β-induced malignant potential of colon cancer. Endocr. Relat. Cancer 2013, 20, 849–859. [Google Scholar] [CrossRef] [Green Version]
  261. Moon, H.-S.; Liu, X.; Nagel, J.M.; Chamberland, J.P.; Diakopoulos, K.N.; Brinkoetter, M.T.; Hatziapostolou, M.; Wu, Y.; Robson, S.C.; Iliopoulos, D.; et al. Salutary effects of adiponectin on colon cancer: In vivo and in vitro studies in mice. Gut 2012, 62, 561–570. [Google Scholar] [CrossRef]
  262. Erarslan, E.; Turkay, C.; Koktener, A.; Koca, C.; Uz, B.; Bavbek, N. Association of Visceral Fat Accumulation and Adiponectin Levels with Colorectal Neoplasia. Dig. Dis. Sci. 2008, 54, 862–868. [Google Scholar] [CrossRef]
  263. Declercq, V.; McMurray, D.; Chapkin, R.S. Obesity promotes colonic stem cell expansion during cancer initiation. Cancer Lett. 2015, 369, 336–343. [Google Scholar] [CrossRef] [Green Version]
  264. Fujisawa, T.; Endo, H.; Tomimoto, A.; Sugiyama, M.; Takahashi, H.; Saito, S.; Inamori, M.; Nakajima, N.; Watanabe, M.; Kubota, N.; et al. Adiponectin suppresses colorectal carcinogenesis under the high-fat diet condition. Gut 2008, 57, 1531–1538. [Google Scholar] [CrossRef] [Green Version]
  265. Takahashi, H.; Hosono, K.; Endo, H.; Nakajima, A. Colon epithelial proliferation and carcinogenesis in diet-induced obesity. J. Gastroenterol. Hepatol. 2013, 28, 41–47. [Google Scholar] [CrossRef] [PubMed]
  266. Ealey, K.N.; Archer, M.C. Elevated circulating adiponectin and elevated insulin sensitivity in adiponectin transgenic mice are not associated with reduced susceptibility to colon carcinogenesis. Int. J. Cancer 2009, 124, 2226–2230. [Google Scholar] [CrossRef]
  267. He, B.; Pan, Y.; Zhang, Y.; Bao, Q.; Chen, L.; Nie, Z.-L.; Gu, L.; Xu, Y.; Wang, S. Effects of genetic variations in the Adiponectin pathway genes on the risk of colorectal cancer in the Chinese population. BMC Med. Genet. 2011, 12, 94. [Google Scholar] [CrossRef] [Green Version]
  268. Otani, K.; Ishihara, S.; Yamaguchi, H.; Murono, K.; Yasuda, K.; Nishikawa, T.; Tanaka, T.; Kiyomatsu, T.; Hata, K.; Kawai, K.; et al. Adiponectin and colorectal cancer. Surg. Today 2016, 47, 151–158. [Google Scholar] [CrossRef]
  269. Yunusova, N.V.; Kondakova, I.V.; Kolomiets, L.A.; Afanasiev, S.G.; Chernyshova, A.L.; Shatokhina, O.V.; Frolova, A.E.; Zhou, Z.; Wang, W. Serum adipokines and their receptors in endometrial and colon cancer patients: Relationship with tumor invasion and metastasis. Vopr. Onkol. 2015, 61, 619–623. [Google Scholar]
  270. Suman, S.; Kallakury, B.V.S.; Fornace, A.J.; Datta, K. Protracted Upregulation of Leptin and IGF1 is Associated with Activation of PI3K/Akt and JAK2 Pathway in Mouse Intestine after Ionizing Radiation Exposure. Int. J. Biol. Sci. 2015, 11, 274–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  271. Song, M.; Zhang, X.; Wu, K.; Ogino, S.; Fuchs, C.S.; Giovannucci, E.L.; Chan, A.T. Plasma Adiponectin and Soluble Leptin Receptor and Risk of Colorectal Cancer: A Prospective Study. Cancer Prev. Res. 2013, 6, 875–885. [Google Scholar] [CrossRef] [Green Version]
  272. Singh, S.; Chouhan, S.; Mohammad, N.; Bhat, M.K. Resistin causes G1 arrest in colon cancer cells through upregulation of SOCS3. FEBS Lett. 2017, 591, 1371–1382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Huang, W.-S.; Yang, J.-T.; Lu, C.-C.; Chang, S.-F.; Chen, C.-N.; Su, Y.-P.; Lee, K.-C. Fulvic Acid Attenuates Resistin-Induced Adhesion of HCT-116 Colorectal Cancer Cells to Endothelial Cells. Int. J. Mol. Sci. 2015, 16, 29370–29382. [Google Scholar] [CrossRef] [Green Version]
  274. Al-Harithy, R.N. Polymorphisms in RETN gene and susceptibility to colon cancer in Saudi patients. Ann. Saudi Med. 2014, 34, 334–339. [Google Scholar] [CrossRef] [PubMed]
  275. Johansen, J.S.; Christensen, I.J.; Jørgensen, L.N.; Olsen, J.; Rahr, H.B.; Nielsen, K.T.; Laurberg, S.; Brünner, N.; Nielsen, H.J. Serum YKL-40 in Risk Assessment for Colorectal Cancer: A Prospective Study of 4496 Subjects at Risk of Colorectal Cancer. Cancer Epidemiol. Biomark. Prev. 2015, 24, 621–626. [Google Scholar] [CrossRef] [Green Version]
  276. Kzhyshkowska, J.; Yin, S.; Liu, T.; Riabov, V.; Mitrofanova, I. Role of chitinase-like proteins in cancer. Biol. Chem. 2016, 397, 231–247. [Google Scholar] [CrossRef]
  277. Abola, M.V.; Thompson, C.L.; Chen, Z.; Chak, A.; Berger, N.A.; Kirwan, J.P.; Li, L. Serum levels of retinol-binding protein 4 and risk of colon adenoma. Endocr. Relat. Cancer 2015, 22, L1–L4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  278. Eichelmann, F.; Schulze, M.B.; Wittenbecher, C.; Menzel, J.; Weikert, C.; Di Giuseppe, R.; Biemann, R.; Isermann, B.; Fritsche, A.; Boeing, H.; et al. Association of Chemerin Plasma Concentration With Risk of Colorectal Cancer. JAMA Netw. Open 2019, 2, e190896. [Google Scholar] [CrossRef]
  279. Erdogan, S.; Yilmaz, F.M.; Yazici, O.; Yozgat, A.; Sezer, S.; Ozdemir, N.; Uysal, S.; Purnak, T.; Sendur, M.A.; Ozaslan, E. Inflammation and chemerin in colorectal cancer. Tumor Biol. 2015, 37, 6337–6342. [Google Scholar] [CrossRef] [PubMed]
  280. Ding, X.; Saxena, N.K.; Lin, S.; Xu, A.; Srinivasan, S.; Anania, F.A. The Roles of Leptin and Adiponectin. Am. J. Pathol. 2005, 166, 1655–1669. [Google Scholar] [CrossRef]
  281. Lin, S.Y.; Chen, W.Y.; Chiu, Y.T.; Lee, W.J.; Wu, H.S.; Sheua, W.H.-H. Different tumor necrosis factor-α–associated leptin expression in rats with dimethylnitrosamine and bile duct ligation–induced liver cirrhosis. Metabolism 2005, 54, 445–452. [Google Scholar] [CrossRef] [PubMed]
  282. Fava, G.; Alpini, G.; Rychlicki, C.; Saccomanno, S.; DeMorrow, S.; Trozzi, L.; Candelaresi, C.; Venter, J.; Di Sario, A.; Marzioni, M.; et al. Leptin enhances cholangiocarcinoma cell growth. Cancer Res. 2008, 68, 6752–6761. [Google Scholar] [CrossRef] [Green Version]
  283. Peng, C.; Sun, Z.; Li, O. Leptin stimulates the epithelial-mesenchymal transition and pro-angiogenic capability of cholangiocarcinoma cells through the miR-122/PKM2 axis. Int. J. Oncol. 2019, 55, 298–308. [Google Scholar] [CrossRef]
  284. Moradi, M.; Doustimotlagh, A.H.; Dehpour, A.R.; Rahimi, N.; Golestani, A. The influence of TRAIL, adiponectin and sclerostin alterations on bone loss in BDL-induced cirrhotic rats and the effect of opioid system blockade. Life Sci. 2019, 233, 116706. [Google Scholar] [CrossRef]
  285. Xia, Y.; Gong, J.-P. Impact of Recombinant Globular Adiponectin on Early Warm Ischemia-Reperfusion Injury in Rat Bile Duct after Liver Transplantation. Sci. Rep. 2014, 4, 6426. [Google Scholar] [CrossRef] [Green Version]
  286. Lin, S.-Y.; Sheu, W.H.-H.; Chen, W.-Y.; Lee, F.-Y.; Huang, C.-J. Stimulated resistin expression in white adipose of rats with bile duct ligation-induced liver cirrhosis: Relationship to cirrhotic hyperinsulinemia and increased tumor necrosis factor-alpha. Mol. Cell. Endocrinol. 2005, 232, 1–8. [Google Scholar] [CrossRef] [PubMed]
  287. Nakeeb, A.; Comuzzie, A.G.; Al-Azzawi, H.; Sonnenberg, E.G.; Kissebah, A.H.; Pitt, A.H. Insulin Resistance Causes Human Gallbladder Dysmotility. J. Gastrointest. Surg. 2006, 10, 940–949. [Google Scholar] [CrossRef]
  288. Avgerinos, K.I.; Spyrou, N.; Mantzoros, C.S.; Dalamaga, M. Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metabolism 2019, 92, 121–135. [Google Scholar] [CrossRef]
  289. Ko, C.W.; Beresford, S.A.A.; Schulte, S.J.; Matsumoto, A.M.; Lee, S.P. Incidence, natural history, and risk factors for biliary sludge and stones during pregnancy. Hepatology 2005, 41, 359–365. [Google Scholar] [CrossRef]
  290. Ruhl, C.E.; Everhart, J.E. Relationship of serum leptin concentration and other measures of adiposity with gallbladder disease. Hepatology 2001, 34, 877–883. [Google Scholar] [CrossRef]
  291. Hyogo, H. Restoration of gallstone susceptibility by leptin in C57BL/6Job/obmice. J. Lipid Res. 2003, 44, 1232–1240. [Google Scholar] [CrossRef] [Green Version]
  292. Lee, S.; Lee, A.; Kweon, O.-K.; Kim, W.H. Presence and distribution of leptin and leptin receptor in the canine gallbladder. Acta Histochem. 2016, 118, 674–678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  293. Lee, S.; Kweon, O.-K.; Kim, W.H. Associations between serum leptin levels, hyperlipidemia, and cholelithiasis in dogs. PLoS ONE 2017, 12, e0187315. [Google Scholar] [CrossRef] [Green Version]
  294. Swartz-Basile, D.A.; Lu, D.; Basile, D.P.; Graewin, S.J.; Al-Azzawi, H.; Kiely, J.M.; Mathur, A.; Yancey, K.; Pitt, H.A. Leptin regulates gallbladder genes related to absorption and secretion. Am. J. Physiol. Liver Physiol. 2007, 293, G84–G90. [Google Scholar] [CrossRef] [Green Version]
  295. Graewin, S.J.; Kiely, J.M.; Lu, D.; Svatek, C.L.; Al-Azzawi, H.H.; Swartz-Basile, D.A.; Pitt, H.A. Leptin Regulates Gallbladder Genes Related to Gallstone Pathogenesis in Leptin-Deficient Mice. J. Am. Coll. Surg. 2008, 206, 503–510. [Google Scholar] [CrossRef]
  296. Goldblatt, M.; Goldblatt, I.M.; Swartz-Basile, A.D.; Svatek, C.L.; Nakeeb, A.; Pitt, H.A. Decreased Gallbladder Response in Leptin-Deficient Obese Mice. J. Gastrointest. Surg. 2002, 6, 438–444. [Google Scholar] [CrossRef]
  297. Zou, H.; Liu, Y.; Wei, D.; Wang, T.; Wang, K.; Huang, S.; Liu, L.; Li, Y.; Ge, J.; Li, X.; et al. Leptin promotes proliferation and metastasis of human gallbladder cancer through OB-Rb leptin receptor. Int. J. Oncol. 2016, 49, 197–206. [Google Scholar] [CrossRef]
  298. Tran, K.Q.; Swartz-Basile, A.D.; Nakeeb, A.; Pitt, H.A. Gallbladder motility in agouti-yellow and leptin-resistant obese mice. J. Surg. Res. 2003, 113, 56–61. [Google Scholar] [CrossRef]
  299. Tran, K.Q.; Graewin, S.J.; Swartz-Basile, D.A.; Nakeeb, A.; Svatek, C.L.; Pitt, H.A. Leptin-resistant obese mice have paradoxically low biliary cholesterol saturation. Surgery 2003, 134, 372–377. [Google Scholar] [CrossRef]
  300. Coe, P.O.; O’Reilly, D.A.; Renehan, A.G. Excess adiposity and gastrointestinal cancer. BJS 2014, 101, 1518–1531. [Google Scholar] [CrossRef]
  301. Ogiyama, H.; Kamada, Y.; Kiso, S.; Araki, H.; Yamada, T.; Nishihara, T.; Watabe, K.; Tochino, Y.; Kihara, S.; Funahashi, T.; et al. Lack of adiponectin promotes formation of cholesterol gallstones in mice. Biochem. Biophys. Res. Commun. 2010, 399, 352–358. [Google Scholar] [CrossRef]
Figure 1. A schematic describing obesity-related digestive diseases. The obesity-related diseases in the digestive tract from the esophagus, stomach, liver, biliary tree, gallbladder to the colon are labelled. Ca: cancer. GERD: gastroesophageal reflux disease; NAFLD: non-alcoholic fatty liver disease.
Figure 1. A schematic describing obesity-related digestive diseases. The obesity-related diseases in the digestive tract from the esophagus, stomach, liver, biliary tree, gallbladder to the colon are labelled. Ca: cancer. GERD: gastroesophageal reflux disease; NAFLD: non-alcoholic fatty liver disease.
Ijms 21 08308 g001
Figure 2. The adipokine-associated signaling pathways in colitis and colon cancer. The altered adipokine and associated pathways are shown on the left for colitis and on the right for colon cancer. Up arrows: upregulation of the signaling pathways under the stimulation of associated adipokines; down arrows: downregulation of the signaling pathways under the stimulation of associated adipokines. MAT: mesenteric adipose tissue; ADSC: adipose tissue-derived stem cells; ObR: leptin receptor; STAT3: signal transducer and activator of transcription 3; NF-êB: nuclear factor ê-light-chain-enhancer of activated B cells; RhoA: Ras homolog gene family, member A; IL-1â: interleukin 1â; TNF-á: tumor necrosis factor-á; PI3K: phosphatidylinositol 3-kinase; mTOR: mammalian target of rapamycin; Nrf2: nuclear factor erythroid 2-related factor 2; SIRT1:silent information regulator 2 homologue 1; ERK1/2: extracellular signal-related kinase 1/2; MAPK: mitogen-activated protein kinase; JAK2: Janus kinase 2; AP-1: activator protein 1; IL-6: interleukin 6; CXCL1: chemokine (C-X-C motif) ligand 1; VEGF: vascular endothelial growth factor; PKC: protein kinase C;Lgr5+: leucine-rich repeat-containing G-protein coupled receptor 5+; AMPK: MP-activated protein kinase; LKB: liver kinase B1; MYD88: myeloid differentiation primary response 88.
Figure 2. The adipokine-associated signaling pathways in colitis and colon cancer. The altered adipokine and associated pathways are shown on the left for colitis and on the right for colon cancer. Up arrows: upregulation of the signaling pathways under the stimulation of associated adipokines; down arrows: downregulation of the signaling pathways under the stimulation of associated adipokines. MAT: mesenteric adipose tissue; ADSC: adipose tissue-derived stem cells; ObR: leptin receptor; STAT3: signal transducer and activator of transcription 3; NF-êB: nuclear factor ê-light-chain-enhancer of activated B cells; RhoA: Ras homolog gene family, member A; IL-1â: interleukin 1â; TNF-á: tumor necrosis factor-á; PI3K: phosphatidylinositol 3-kinase; mTOR: mammalian target of rapamycin; Nrf2: nuclear factor erythroid 2-related factor 2; SIRT1:silent information regulator 2 homologue 1; ERK1/2: extracellular signal-related kinase 1/2; MAPK: mitogen-activated protein kinase; JAK2: Janus kinase 2; AP-1: activator protein 1; IL-6: interleukin 6; CXCL1: chemokine (C-X-C motif) ligand 1; VEGF: vascular endothelial growth factor; PKC: protein kinase C;Lgr5+: leucine-rich repeat-containing G-protein coupled receptor 5+; AMPK: MP-activated protein kinase; LKB: liver kinase B1; MYD88: myeloid differentiation primary response 88.
Ijms 21 08308 g002
Table 1. Adipokine alterations in various digestive diseases *.
Table 1. Adipokine alterations in various digestive diseases *.
DiseasesAdipokinesIncreased (I), Decreased (D), or No Changes (N)Associated Findings (References)
NAFLDLeptinIIncreased severity [22]
AdiponectinDInversely related to the severity of steatosis [29], necroinflammation, and fibrosis [28]
PAI-1IIndependently associated with NAFLD [39]
Hepatitis BLeptinI/DAssociated with fibrosis/cirrhosis [44,45,46]/with cirrhosis/HCC [47]
AdiponectinI/DAssociated with viral load [48,49]/Viral load inversely associated with HDL-C [50]
ResistinIAssociated with hepatic necroinflammation [54]
VisfatinINegatively correlated with haptoglobin and fibrinogen [56]
Hepatitis CLeptinI/N[65,66]/[67,68]
AdiponectinI/N/DAssociated with fibrosis [74,75,76,77,78,81] and inflammation [85]/[67]/in G1 and G3 HCV patients [84], associated with steatosis [79,84,86,87,88,89]
VisfatinI[104,105]
RBP4DInversely associated with hepatic fibrosis [110,111,112]
ResistinIAssociated with hepatic fibrosis [115,116], reversed after viral clearance [55,120,121], associated with hepatic fibrosis [117]
ChemerinI[122]
PBCLeptinI/D[126,127]/[128,129,130]
AdiponectinI[127]
ResistinI[127]
ALDLeptinI, N or D[136]
AdiponectinI or D[136]
ChemerinI[141]
Pancreatic cancerLeptinI[145]
AdiponectinD[145]
DiabetesLeptinI[141]
GERDLeptinI[167,168,173]
Barrett’s esophagusLeptinI[171,185,186], stronger in men [166,172]
AdiponectinI/D/N[184]/Among patients with GERD and among smokers [181], especially in patients with GERD [180,182]/[186]
Esophageal cancerLeptinIIncreased cellular response to radiation [176], angiogenesis and lymphangiogenesis [177], chemoresistance of gastro-oesophageal adenocarcinomas [178].
ColitisLeptinI[203]
DiverticulosisLeptinI[218]
AdiponectinD[218]
Colon polypLeptinISerum leptin associated with tubular adenoma [221], local leptin with colonic adenoma [222]
AdiponectinD[247]
RBP4I[277]
Colon cancerLeptinN[227]
AdiponectinD[248,249]
ResistinI[262,263]
YKL-40IIn subjects without comorbidity [275] and correlated with poor prognosis in patients with colon cancers [276]
CholelithiasisLeptinI[289]
NAFLD: Non-alcoholic fatty liver disease; HCC: Hepatocellular carcinoma; HDL-C: high-density lipoprotein cholesterol; G1 and G3: genotype 1 and genotype 3; HCV: hepatitis C virus; PBC: primary biliary cholangitis; ALD: alcoholic liver disease; GERD: gastroesophageal reflux disease. *: data of in vivo or animal studies were not listed in the current table.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chang, M.-L.; Yang, Z.; Yang, S.-S. Roles of Adipokines in Digestive Diseases: Markers of Inflammation, Metabolic Alteration and Disease Progression. Int. J. Mol. Sci. 2020, 21, 8308. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21218308

AMA Style

Chang M-L, Yang Z, Yang S-S. Roles of Adipokines in Digestive Diseases: Markers of Inflammation, Metabolic Alteration and Disease Progression. International Journal of Molecular Sciences. 2020; 21(21):8308. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21218308

Chicago/Turabian Style

Chang, Ming-Ling, Zinger Yang, and Sien-Sing Yang. 2020. "Roles of Adipokines in Digestive Diseases: Markers of Inflammation, Metabolic Alteration and Disease Progression" International Journal of Molecular Sciences 21, no. 21: 8308. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21218308

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop