Hostname: page-component-745bb68f8f-mzp66 Total loading time: 0 Render date: 2025-02-06T10:46:53.318Z Has data issue: false hasContentIssue false

The potential role of FNDC5/irisin in various liver diseases: awakening the sleeping beauties

Published online by Cambridge University Press:  13 June 2022

Xiaoyu Wang
Affiliation:
Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
Lihong Mao
Affiliation:
Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
Chaoqun Li
Affiliation:
Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China Department of Internal Medicine, Tianjin Hexi Hospital, Qiongzhou Road 43, Hexi District, Tianjin 300202, China
Yangyang Hui
Affiliation:
Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
Zihan Yu
Affiliation:
Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
Mingyu Sun
Affiliation:
Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
Yifan Li
Affiliation:
Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
Gaoyue Guo
Affiliation:
Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
Wanting Yang
Affiliation:
Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
Binxin Cui
Affiliation:
Department of Gastroenterology, Tianjin Medical University General Hospital Airport Hospital, East Street 6, Tianjin Airport Economic Area, Tianjin 300308, China
Xiaofei Fan
Affiliation:
Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
Chao Sun*
Affiliation:
Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China Department of Gastroenterology, Tianjin Medical University General Hospital Airport Hospital, East Street 6, Tianjin Airport Economic Area, Tianjin 300308, China
*
Author for correspondence: Chao Sun, E-mail: chaosun@tmu.edu.cn
Rights & Permissions [Opens in a new window]

Abstract

Fibronectin type III domain-containing protein 5 (FNDC5) is a transmembrane protein and the precursor of irisin, which serves as a systemic exerkine/myokine with multiple origins. Since its discovery in 2012, this hormone-like polypeptide has rapidly evolved to a component significantly involved in a gamut of metabolic dysregulations and various liver diseases. After a decade of extensive investigation on FNDC5/irisin, we are still surrounded by lots of open questions regarding its diagnostic and therapeutic values. In this review, we first concentrated on the structure–function relationship of FNDC5/irisin. Next, we comprehensively summarised the current knowledge and research findings regarding pathogenic roles/therapeutic applications of FNDC5/irisin in the context of non-alcoholic fatty liver disease, fibrosis, liver injury due to multiple detrimental insults, hepatic malignancy and intrahepatic cholestasis of pregnancy. Moreover, the prominent molecules involved in the underlying mechanisms and signalling pathways were highlighted. As a result, emerging evidence reveals FNDC5/irisin may act as a proxy for diagnosing liver disease pathology, a sensitive biomarker for assessing damage severity, a predisposing factor for surveilling illness progression and a treatment option with protective/preventive impact, all of which are highly dependent on disease grading and contextually pathological features.

Type
Review
Copyright
Copyright © The Author(s), 2022. Published by Cambridge University Press

Introduction

Fibronectin type III domain-containing protein 5 (FNDC5) was originally identified and characterised by two independent research teams in 2002, which is mainly expressed in the skeletal muscle along with the heart, adipose tissue, brain and liver but with relatively small amount (Ref. Reference Aydin1). The synthesis and secretion of FNDC5 is stimulated by peroxisome proliferator-activated receptor-γ co-activator 1α (PGC1α), which serves as a transcription cofactor in response to endurance exercise in muscle tissue (Ref. Reference Bostrom2). As a glycosylated type I transmembrane protein, FNDC5 is proteolytically cleaved and released into the circulation constituting a soluble polypeptide, namely irisin of 112 amino acids (aa). It is widely evidenced that FNDC5/irisin can potentiate energy expenditure by browning white adipose tissue and be responsible for glucose homeostasis by ameliorating insulin resistance, yet controversies also exist (Ref. Reference Perakakis3). In addition to its myokine activity, FNDC5/irisin can also serve as a type of adipokine. Reports have indicated that irisin suppresses lipids accumulation via modifying adipose triglyceride lipase and fatty acid synthase (Refs Reference Huh4, Reference Ma5). Furthermore, overexpressed FNDC5/irisin in obese mice led to reduced adipocyte size in subcutaneous adipose tissue and induced lipolysis (Ref. Reference Xiong6). All these results revealed that the main facet of FNDC5/irisin is governing fat size of the body and in consequence the metabolic rate of fat tissue. Taking into account the exercise-induced property of irisin (myokine) in the physiological context, it has extensively raised concerns of the scientific community to investigate its pathobiological role, clinical relevance and therapeutic potential in a wide spectrum of diseases. Since the inspiring discovery of irisin in 2012, approximately 1400 researches have been published with stable annual increment.

Liver disease accounts for a heavy burden on global healthcare and the economic system in line with increased prevalence and related morbidity/mortality (Ref. Reference Cui7). Accordingly, around 2 million yearly deaths have been attributed to predominant liver diseases including but not limited to viral hepatitis infection, liver cirrhosis and various hepatic malignancies (Ref. Reference Byass8). The development and widespread application of vaccines and antiviral drugs have remarkably reduced the incidence of hepatitis B virus (HBV) infection, but the resultant number of chronic HBV patients and carriers remains enormous (Ref. Reference Trepo, Chan and Lok9). Gloomily, 2.4 billion people are consuming alcohol with 950 million designated as highly addictive, giving rise to a lifetime prevalence of alcohol use disorder as being 5.1–8.6% (Ref. Reference Asrani10). On the other hand, non-alcoholic fatty liver disease (NAFLD) has been reported to be epidemic in approximately 25% of the adult population but geographic variability (Ref. Reference Younossi11). Of note, the burden of NAFLD may become the leading cause of hepatic disorders during the coming decades in some countries/regions currently undergoing rapid urbanisation (e.g. Asia) (Ref. Reference Kabarra, Golabi and Younossi12). The increasing and high rate of NAFLD is proportionate with the occurrence of obesity, diabetes mellitus (DM) and metabolic diseases. As a matter of fact, NAFLD refers to a pathological defect belonging to metabolic syndrome manifested in the liver, inspiring scientific endeavour to explore whether exercise-derived irisin may act as a possible therapeutic target as far back as 2014. Although lots of attempts have been made to characterise the clinical implication of irisin in the context of liver diseases, literature data appear to be irreproducible and enigmatic probably due to flawed testing assays by using dramatically discrepant reference range and methodology (Ref. Reference Maak13). However, it has been well documented with regard to the anti-inflammatory, antioxidative and antiapoptotic activities by administering recombinant irisin or employing genetic depletion mice within different pathological conditions (Ref. Reference Askari14).

In this article, we comprehensively mine and review the literature data with regard to the purported myokine irisin and its precursor FNDC5 in the pathogenesis of several hepatic pathologies, if available, as the therapeutic scale. The collected information and retrieved findings in relation to basic research and clinical studies of FNDC5/irisin are depicted in the separated section for more explicit statement and better understanding for our authors. Some basic knowledge and future research perspectives are also discussed in this article.

Structure–function basis of FNDC5/irisin

In rodents FNDC5 comprising six exons encodes a protein of 209 amino acids. FNDC5 protein consists of a 39-aa cytoplasmic tail (171–209), a 21-aa transmembrane domain (150–170), a 92-aa FNIII domain (33–124) and a 28-aa N-terminal signal peptide (1–28) (Ref. Reference Young, Valaris and Wrann15). The N-terminal sequence, responsible for transporting the protein across the membrane, is then cleaved (Ref. Reference Schumacher16). Irisin is proposed to result from proteolytically cleaved transmembrane protein FNIII plus a 19-aa linker peptide (29–140). Overall, FNDC5 appears to be a highly conserved gene except for a mutant start codon referring to canonical ATG to ATA (Ref. Reference Raschke17). The crystal structure of irisin has been deciphered with a continuous inter-subunit β-sheet dimer at the FNIII domain. The sequence of irisin exhibits 100% conserved between rodents and humans. The 112-aa irisin polypeptide possesses a theoretical molecular weight of 12 600 in response to deglycosylation. Accordingly, the glycosylated irisin at two N-linked sites is supposed to increase its expected molecular weight to a total of 28 000. Plenty of analytical metrics have been instigated to detect and quantify FNDC5/irisin, including antibody-based as well as label-free methods (e.g. western blot, enzyme-linked immunosorbent assay [ELISA], protein-lipid chip assay and mass spectrometry [MS]). However, we are far away from getting valid and reproducible measurements due to drastically methodological variability and heterogeneity. Furthermore, the reported contents/levels in the existing literature of irisin in the circulation represented a wide range from pg/ml to μg/ml, leading to more discordance and ambiguities.

Signalling pathways pertaining to cellular activities of FNDC5/irisin

A multitude of signalling pathways have been addressed to regulate or be activated by FNDC5/irisin with the purpose of delineating its pathobiological roles. More recently, Rabiee et al. have summarised and provided an outstanding article with respect to potentially intricate pathways facilitating intracellular functions of irisin circumstantially (Ref. Reference Rabiee18). In that paper, the authors denoted that FNDC5/irisin exerts multifaceted roles including anti-inflammation, anti-tumour, anti-depression, maintenance of metabolic homeostasis and bridging crosslink between the muscle and other organs/tissues. Notably, we intended to provide insight into signalling pathways specific to diverse liver diseases contextually in the following sections. Herein we briefly discussed the assumed receptor of irisin, since this ligand–receptor interaction takes responsibility for activating downstream pathways in charge of cellular processes. Although the receptor specific to irisin is yet to be identified until now, Kim et al. showed irisin could bind to integrins, a family of transmembrane cell adhesion molecules mediating cell-matrix and intercellular interactions (Ref. Reference Kim19). Using crosslinking and MS on the cultured cells, their observations implicated that αVβ5, known as the favoured receptor, represents the most strong response among screening determination, which is further verified by following research (Refs Reference Estell20Reference Bi22). Further experiment indicated that CD81 serves as a partner in the integrins signalling by sensitizing the irisin–receptor interaction in HEK293T cells (Ref. Reference Oguri23). Intriguingly, accumulating evidence has proved αV integrins also play pivotal roles in the fibrogenic development, metastatic activity and anti-inflammatory response in a variety of liver diseases (Refs Reference Zhang24, Reference Sun25).

Association between FNDC5/irisin and NAFLD (and relevant metabolic disorders)

NAFLD covers a broad spectrum of hepatic pathology entities ranging from simple fatty infiltration in hepatic parenchyma to the potentially progressive type designated as non-alcoholic steatohepatitis (NASH). In alignment with global obesity epidemic, NAFLD and NASH have approached alarming levels worldwide. In T2DM subjects, 55% of the population is suggested to be accompanied with NAFLD closely relevant to visceral obesity, hyperlipidaemia and hypertension (Ref. Reference Kabarra, Golabi and Younossi12). Notably, NASH is histologically evidenced by diffused steatosis, multi-lobular inflammation, hepatocellular ballooning, appearance of Mallory–Denk bodies along with fibrosis pericellularly. About 15–20% patients with NASH may proceed to cirrhosis when compared with 5–10% advancing to other end-stage liver diseases like hepatocellular carcinoma (HCC) requiring transplantation (Ref. Reference Younossi26). The incidence of HCC among NASH subjects with cirrhosis is estimated to be 0.5–2.6% (Ref. Reference Huang, El-Serag and Loomba27). Nonetheless, HCC has occasionally been diagnosed among patients with NAFLD and metabolic syndrome rather than those with obvious fibrosis or cirrhosis, giving rise to a challenge regarding a lack of regular and well-scheduled surveillance. Actually, this subset is prone to more advanced malignancies with dismal outcome compared with HCC on account of chronic viral hepatitis load (Ref. Reference Hester28). Until now, there are no specific and available pharmacological regimens to cure NAFLD/NASH approved by the FDA, although some natural components with anti-inflammation and antioxidant capabilities might be promising candidates (Refs Reference Soleimani29, Reference Soleimani30). The gold standard therapy recommended by the major scientific associations is to lose weight through calorie restriction in addition to physical exercise (Ref. Reference Chalasani31). Keeping in view the property of FNDC5/irisin in response to exercise, it has greatly inspired scholars' interest to deepen the potentials by targeting this novel and pleiotropic myokine as a biomarker or treatment of choice.

Basic researches

Soon after the discovery of irisin, mounting attempts have been made to reveal its pathogenic facet and therapeutic application in the context of NAFLD and relevant metabolic disorders (Table 1). Park et al. incubated mouse AML12 hepatic cells and primary hepatocytes with palmitic acid (PA) to mimic insulin resistance and steatosis, and found recombinant irisin markedly suppresses PA-induced lipogenesis and hepatocellular lipid accumulation (Ref. Reference Park32). Moreover, the nuclear localisation, lipogenic regulator expression/transcriptional activities, oxidative stress and inflammatory biomarkers were all markedly alleviated upon irisin treatment. These protective effects of irisin against hepatic steatosis may be accomplished by suppressing protein arginine methyltransferase-3, nuclear factor κB (NF-κB) or p38 mitogen-activated protein kinase signalling pathways. Liu et al. showed FNDC5 deficiency aggravates hyperlipidaemia, hepatic fatty acid oxidation/lipogenesis, lipid accumulation and autophagy flux blockage, which can be effectively reversed by exogenous prescription or FNDC5 overexpression through modulating adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin complex 1/peroxisome proliferator-activated receptor α (PPARα) axis (Ref. Reference Liu33). Similarly, liver-derived irisin functioned in both autocrine and paracrine modalities to surpass lipogenesis and reverse steatosis, responding to constitutive androstane receptor by activating the AMPK pathway with dose response in primary human hepatocytes (Ref. Reference Mo34). Relief of hepatic steatosis was also observed in irisin transgenic mice in correspondence with reduced lipogenic gene expression including sterol regulatory element binding protein-1c, fatty-acid synthase and stearoyl-CoA desaturase-1 (SCD-1). The connection between AMPK activation and administered irisin has been well documented in relation to lesser hepatic lipid accumulation and glucose output, increased fatty acid oxidation/glycolysis in addition to reduced lipogenesis/gluconeogenesis within different experimental models (Refs Reference So and Leung35, Reference Tang36). By conducting in vitro study, it has been confirmed that irisin reduces hepatic triglyceride/glucose content, but increases glycogen content and promotes HepG2 cells survival (Ref. Reference So and Leung35). In addition, subcutaneous infusion of irisin significantly reduced plasma and hepatic cholesterol in vivo, which proposes a promising therapeutic target of irisin for hypercholesterolaemia (Ref. Reference Tang36). In mice subjected to a fructose-rich diet, Motta et al. found liver steatosis develops with adipocyte hypertrophy, and these detrimental events were relevant to altered gene expressions including FNDC5, PGC1α and PGC1β, all of which were efficiently prevented upon high-intensity interval training (HIIT) (Ref. Reference Motta37). The authors concluded that the beneficial influences of HIIT, in parallel to the medicinal properties of metformin, may partially alleviate the liver injury and prevent progressive NAFLD from more advanced pathological stages. In contrast, the addition of recombinant irisin had no effect on lipid contents in HepG2 cells exposed to oleic acid and PA in line with unchanged mRNA expression of PPARα and SCD, while no significant hepatic expression of irisin was observed in high-fat diet (HFD) inducing NAFLD mice model (Ref. Reference Petta38). In two NAFLD models challenged by HFD or a methionine- and choline-deficient (MCD) diet, Canivet et al. implied hepatic FNDC5 expression increases with steatotic changes and liver damages, whereas circulating irisin did not proportionate to consequential complications in the liver (Ref. Reference Canivet39). Both hepatocytes and non-parenchymal cells incorporating innate immune cells and activated HSC dominated the production of FNDC5 in fatty liver, wherein hepatocellular source was stimulated by genotoxic stress and p53-PGC1α signalling pathway. Knockdown of FNDC5 in HepG2 cells and in mouse primary hepatocytes resulted in increased expression of glucogenesis gene (phosphoenolpyruvate carboxykinase) and decreased expression of very-low-density lipoprotein synthesis genes (such as apolipoprotein B). Using in vitro modalities, it has also been demonstrated that FNDC5 can suppress hepatocellular fat accumulation by enhancing the export of lipids, facilitating autophagic flux in addition to decreasing the cytokine-mediated apoptosis. Another research intended to delineate the therapeutic potential of nicotinamide riboside (NR) against NAFLD in similarly built models (i.e. mice fed an HFD or MCD) between wild-type and FNDC5−/− mice (Ref. Reference Li40). Their findings implicated the FNDC5 gene expression is dramatically up-regulated in the skeletal muscle, adipose and liver tissue along with plasma irisin values upon NR treatment. The medicinal properties were evidenced by alterations with respect to hepatic steatosis, steatohepatitis, insulin sensitivity, mitochondrial dysfunction and apoptotic cell death, which were all compromised in FNDC5 knockout mice. Mechanistically, NR treatment displayed its hepatoprotective action by regulating sirtuin 2-dependent FNDC5 deacetylation and deubiquitination to foster protein stability as well as contents expressed. Moreover, the exerkine nature of irisin has been explored and corroborated in another HFD-induced NAFLD (Ref. Reference Zhu41). Accordingly, exercise training markedly ameliorated hepatic steatosis and fibrosis with concurrent enhancement in circulating irisin levels and muscular irisin expression. By using PA-incubated AML12 cells in vitro, it has been suggested that irisin is capable of disturbing myeloid differentiation factor 2-Toll-like receptor 4 (TLR4) complex formation to exert anti-inflammatory activities dose-dependently. Moreover, irisin could improve lipid metabolism and fibrosis by attenuating expression of fatty acid-binding protein 4, collagen type 1 and transforming growth factor-β (TGF-β). As a matter of fact, there are concerns about the synchronizing impacts and reciprocal reactions between NAFLD and other entities. For instance, Medhat et al. determined the influence of irisin in an experimental model suffering from NAFLD due to metabolic syndrome induced by a high-fat high-carbohydrate diet (Ref. Reference Medhat42). Their findings unveiled that administered irisin can attenuate dysregulated hepatic architecture characterised by diminished foam cells, foster energy expenditure via PGC1α and repress hepatic cholesterol synthesis relying on AMPK-sterol regulatory element-binding transcription factor 2 axis. In a study exploring the pathogenic link between NAFLD and inflammatory bowel disease, Kwon et al. showed dextran sodium sulphate-induced colitis leads to a significant down-regulation of the skeletal muscle-derived irisin accompanied by perturbing impacts on overall metabolic processes, ultimately presented as hepatic steatosis- and dyslipidaemia-like phenotypes (Ref. Reference Kwon43). MicroRNAs (miRNAs) belong to a setting of single-stranded, small-sized non-coding RNAs negatively modifying gene expression at the post-transcriptional level bound to the 3ʹ-untranslated region (UTR) of target miRNAs (Ref. Reference Alamdari44). Diversely dysregulated miRNAs expressions have been linked to the pathological advancement in patients with NAFLD (Ref. Reference Gjorgjieva45). Most recently, Yu et al. unveiled miR-665-3p binds directly to the 3ʹ-UTR of FNDC5, in consequence down-regulates its expression, inactivates the downstream AMPKα signalling pathway, and thereafter mediates oxidative stress, inflammatory reaction and NAFLD progression (Ref. Reference Yu46). Correspondingly, increased hepatic irisin levels were found by antagonizing miR-665-3p. Knockdown of endogenous FNDC5 expression in the liver abrogated the miR-665-3p antagomir-mediated protection, which was evidenced by increased serum transaminase levels and hepatic hydroxyproline content. A schematic diagram was depicted in Figure 1.

Fig. 1. Schematic diagram depicting the pathogenic role and therapeutic potential of FNDC5/irisin in experimental NAFLD models. FNDC5, fibronectin type III domain-containing protein 5; HFD, high-fat diet; MCD, methionine- and choline-deficient; SIRT, sirtuin; UCP1, uncoupling protein 1; PA, palmitic acid; AMPK, adenosine monophosphate-activated protein kinase; mTORC1, mammalian target of rapamycin complex 1; PPARα, peroxisome proliferator-activated receptor α; TLR4, Toll-like receptor 4; MD2, myeloid differentiation factor 2; PGC1α, proliferator-activated receptor-γ co-activator 1α; NAD+, nicotinamide adenine dinucleotide+; MyD88, myeloid differentiation factor 88; MAPK, mitogen-activated protein kinase.

Table 1. The pathogenic role and therapeutic potential of FNDC5/irisin in experimental NAFLD models

FNDC5, fibronectin type III domain-containing protein 5; HFD, high-fat diet; N.A., not applicable; T2DM, type 2 diabetes mellitus; PI3K, phosphoinositide 3-kinase; FOXO-1; forkhead box transcription factor O1; GSK3, glycogen synthase kinase-3; PEPCK, phosphoenolpyruvate carboxykinase; G6Pase, glucose-6-phosphatase; GS, glycogen synthase; PRMT3, protein arginine methyltransferase-3; AMPK, adenosine monophosphate-activated protein kinase; mTORC1, mammalian target of rapamycin complex 1; PPARα, peroxisome proliferator-activated receptor α; HSCs, hepatic stellated cells; MCDD, methionine- and choline-deficient diet; SIRT, sirtuin; NAD+, nicotinamide adenine dinucleotide+; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor-κB; IBD, inflammatory bowel disease; DSS, dextran sodium sulphate; miR, microRNA; UTR, untranslated region.

DM is a major metabolic disorder influencing multiple organs/tissues, and the generally acting mechanisms designated as insulin resistance and in consequence hyperglycaemia may give rise to NAFLD or NASH by interrupting carbohydrate, lipid and protein metabolisms and HCC in circumstances (Ref. Reference Chen47). In obese rats challenged by HFD, exercise and exogenous supplementation with potent antioxidant aged garlic extract exerted no influence on liver/gastrocnemius muscle masses in line with unchanged plasma irisin or skeletal muscle FNDC5 contents, counteracting its impact on homeostatic metabolism (Ref. Reference Seo48). In vitro study showed that irisin significantly dampens gluconeogenesis and enhances glycogenesis in HepG2 cells and mouse primary hepatocytes undergoing insulin resistance induced by glucosamine or palmitate via activation of the phosphoinositide 3-kinase (PI3K)/Akt pathway, which is mechanistically shared with insulin (Ref. Reference Liu49). Niranjan et al. also treated HFD-triggered obesity mice with recombinant irisin presented with significant up-regulation of uncoupling protein 1 (UCP1) in a dose-respondent manner (Ref. Reference Niranjan50). The pivotal role of UCP1 in dissipating energy in the form of heat rather than stored fat depots may link the pathophysiological facet regarding irisin to mitigate hepatic steatosis. Another metabolic and inflammatory regulation mechanism of irisin relied on its effect on brain-derived neurotrophic factor, fatty acid binding protein 4 and PPARγ in spleen. During the last 10 years, there has been a surge delineating the involvement of gut microbiota to the pathogenesis of obesity and its complications including DM, obesity, hyperlipidaemia and NAFLD (Refs Reference Cui7, Reference Canfora51). In this regard, Kwon et al. stated the Lactobacillus plantarum strain exerts favourable metabolic regulations including improvement in hepatic steatosis in HFD-induced obese mice along with up-regulated irisin mRNA expression in the skeletal muscle and subcutaneous fat tissue (Ref. Reference Kwon52). The protective effect of L. plantarum to improve lipid oxidation and alleviate adipose accumulation was implemented via SIRT1/PGC1α signalling pathway in addition to fighting against metabolic dysregulation and favouring hepatic cholesterol clearance (Ref. Reference Chang and Guarente53).

Two research teams used substances from natural products of choice, namely silymarin (belongs to flavonoid) and genistein (belongs to isoflavone) for treating metabolic disorders-related deleterious liver damages. Their findings demonstrated that these medicinal agents can significantly enhance serum irisin concentration and hepatic FNDC5 expression responsible for unobvious histopathological changes in the parenchyma and amended metabolism homeostasis (Refs Reference Kheiripour54, Reference Shen55). A plethora of hepatoprotective machinery is pointed out including antioxidant effects, anti-endoplasmic reticulum (ER) stress as well as anti-inflammatory activities. In this regard, garlic oil, a natural constituent, mitigated the diabetic liver injury by repressing oxidative stress-related culprits and modulating serum irisin values along with PGC1α, FNDC5, p38 in liver tissue to maintain insulin sensitivity and balance carbohydrate metabolism (Ref. Reference Eser56).

Clinical studies

As far back as 2013, Zhang et al. showed for the first time serum irisin levels are inversely associated with intrahepatic triglyceride contents and significantly decreased among NAFLD in 296 obese Chinese adults (Ref. Reference Zhang57). Furthermore, inverse correlation between circulating irisin levels and liver enzymes (i.e. ALT and AST) reflected a protective role of irisin against hepatic steatosis. In the same year, Polyzos et al. reported higher systemic irisin contents in lean controls when compared with obese subjects, biopsy-proven patients with simple steatosis and NASH participants (Ref. Reference Polyzos58). However, no difference was observed in patients with simple steatosis relative to NASH, despite higher irisin levels appeared to be independently associated with obvious portal inflammation (36.7 versus 30.8 ng/ml). Similarly, in a setting of patients with NAFLD, T2DM or their concurrence, the plasma irisin levels exhibited gradual reduction in that order (2.18 versus 1.96 versus 1.45 μg/ml) with highest values in healthy controls (Ref. Reference Shanaki59). It is suggested that irisin can predict liver stiffness independently, and elevated circulating irisin levels may reduce the risk of these metabolic disorders in addition to moderately diagnostic utility for NAFLD. Waluga and colleagues consistently addressed lower plasma irisin concentrations in the NAFLD group in comparison with healthy control and an inverse correlation with grading inflammation in NAFLD, which was contrary to the results by Polyzos (Ref. Reference Waluga60). The authors speculated the severity of liver histopathological lesion dominates these relationships.

Oppositely, another study indicated the serum irisin levels in NAFLD group are higher than those of control group, suggesting increased irisin as a defensive partner during the early stage of NAFLD. This compensatory action would be exhausted; simultaneously, the steatotic advancement is accompanied by a decrease in irisin contents (Ref. Reference Choi61). These observations were verified in patients with metabolic syndrome and concurrent fatty liver, whose systemic irisin concentrations were drastically up-regulated and represented a positive correlation with liver enzymes, homeostatic model assessment of insulin resistance index (HOMA-IR), anthropometry data as well as serum triglycerides (Ref. Reference Rizk, Elshweikh and Abd El-Naby62). Likewise, male HIVs without diabetes had higher irisin concentrations correlated to HOMA-IR and NAFLD identified by higher hepatic triglyceride contents via 1-H magnetic resonance spectroscopy (Ref. Reference Moreno-Perez63). Another study reported morbidly obese patients with hepatic steatosis and NASH have up-regulated FNDC5 mRNA expression in liver, closely relevant to the grade of steatosis, NAFLD activity score and ALT levels, rather than impact on the systemic levels of irisin (Ref. Reference Canivet39). Taken together, unlike the local involvement of FNDC5 to the onset and progression of NAFLD, increased and decreased systemic irisin have both been addressed in the existing literature. One justified interpretation refers to muscle-derived irisin constituting approximately 70% of the total circulating protein levels, while sarcopaenia (muscle wasting) has a reciprocal relationship with NAFLD as being a risk factor or a complication (Refs Reference Perakakis3, Reference Bhanji64). More recently, Armandi et al. denoted circulating irisin levels are increased (in the absence of sarcopaenia) in individuals with marked fibrosis and correlated with liver fibrogenesis, potentially identifying a more aggressive phenotype of liver disease with severe damage (Ref. Reference Armandi65). The authors speculated that a harmonic relationship between irisin and liver fibrogenesis, indicative of hepatic response to the inflammatory injury, may be attributed to the selection of a well-characterised cohort of biopsy-proven NAFLD for the absence of the major metabolic confounders. It is suggested that metabolic burdens such as DM and obesity could have affected the circulating irisin pool. Therefore, we believe future larger studies are merited to further uncover the association between circulating irisin levels and liver damages in patients with NAFLD.

Emerging evidence suggests a variety of inherited factors, especially single-nucleotide polymorphisms (SNPs) may potentiate the susceptibility of individual regarding the development and progression as well as therapeutic response in the context of NAFLD (Ref. Reference Martinez-Montoro66). However, no association was observed between FNDC5 rs3480 and anthropometric, metabolic and biochemical indices on the basis of 593 liver biopsy-confirmed NAFLD participants compared to healthy controls (Ref. Reference Petta38). In contrast, according to another investigation enrolling 987 Caucasian NAFLD patients, Metwally et al. reported FNDC5 rs3480 minor (G) allele associates with advanced steatotic changes, an independent variant but additive to patatin-like phospholipase domain-containing 3 and the transmembrane 6 superfamily member 2 responsible for hepatic fat accumulation (Ref. Reference Metwally67). Alteration of the above-described three variants concomitantly with ≥1 copy at respective region increased hepatic steatosis risk by 1.4- to 6.8-fold in comparison with subjects without any risk allele. The underpinning mechanism unravels marked FNDC5 mRNA degradation embracing G allele (versus A allele), which was bound with miR-135a-5P at its SNP in the 3’UTR resulting in impaired FNDC5 expression. Accordingly, NAFLD patients represented higher hepatic contents of miR-135a-5P and lower FNDC5, but increased serum irisin levels closely relevant to improved steatosis and metabolic profiles in the circulation. Taken together, abovementioned discordance among studies with respect to inherited determinants facilitating NAFLD probably results from divergent recruited population, different disease stage/severity and discordant detecting approaches, which warrant further validation with solid evidence.

From therapeutic perspectives, a clinical trial indicated green cardamom supplementation exerts medicinal influences on fatty liver grading, serum glucose parameters and lipid components within obese NAFLD patients, which can be mediated by increased SIRT1 and irisin levels (Ref. Reference Daneshi-Maskooni68). However, another study conducted on NAFLD patients showed α-lipoic acid, a cofactor for mitochondrial enzymes with multifaceted properties, administered for 12 weeks leads to improved insulin resistance and serum adipokines without affecting anthropometry data, liver steatosis intensity and irisin levels (Ref. Reference Rahmanabadi69).

Fibrosis/cirrhosis

Hepatic fibrosis stands for a common pathology subsequent to a majority of chronic liver diseases, ultimately giving rise to cirrhosis, liver failure and in more aggressive type like HCC (Ref. Reference Trautwein70). The hallmark and cornerstone pertaining to the pathogenesis of hepatic fibrosis is the activation of hepatic stellate cell (HSC) and in consequence diffused deposition of extracellular matrix (ECM) proteins. Emerging evidence implicates the fibrotic ECM can undergo remodelling and convert into near-normal hepatic architecture by injury cessation (Ref. Reference Ellis and Mann71). Thus it is practical to terminate the fibrogenic process aimed at slowing progressive fibrosis and even cirrhosis (Ref. Reference Elpek72).

Basic researches

Petta et al. showed irisin expression increases in line with HSC activation, evident by collagen α1 and α-smooth muscle actin (α-SMA) mRNA expression (Ref. Reference Petta38). Furthermore, exogenous irisin triggered profibrogenic genes monocyte chemoattractant protein-1 (MCP-1) and tissue inhibitors of metalloproteinases-1 (TIMP-1) in activated HSCs, and its expression was up-regulated in experimental hepatic fibrosis exposed to carbon tetrachloride (CCl4). In a study conducted by Zhou et al., FNDC5 deficiency exacerbated hepatic fibrosis induced by HFD in mice (Ref. Reference Zhou73). The underpinning molecular basis was built on AMPK phosphorylation-mediated inhibition of HSCs activation and up-regulation of connective tissue growth factor/TGF-β. Using oxidised low-density lipoprotein triggered primary mouse HSCs and human LX-2 cells in vitro, exogenous FNDC5 treatment also effectively mitigated HSC activation and ECM deposition indicative of gradually down-regulated α-SMA and collagen expression. Additionally, the medicinal potency of irisin for improving fibrogenic process was corroborated in immortalised LX-2 cells via modifying HSC activation, proliferation, migration, contractility and production of interleukin-(IL-)1β/IL-6, given the fundamental participation of inflammation in activating and trans-differentiating resting HSC to myofibroblasts (Ref. Reference Dong74). More recently, Liao et al. used recurrent CCl4 exposure to establish in vivo fibrosis model and clarified that co-treatment with irisin remarkably attenuates collagen deposition and the expression of fibrogenic biomarkers in mice (Ref. Reference Liao75). This therapeutic approach was in part attributed to inhibitory ER stress via regulating PKR-like ER kinase mediated heterogeneous nuclear ribonucleoprotein A1 destabilisation. A schematic diagram was depicted in Figure 2.

Fig. 2. Schematic diagram depicting the pathogenic role and therapeutic potential of FNDC5/irisin in experimental fibrosis models. FNDC5, fibronectin type III domain-containing protein 5; HSC, hepatic stellate cell; ECM, extracellular matrix; AMPK, adenosine monophosphate-activated protein kinase; α-SMA, α-smooth muscle actin; TGF-β, transforming growth factor-β; IL-, interleukin-; HFD, high-fat diet; CCl4, carbon tetrachloride; LPS, lipopolysaccharide; PERK, PKR-like ER kinase; HNRNPA1, heterogeneous nuclear ribonucleoprotein A1; oxLDL, oxidised low density lipoprotein.

Clinical studies

In a cohort of 593 patients prone to NASH, FNDC5 rs3480 A > G was in relation to protective impact against clinically significant fibrosis (Ref. Reference Petta38). Mechanistically, the authors proposed FNDC5 rs3480 may influence fibrosis by regulating irisin expression mainly derived from HSCs and adipocytes, in other words, irisin from distinct sites/origins may enter into plasma to elicit proinflammatory (e.g. MCP-1) and fibrogenic (e.g. TIMP-1) factors and consequently hepatic fibrogenesis.

Like elusive role of serum irisin in the context of NAFLD, the association between circulating irisin and liver cirrhosis remains unclear. A clinical study recruited 43 virus-related liver cirrhosis of whom 36 participants had HCC (Ref. Reference Pazgan-Simon76). The authors showed cirrhotics with concomitant HCC exhibit lower irisin concentrations than do healthy controls, although non-significantly lower irisin levels were observed in patients at more severe stage stratified by Child–Pugh classification. Kukla et al. intended to analyse the relationship between serum irisin levels and sarcopaenia in a total of 88 patients with decompensated cirrhosis with median model for end-stage liver disease (MELD) scores of 15 points (Ref. Reference Kukla77). Their findings suggested there is no marked difference regarding serum irisin levels among cirrhotic patients stratified by Child–Pugh classification and MELD score. Moreover, despite irisin primarily standing for a myokine, it did not correlate with sarcopaenia defined by low handgrip strength or central body muscle loss in foresaid work. On the other hand, existing insulin resistance may explain less relevance between cirrhosis and complication/comorbidity. On the contrary, Zhao et al. performed a cross-sectional study on 187 patients with cirrhosis, among whom a total of 39% presented sarcopaenia, and described plasma irisin as an independent biomarker relevant to sarcopaenia (Ref. Reference Zhao78). Other novel findings included positive correlation between skeletal muscle index at the third lumbar vertebra, namely an index of sarcopaenia, as well as correspondingly decreased irisin levels with worsening hepatic function (Ref. Reference Hou79). Collectively, it must be noted that all described studies were on the basis of relatively small sample size and their retrospective nature in design. The extremely wide reference values of retrieved irisin levels in serum (from pg/ml to μg/ml) and methodological heterogeneity may give rise to inconsistent results.

Various liver injury and relevant illness

Liver injury provides a general idea of hepatocytes damage resulting from a wealth of detrimental insults, including but not limited to toxicants, chemical agents, drugs, viral infection as well as binge. Additionally, it can be presented as accompanying symptom under deleterious attack to other organ/tissue. Within persistent and intensive pathobiological stress, hepatic damage proceeds to advanced or end-stage disease such as liver failure (Ref. Reference Wang80). Of note, acute liver failure brings high mortality rates due to insufficient transplant organ allocation and relatively limited treatment of choice.

Inadequate coronary blood supply against normal myocardial demand results in the decease of myocardial tissue designated as myocardial infarction (MI), which also leads to significant liver function abnormality (Refs Reference Barbagelata81, Reference Thirupurasundari, Jayalakshmi and Niranjali Devaraj82). Kuloglu et al. demonstrated that liver irisin levels, generated from both hepatocytes and sinusoidal cells, drastically decrease in the presence of MI induced by isoproterenol and considered decreased irisin as an uncoupling agent in the circulation to surpass ATP consumption and maintain cell viability by inhibiting necrotic process (Ref. Reference Kuloglu83). Intriguingly, a similar study conducted by Aydin et al. on myocardial ischaemic rats implied both iloprost and sildenafil can reverse down-regulated irisin immunoreactivity in the hepatocytes of liver tissue (Ref. Reference Aydin84). These tissue ischaemia-eliminating effects to some extent accelerated the blood supply through vasodilatation, cellular reoxygenation and enhanced irisin synthesis. The same research team clarified formaldehyde, a normal metabolite conditionally deleterious to liver tissue, increases free radical and poses oxidative stress on hepatocytes manifested by cell death in dose-dependent way (Ref. Reference Aydin85). The authors hypothesised the damaged cells may circumvent irisin synthesis aimed at preventing energy waste, evident from the reduced irisin in liver and the circulation. Furthermore, the histopathological and biochemical signs due to formaldehyde exposure were restored upon carnosine supplementation, a potent antioxidant capable of scavenging reactive oxygen species, which was usually found in the muscle and brain tissues. Accordingly, the antioxidant activity of irisin was validated. Further research intended to explore the relationship between irisin activity and methotrexate, known as a chemotherapeutic and immunosuppressive agent, triggered hepatotoxicity in rats (Ref. Reference Erdogan and Yalcin86). It was found that an increase in irisin activity is helpful for diminishing oxidative stress against hepatic toxicity and compensating for the increased energy demand, evident from reduced immunoreactivity of irisin in liver responding to benfotiamine (a thiamine pyrophosphate metabolite with antioxidant effect). CCl4 has been widely used to elicit hepatotoxicity experimentally and characterised by multiple biological facets including lipid peroxidation, oxidative stress, autophagy and ferroptotic cell death (Refs Reference Unsal, Cicek and Sabancilar87, Reference Zhao88). Rabey et al. reported CCl4 toxicity promotes inflammatory reaction at aggravated site of liver damage and potentiates oxidative stress (Ref. Reference Rabey89). However, green coffee methanolic extract exerted hepatoprotection by improving irisin and other antioxidants to reverse liver dysfunction, and converting inflammatory cytokines/oxidative stress indices to their normal values in the exposed rats. These medicinal effects are in alignment with other natural components such as propolis in the context of liver injury (Ref. Reference Pahlavani90).

Ischaemia-reperfusion (I/R) injury is subsequent to inflammatory processes in the case of target organ undergoing a cessation of blood flow or transient reduction, followed by perfusion re-establishment. Following liver surgery, haemorrhagic shock and severe sepsis, liver I/R injury results in organ dysfunction, liver failure and vice versa. Importantly, it represents a predisposing factor to the onset of graft rejection during liver transplantation (Ref. Reference Lu91). There is currently no clinically applicable therapeutics specific to I/R injury with the exception of supportive care medicine. Bi et al. demonstrated exogenous irisin markedly improves hepatic function, inhibits cell death and relieves inflammatory response in the setting of liver I/R (Ref. Reference Bi92). It is suggested that mitochondria constantly undergoes fission and fusion, which is biologically essential to maintain internal homeostasis (Ref. Reference Youle and van der Bliek93), and irisin significantly restrains mitochondrial fission-related protein, dynamin-related protein 1 and fission 1 for normally functioning. On the contrary, the mitochondrial content/biogenesis was fostered along with quenched oxidative stress by regulating PGC1α and uncoupling proteins 2 expression. In one study performed on hepatic I/R in HFD-fed mice, administered irisin ameliorated hepatic injury, eradicated oxidative/ER stress and restored mitochondrial function in steatotic liver (Ref. Reference Zhang94). However, inhibition of kindlin-2, an important regulator of αVβ5 integrin function, by RNA interference offsets irisin's influence on aforementioned cellular mechanisms in cultured hepatocytes. Intestinal I/R can give rise to local and remote tissue/organ necrosis, rendering it to be vital in the progressively deteriorated state of an illness (Ref. Reference Jing95). Notably, liver is among the most vulnerable organs after intestinal I/R, since these two organs share a common anatomical passage (Ref. Reference Saidi96). In rats with concurrent intestine and liver injuries, Fan et al. unravelled decreased circulating irisin levels in contrast to increased hepatic irisin and inflammasome expressions after intestinal ischaemia perfusion (Ref. Reference Fan97). In addition, plasma irisin contents inversely correlated with hepatic damage severity and proinflammatory cytokines such as IL-6/tumour necrosis factor-(TNF-) α. Looking into the therapeutic potential, an analgesic dexmedetomidine can promote irisin expression to alleviate intestinal I/R-mediated liver injury by dampening NOD-like receptor protein 3 (NLRP3) inflammasome activation. Intriguingly, irisin-treated hepatic I/R old rats exhibited restored reparative capacity (Ref. Reference Bi98). This conspicuous benefit of irisin may be attributed to the activation of autophagy and in consequence telomerase activity via regulating phosphorylated JNK during hepatic I/R advancement.

Infection-associated liver injury

It has been well documented that severe acute pancreatitis can lead to the development of systemic inflammatory response syndrome, which imposes additional burden on multiple organs incorporating liver and the intestine (Refs Reference Cheng99, Reference Tan100). In L-arginine-induced AP mice, intraperitoneal injection of irisin can effectively mitigate intestinal cell injury by blocking ER stress and diminishing oxidative stress in combination with attenuated hepatic injury (Ref. Reference Ren101). Sepsis exhibits a leading cause of in-hospital mortality and morbidity, and early liver dysfunction serves as an independent risk factor for dismal outcomes and deterioration of sepsis (Refs Reference Rhee102, Reference Kobashi, Toshimori and Yamamoto103). A multitude of regulated cell death (RCD) has been linked to the development and progression of sepsis, including ferroptosis and pyroptosis. Ferroptosis is a type of RCD sensitive to iron accumulation which is then executed by glutathione peroxidase 4 (GPx4) deletion and subsequently overwhelming lipid peroxidation, while pyroptosis a lytic form triggered by invasive pathogens/tissue injury to amplify inflammation. Both described RCD modalities are involved in the pathogenesis of various liver diseases (Refs Reference Mao104, Reference Knorr, Wree and Feldstein105). In this context, the interplay between RCD and septic liver injury has been concerns of scientific endeavour. Wei et al. stated serum levels of irisin decrease in septic patients (12.85 versus 16.35 ng/ml), inversely correlated with disease severity, and in septic mice subjected to caecal ligation and puncture (CLP) (Ref. Reference Wei106). Overall, these observations indicated that there may be an irisin deficiency in sepsis. Moreover, ferroptosis were instigated in septic mice liver indicative of decreased GPx4 expression, Fe2+ accumulation and increased malondialdehyde content, whereas exogenous irisin treatment remarkably reversed their expression and mitigated mitochondrial impairment in morphology. These protective effects of irisin against ferroptosis were eliminated by inhibiting GPx4 or blocking αV integrin in lipopolysaccharide- (LPS-) treated hepatocytes and CLP-induced septic mice. Li et al. denoted that irisin intervention counters LPS-mediated septic liver injury by eradicating NF-κB signalling and downstream components responsible for inflammasome assembly and the activation of pyroptosis, such as caspase-1, NLRP3 and gasdermin D at both animal and cell levels (Ref. Reference Li107).

Hepatocellular carcinoma

HCC is one of the most common liver cancers worldwide. It carries a variety of risk factors, including alcohol addiction, concurrent NAFLD, obesity, chronic viral hepatitis infection, iron accumulation and family history (Ref. Reference Erstad and Tanabe108). A variety of multifunctional proteins have been proposed to contribute to HCC development, progression, metastasis and recurrence such as midkine, forkhead box O and osteopontin (Refs Reference Gowhari Shabgah109, Reference Yang110, Reference Wen111). Although great efforts have been carried out with respect to therapeutic strategy and supportive management for patients with HCC, such as surgery, chemotherapy, interventional therapy, immunotherapy and liver transplantation, the survival rate remains approximately 3–5% in developed countries. Thus, it is crucial to figure out more effective treatment by concentrating on novel molecular targets and underpinning mechanisms.

Basic researches

Shi et al. showed irisin enhances two HCC cell lines, namely HepG2 and SMCC7721, proliferation, migration and invasiveness in a dose-dependent manner through activating PI3K/Akt signalling pathway as evidenced by opposing impact after LY294002 (a PI3K inhibitor) administration (Ref. Reference Shi112). Mounting evidence has proved the PI3K/Akt pathway is involved in antiapoptotic activity during HCC development (Ref. Reference Sun113). Accordingly, treatment with irisin rendered protective impact on HepG2 apoptotic cells evoked by doxorubicin, which indicates the detrimental role of irisin pertaining to hepatic malignancy progression and decreased sensitivity to chemotherapy. Given the hallmark of tumour microenvironment on biological actions within HCC, it is appealing to modulate this peri-cancerous tissue as a novel therapeutic scale (Ref. Reference Pathria, Louis and Varner114). Notably, inflammation has become the hallmark as featured malignancy, and it is ascertained that tumour-associated inflammation facilitates the contribution of monocyte in HCC growth and metastasis. Tumour-associated macrophages (TAMs) play a pivotal role in crosstalk between tumour and stromal cells, actively participating in tumour-related inflammation (Ref. Reference Arvanitakis115). By using co-culture system embracing both HCC cells and TAMs, the same research group denoted HepG2/SMCC7721-derived FNDC5 overproduction results in increased M2 phenotype and decreased M1 phenotype in THP-1 induced macrophages (Ref. Reference Liu116). Further mechanistic investigation implied that high FNDC5-expressed HCC cells may influence the PPARγ/NF-κB/NLRP3 pathway in TAMs, in consequence, promoting M2 polarisation.

Clinical studies

Initially, Aydin et al. determined irisin immunoreactivity in various gastrointestinal cancers, and showed strong staining in all tissues except for liver (Ref. Reference Aydin117). No significant histoscores of irisin were observed between HCC and normal liver tissue. On the contrast, another two investigations reported discordant findings. Gaggini et al. found the highest expression of hepatic FNDC5 mRNA in patients with HCC relative to the transplantation donors (Ref. Reference Gaggini118). Moreover, hepatic FNDC5/irisin was transcriptional associated with sterol regulatory element-binding factor 1, SCD-1, proinflammatory cytokines (i.e. TNF-α/IL-6) and neurogenic locus notch homolog protein 1, all of which accounted for lipogenesis, inflammation and tumourigenesis. Mechanistically, the overexpressed FNDC5/irisin may exert compensatory effect and protective role to limit de novo lipogenesis during HCC progression as a paracrine hormone. Consistent with prior results, Shi et al. also indicated FNDC5 levels increase in the hepatic tissue of HCC patients (Ref. Reference Shi112). Intriguingly, no correlation was found between circulating irisin levels and hepatic FNDC5/irisin according to described studies, probably due to multiple origins of this myokine via distinct regulation mechanisms (Ref. Reference Varela-Rodriguez119). Nevertheless, a study recruiting 117 HCC participants and 102 healthy controls measured preoperative irisin contents by ELISA (Ref. Reference Zhang120). The authors demonstrated irisin levels (at μg/ml) decrease in patients with HCC undergoing hepatectomy on the basis of enrolled population as well as the Cancer Genome Atlas cohort, and supposed lower irisin expression may prevent patients from cachexia through restricting energy consumption. Another study further corroborated reduced serum irisin levels in HCC patients compared to controls (2.52 versus 4.46 μg/ml, P = 0.02) and clarified its negative correlation with more advanced HCC grade (Barcelona clinic liver cancer classification) and impaired live function reserve (Child–Pugh classification) (Ref. Reference Pazgan-Simon121). Supposable, a decrease of systemic irisin may promote invasive and metastatic behaviours of HCC by suppressing epithelial-to-mesenchymal transition and resultant fibrosis. Taken together, we are currently as its infancy to regard irisin as a surrogate associated with poor outcomes in the context of HCC taken into consideration of existing literature data (Ref. Reference Kim122).

Intrahepatic cholestasis of pregnancy

Intrahepatic cholestasis of pregnancy (ICP) is the most common pregnancy-specific liver disorder typically commencing in the late second and third trimester, which can dissolve within 48 h subsequent to delivery of the foetus (Refs Reference Williamson and Geenes123, Reference Geenes and Williamson124). It is characterised by unexplained mild to severe itching, increased serum bile acids and abnormal liver functional tests. Emerging evidence has suggested that more prevalent ICP is associated with abnormal metabolic profiles, including glucose intolerance and dyslipidaemia secondary to maternal aberrant bile acid homeostasis (Ref. Reference Menzyk125). Kirbas et al. conducted a cross-sectional study on 59 consecutive pregnant ICP women, and stated maternal systemic irisin levels are markedly higher in ICP patients relative to healthy controls (Ref. Reference Kirbas126). By using an established cut-off, that is 908.875 pg/ml with 72.5% sensitivity and 86.8% specificity, the risk of cholestasis appeared to be approximately 16 times higher (odds ratio = 16.972, P < 0.001). One interpretation built on these changes referred to irisin's adaptive and compensatory response to attenuate metabolic disturbances within ICP progression. Similarly, Chen et al. also demonstrated the values of irisin in the serum and umbilical vein blood increases with more aggressive ICP severity (Ref. Reference Chen, Li and Ma127). Serum irisin levels may serve as a proxy for diagnosing and indexing ICP subjects along with its role in diminishing oxidative stress and improving lipid metabolism in the pathogenesis of ICP.

Conclusions and future perspectives

Accumulating data have implicated liver diseases are on the steep rise and account for a large amount of deaths worldwide. Understanding the precise role of a target protein at the cellular, tissue and systemic levels is of upmost significance to dissect its potentials as therapeutic approach or prognostic/diagnostic surrogate. After a decade of extensive investigation and research on FNDC5/irisin, we are still surrounded by a gamut of open questions. FNDC5, as an overlooked transmembrane protein with a sleeping beauty-like fate in 2012, was kissed awake to be the precursor of irisin, known as a systemic exerkine/myokine with multiple origins. Since then, this hormone-like polypeptide has rapidly evolved into a component significantly involved in amazingly diverse diseases and metabolic dysregulations (Refs Reference Perakakis3, Reference Cao128, Reference Zhao129). In this article, we first concentrated on the structure–function relationship of FNDC5/irisin. Next, we comprehensively summarised the current knowledge and findings regarding pathogenic roles/therapeutic applications of FNDC5/irisin in the context of NAFLD, fibrosis, liver injury due to multiple detrimental insults and hepatic malignancy. Furthermore, the prominent molecules involved in underlying mechanisms and signalling pathways were highlighted. As a result, emerging evidence reveals FNDC5/irisin may act as a surrogate for diagnosing liver pathology, a sensitive biomarker for assessing damage severity, a predisposing factor for surveilling illness progression and a treatment option with protective/preventive effect, all of which are highly dependent on disease grading and contextually pathological features.

Although much progress has been made in our understanding of the contribution of FNDC5/irisin to the onset and development of liver diseases, several critical issues are supposed to be addressed for directing future investigation. First, there is a substantial gap between basic researches and clinical studies according to existing literature data, probably on account of flawed methods for detection and quantification. Additionally, the dosage of recombinant irisin applied to cells or injected to animals appears to exceed normal range among human beings in physiological context, hindering the smooth translation of available findings to clinical practice. Second, the medicinal impact of irisin to treat liver diseases is likely specific to cell types, with distinct effect on parenchymal versus non-parenchymal cells within liver tissue. More precise manipulation of hepatocytes subroutine is merited. Last, it is more practical and beneficial to target irisin in certain hepatic pathological condition and disease stage aimed at maximizing therapeutic efficacies and avoiding adverse events. Our capability to achieve selectivity when targeting this protein will be challenging in this direction.

Acknowledgements

None.

Author contributions

Xiaoyu Wang: writing – original draft, visualisation. Lihong Mao: writing – original draft, visualisation. Chaoqun Li: writing – original draft, visualisation. Yangyang Hui: conceptualisation. Zihan Yu: conceptualisation. Mingyu Sun: conceptualisation. Yifan Li: conceptualisation. Gaoyue Guo: conceptualisation. Wanting Yang: conceptualisation. Binxin Cui: conceptualisation. Xiaofei Fan: conceptualisation. Chao Sun: conceptualisation, writing – review and editing, supervision.

Financial support

This research received no specific grant from any funding agency, commercial or not-for-profit sectors.

Conflict of interest

None.

Footnotes

*

These authors have contributed equally to this work and share first authorship.

References

Aydin, S et al. (2014) A comprehensive immunohistochemical examination of the distribution of the fat-burning protein irisin in biological tissues. Peptides 61, 130136.10.1016/j.peptides.2014.09.014CrossRefGoogle ScholarPubMed
Bostrom, P et al. (2012) A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 481, 463468.10.1038/nature10777CrossRefGoogle ScholarPubMed
Perakakis, N et al. (2017) Physiology and role of irisin in glucose homeostasis. Nature Reviews. Endocrinology 13, 324337.10.1038/nrendo.2016.221CrossRefGoogle ScholarPubMed
Huh, JY et al. (2014) Irisin stimulates muscle growth-related genes and regulates adipocyte differentiation and metabolism in humans. International Journal of Obesity 38, 15381544.10.1038/ijo.2014.42CrossRefGoogle ScholarPubMed
Ma, EB et al. (2019) Irisin exerts inhibitory effect on adipogenesis through regulation of Wnt signaling. Frontiers in Physiology 10, 1085.10.3389/fphys.2019.01085CrossRefGoogle ScholarPubMed
Xiong, XQ et al. (2015) FNDC5 overexpression and irisin ameliorate glucose/lipid metabolic derangements and enhance lipolysis in obesity. Biochimica et Biophysica Acta 1852, 18671875.10.1016/j.bbadis.2015.06.017CrossRefGoogle ScholarPubMed
Cui, B et al. (2022) Therapeutic potential of Saccharomyces boulardii in liver diseases: from passive bystander to protective performer? Pharmacological Research 175, 106022.10.1016/j.phrs.2021.106022CrossRefGoogle ScholarPubMed
Byass, P (2014) The global burden of liver disease: a challenge for methods and for public health. BMC Medicine 12, 159.CrossRefGoogle ScholarPubMed
Trepo, C, Chan, HL and Lok, A (2014) Hepatitis B virus infection. Lancet 384, 20532063.10.1016/S0140-6736(14)60220-8CrossRefGoogle ScholarPubMed
Asrani, SK et al. (2021) Reducing the global burden of alcohol-associated liver disease: a blueprint for action. Hepatology 73, 20392050.10.1002/hep.31583CrossRefGoogle Scholar
Younossi, ZM et al. (2016) Global epidemiology of nonalcoholic fatty liver disease – meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 64, 7384.10.1002/hep.28431CrossRefGoogle ScholarPubMed
Kabarra, K, Golabi, P and Younossi, ZM (2021) Nonalcoholic steatohepatitis: global impact and clinical consequences. Endocrine Connections 10, R240RR47.10.1530/EC-21-0048CrossRefGoogle ScholarPubMed
Maak, S et al. (2021) Progress and challenges in the biology of FNDC5 and irisin. Endocrine Reviews 42, 436456.10.1210/endrev/bnab003CrossRefGoogle ScholarPubMed
Askari, H et al. (2018) A glance at the therapeutic potential of irisin against diseases involving inflammation, oxidative stress, and apoptosis: an introductory review. Pharmacological Research 129, 4455.CrossRefGoogle Scholar
Young, MF, Valaris, S and Wrann, CD (2019) A role for FNDC5/irisin in the beneficial effects of exercise on the brain and in neurodegenerative diseases. Progress in Cardiovascular Diseases 62, 172178.CrossRefGoogle ScholarPubMed
Schumacher, MA et al. (2013) The structure of irisin reveals a novel intersubunit beta-sheet fibronectin type III (FNIII) dimer: implications for receptor activation. Journal of Biological Chemistry 288, 3373833744.10.1074/jbc.M113.516641CrossRefGoogle ScholarPubMed
Raschke, S et al. (2013) Evidence against a beneficial effect of irisin in humans. PLoS ONE 8, e73680.10.1371/journal.pone.0073680CrossRefGoogle ScholarPubMed
Rabiee, F et al. (2020) New insights into the cellular activities of Fndc5/irisin and its signaling pathways. Cell & Bioscience 10, 51.10.1186/s13578-020-00413-3CrossRefGoogle ScholarPubMed
Kim, H et al. (2018) Irisin mediates effects on bone and Fat via alphaV integrin receptors. Cell 175, 17561768 e17.CrossRefGoogle Scholar
Estell, EG et al. (2020) Irisin directly stimulates osteoclastogenesis and bone resorption in vitro and in vivo. Elife 9, e58172.10.7554/eLife.58172CrossRefGoogle ScholarPubMed
Bi, J et al. (2020) Irisin reverses intestinal epithelial barrier dysfunction during intestinal injury via binding to the integrin alphaVbeta5 receptor. Journal of Cellular and Molecular Medicine 24, 9961009.10.1111/jcmm.14811CrossRefGoogle Scholar
Bi, J et al. (2020) Exercise hormone irisin mitigates endothelial barrier dysfunction and microvascular leakage-related diseases. JCI Insight 5, e136277.10.1172/jci.insight.136277CrossRefGoogle ScholarPubMed
Oguri, Y et al. (2020) CD81 controls beige fat progenitor cell growth and energy balance via FAK signaling. Cell 182, 563577. e20.10.1016/j.cell.2020.06.021CrossRefGoogle ScholarPubMed
Zhang, N et al. (2020) A novel biphenyl compound IMB-S7 ameliorates hepatic fibrosis in BDL rats by suppressing Sp1-mediated integrin alphav expression. Acta Pharmacologica Sinica 41, 661669.CrossRefGoogle ScholarPubMed
Sun, F et al. (2019) Interleukin-8 promotes integrin beta3 upregulation and cell invasion through PI3K/Akt pathway in hepatocellular carcinoma. Journal of Experimental & Clinical Cancer Research: CR 38, 449.10.1186/s13046-019-1455-xCrossRefGoogle ScholarPubMed
Younossi, Z et al. (2018) Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nature Reviews. Gastroenterology & Hepatology 15, 1120.CrossRefGoogle ScholarPubMed
Huang, DQ, El-Serag, HB and Loomba, R (2021) Global epidemiology of NAFLD-related HCC: trends, predictions, risk factors and prevention. Nature Reviews. Gastroenterology & Hepatology 18, 223238.10.1038/s41575-020-00381-6CrossRefGoogle ScholarPubMed
Hester, D et al. (2020) Among Medicare patients with hepatocellular carcinoma, non-alcoholic fatty liver disease is the most common etiology and cause of mortality. Journal of Clinical Gastroenterology 54, 459467.10.1097/MCG.0000000000001172CrossRefGoogle ScholarPubMed
Soleimani, D et al. (2021) Protective effects of propolis on hepatic steatosis and fibrosis among patients with nonalcoholic fatty liver disease (NAFLD) evaluated by real-time two-dimensional shear wave elastography: a randomized clinical trial. Phytotherapy Research 35, 16691679.10.1002/ptr.6937CrossRefGoogle ScholarPubMed
Soleimani, D et al. (2021) Effect of propolis supplementation on athletic performance, body composition, inflammation, and oxidative stress following intense exercise: a triple-blind randomized clinical trial. Food Science & Nutrition 9, 36313640.CrossRefGoogle ScholarPubMed
Chalasani, N et al. (2018) The diagnosis and management of nonalcoholic fatty liver disease: practice guidance from the American Association for the Study of Liver Diseases. Hepatology 67, 328357.10.1002/hep.29367CrossRefGoogle ScholarPubMed
Park, MJ et al. (2015) New role of irisin in hepatocytes: the protective effect of hepatic steatosis in vitro. Cellular Signalling 27, 18311839.10.1016/j.cellsig.2015.04.010CrossRefGoogle ScholarPubMed
Liu, TY et al. (2016) FNDC5 alleviates hepatosteatosis by restoring AMPK/mTOR-mediated autophagy, fatty acid oxidation, and lipogenesis in mice. Diabetes 65, 32623275.10.2337/db16-0356CrossRefGoogle ScholarPubMed
Mo, L et al. (2016) Irisin is regulated by CAR in liver and is a mediator of hepatic glucose and lipid metabolism. Molecular Endocrinology 30, 533542.CrossRefGoogle ScholarPubMed
So, WY and Leung, PS (2016) Irisin ameliorates hepatic glucose/lipid metabolism and enhances cell survival in insulin-resistant human HepG2 cells through adenosine monophosphate-activated protein kinase signaling. International Journal of Biochemistry & Cell Biology 78, 237247.CrossRefGoogle ScholarPubMed
Tang, H et al. (2016) Irisin inhibits hepatic cholesterol synthesis via AMPK-SREBP2 signaling. EBioMedicine 6, 139148.10.1016/j.ebiom.2016.02.041CrossRefGoogle ScholarPubMed
Motta, VF et al. (2017) Treating fructose-induced metabolic changes in mice with high-intensity interval training: insights in the liver, white adipose tissue, and skeletal muscle. Journal of Applied Physiology (1985) 123, 699709.10.1152/japplphysiol.00154.2017CrossRefGoogle ScholarPubMed
Petta, S et al. (2017) Fibronectin type III domain-containing protein 5 rs3480 A > G polymorphism, irisin, and liver fibrosis in patients with nonalcoholic fatty liver disease. Journal of Clinical Endocrinology and Metabolism 102, 26602669.10.1210/jc.2017-00056CrossRefGoogle Scholar
Canivet, CM et al. (2020) Hepatic FNDC5 is a potential local protective factor against non-alcoholic fatty liver. Biochimica et Biophysica Acta, Molecular Basis of Disease 1866, 165705.10.1016/j.bbadis.2020.165705CrossRefGoogle ScholarPubMed
Li, DJ et al. (2021) NAD(+)-boosting therapy alleviates nonalcoholic fatty liver disease via stimulating a novel exerkine Fndc5/irisin. Theranostics 11, 43814402.CrossRefGoogle ScholarPubMed
Zhu, W et al. (2021) Exercise-Induced irisin decreases inflammation and improves NAFLD by competitive binding with MD2. Cells 10, 3306.10.3390/cells10123306CrossRefGoogle ScholarPubMed
Medhat, D et al. (2021) Influence of irisin on diet-induced metabolic syndrome in experimental rat model. Journal of Complementary & Integrative Medicine 18, 347354.10.1515/jcim-2020-0030CrossRefGoogle ScholarPubMed
Kwon, J et al. (2021) DSS-induced colitis is associated with adipose tissue dysfunction and disrupted hepatic lipid metabolism leading to hepatosteatosis and dyslipidemia in mice. Scientific Reports 11, 5283.10.1038/s41598-021-84761-1CrossRefGoogle ScholarPubMed
Alamdari, AF et al. (2021) Melatonin as a promising modulator of aging related neurodegenerative disorders: role of microRNAs. Pharmacological Research 173, 105839.10.1016/j.phrs.2021.105839CrossRefGoogle ScholarPubMed
Gjorgjieva, M et al. (2019) miRNAs and NAFLD: from pathophysiology to therapy. Gut 68, 20652079.10.1136/gutjnl-2018-318146CrossRefGoogle ScholarPubMed
Yu, Y et al. (2022) MicroRNA-665-3p exacerbates nonalcoholic fatty liver disease in mice. Bioengineered 13, 29272942.CrossRefGoogle ScholarPubMed
Chen, J et al. (2022) Association of metabolic traits with occurrence of nonalcoholic fatty liver disease-related hepatocellular carcinoma: a systematic review and meta-analysis of longitudinal cohort studies. Saudi Journal of Gastroenterology 28, 92100.Google ScholarPubMed
Seo, DY et al. (2014) Effects of aged garlic extract and endurance exercise on skeletal muscle FNDC-5 and circulating irisin in high-fat-diet rat models. Nutrition Research and Practice 8, 177182.10.4162/nrp.2014.8.2.177CrossRefGoogle ScholarPubMed
Liu, TY et al. (2015) Irisin inhibits hepatic gluconeogenesis and increases glycogen synthesis via the PI3K/Akt pathway in type 2 diabetic mice and hepatocytes. Clinical Science 129, 839850.CrossRefGoogle ScholarPubMed
Niranjan, SB et al. (2019) Recombinant irisin induces weight loss in high fat DIO mice through increase in energy consumption and thermogenesis. Biochemical and Biophysical Research Communications 519, 422429.10.1016/j.bbrc.2019.08.112CrossRefGoogle ScholarPubMed
Canfora, EE et al. (2019) Gut microbial metabolites in obesity, NAFLD and T2DM. Nature Reviews. Endocrinology 15, 261273.10.1038/s41574-019-0156-zCrossRefGoogle ScholarPubMed
Kwon, J et al. (2020) Comprehensive amelioration of high-fat diet-induced metabolic dysfunctions through activation of the PGC-1alpha pathway by probiotics treatment in mice. PLoS ONE 15, e0228932.10.1371/journal.pone.0228932CrossRefGoogle ScholarPubMed
Chang, HC and Guarente, L (2014) SIRT1 and other sirtuins in metabolism. Trends in Endocrinology and Metabolism 25, 138145.10.1016/j.tem.2013.12.001CrossRefGoogle ScholarPubMed
Kheiripour, N et al. (2019) Hepatoprotective effects of silymarin on liver injury via irisin upregulation and oxidative stress reduction in rats with type 2 diabetes. Iranian Journal of Medical Sciences 44, 108117.Google ScholarPubMed
Shen, HH et al. (2019) Genistein ameliorated obesity accompanied with adipose tissue browning and attenuation of hepatic lipogenesis in ovariectomized rats with high-fat diet. Journal of Nutritional Biochemistry 67, 111122.CrossRefGoogle ScholarPubMed
Eser, N et al. (2021) Ameliorative effects of garlic oil on FNDC5 and irisin sensitivity in liver of streptozotocin-induced diabetic rats. Journal of Pharmacy and Pharmacology 73, 824834.CrossRefGoogle ScholarPubMed
Zhang, HJ et al. (2013) Irisin is inversely associated with intrahepatic triglyceride contents in obese adults. Journal of Hepatology 59, 557562.10.1016/j.jhep.2013.04.030CrossRefGoogle ScholarPubMed
Polyzos, SA et al. (2014) Irisin in patients with nonalcoholic fatty liver disease. Metabolism: Clinical and Experimental 63, 207217.10.1016/j.metabol.2013.09.013CrossRefGoogle ScholarPubMed
Shanaki, M et al. (2017) Lower circulating irisin is associated with nonalcoholic fatty liver disease and type 2 diabetes. Diabetes & Metabolic Syndrome 11(suppl. 1), S467SS72.CrossRefGoogle ScholarPubMed
Waluga, M et al. (2019) Omentin, vaspin and irisin in chronic liver diseases. Journal of Physiology and Pharmacology 70, 277285.Google ScholarPubMed
Choi, ES et al. (2014) Association between serum irisin levels and non-alcoholic fatty liver disease in health screen examinees. PLoS ONE 9, e110680.CrossRefGoogle ScholarPubMed
Rizk, FH, Elshweikh, SA and Abd El-Naby, AY (2016) Irisin levels in relation to metabolic and liver functions in Egyptian patients with metabolic syndrome. Canadian Journal of Physiology and Pharmacology 94, 359362.10.1139/cjpp-2015-0371CrossRefGoogle ScholarPubMed
Moreno-Perez, O et al. (2018) High irisin levels in nondiabetic HIV-infected males are associated with insulin resistance, nonalcoholic fatty liver disease, and subclinical atherosclerosis. Clinical Endocrinology 89, 414423.CrossRefGoogle ScholarPubMed
Bhanji, RA et al. (2017) Sarcopenia in hiding: the risk and consequence of underestimating muscle dysfunction in nonalcoholic steatohepatitis. Hepatology 66, 20552065.10.1002/hep.29420CrossRefGoogle ScholarPubMed
Armandi, A et al. (2022) Crosstalk between irisin levels, liver fibrogenesis and liver damage in non-obese, non-diabetic individuals with non-alcoholic fatty liver disease. Journal of Clinical Medicine 11, 635.CrossRefGoogle ScholarPubMed
Martinez-Montoro, JI et al. (2021) Impact of genetic polymorphism on response to therapy in non-alcoholic fatty liver disease. Nutrients 13, 4077.CrossRefGoogle ScholarPubMed
Metwally, M et al. (2019) A polymorphism in the irisin-encoding gene (FNDC5) associates with hepatic steatosis by differential miRNA binding to the 3'UTR. Journal of Hepatology 70, 494500.CrossRefGoogle Scholar
Daneshi-Maskooni, M et al. (2019) Green cardamom supplementation improves serum irisin, glucose indices, and lipid profiles in overweight or obese non-alcoholic fatty liver disease patients: a double-blind randomized placebo-controlled clinical trial. BMC Complementary and Alternative Medicine 19, 59.10.1186/s12906-019-2465-0CrossRefGoogle ScholarPubMed
Rahmanabadi, A et al. (2019) Oral alpha-lipoic acid supplementation in patients with non-alcoholic fatty liver disease: effects on adipokines and liver histology features. Food & Function 10, 49414952.CrossRefGoogle ScholarPubMed
Trautwein, C et al. (2015) Hepatic fibrosis: concept to treatment. Journal of Hepatology 62(suppl. 1), S15S24.10.1016/j.jhep.2015.02.039CrossRefGoogle ScholarPubMed
Ellis, EL and Mann, DA (2012) Clinical evidence for the regression of liver fibrosis. Journal of Hepatology 56, 11711180.10.1016/j.jhep.2011.09.024CrossRefGoogle ScholarPubMed
Elpek, GO (2014) Cellular and molecular mechanisms in the pathogenesis of liver fibrosis: an update. World Journal of Gastroenterology 20, 72607276.CrossRefGoogle ScholarPubMed
Zhou, B et al. (2018) Fibronectin type III domain-containing 5 attenuates liver fibrosis via inhibition of hepatic stellate cell activation. Cellular Physiology and Biochemistry 48, 227236.10.1159/000491722CrossRefGoogle ScholarPubMed
Dong, HN et al. (2020) Irisin regulates the functions of hepatic stellate cells. Endocrinology and Metabolism 35, 647655.10.3803/EnM.2020.658CrossRefGoogle ScholarPubMed
Liao, X et al. (2021) Irisin ameliorates endoplasmic reticulum stress and liver fibrosis through inhibiting PERK-mediated destabilization of HNRNPA1 in hepatic stellate cells. Biological Chemistry 402, 703715.10.1515/hsz-2020-0251CrossRefGoogle ScholarPubMed
Pazgan-Simon, M et al. (2020) Serum concentrations of selected adipokines in virus-related liver cirrhosis and hepatocellular carcinoma. Clinical and Experimental Hepatology 6, 235242.10.5114/ceh.2020.99517CrossRefGoogle ScholarPubMed
Kukla, M et al. (2020) Irisin in liver cirrhosis. Journal of Clinical Medicine 9, 3158.CrossRefGoogle ScholarPubMed
Zhao, M et al. (2020) Association between serum irisin concentrations and sarcopenia in patients with liver cirrhosis: a cross-sectional study. Scientific Reports 10, 16093.10.1038/s41598-020-73176-zCrossRefGoogle ScholarPubMed
Hou, L et al. (2021) A sex-stratified prognostic nomogram incorporating body compositions for long-term mortality in cirrhosis. JPEN. Journal of Parenteral and Enteral Nutrition 45, 403413.CrossRefGoogle ScholarPubMed
Wang, Y et al. (2019) Genipin ameliorates carbon tetrachloride-induced liver injury in mice via the concomitant inhibition of inflammation and induction of autophagy. Oxidative Medicine and Cellular Longevity 2019, 3729051.10.1155/2019/3729051CrossRefGoogle Scholar
Barbagelata, A et al. (2007) Time to reperfusion in acute myocardial infarction. It is time to reduce it!. Journal of Electrocardiology 40, 257264.10.1016/j.jelectrocard.2007.01.007CrossRefGoogle ScholarPubMed
Thirupurasundari, CJ, Jayalakshmi, R and Niranjali Devaraj, S (2005) Liver architecture maintenance by tincture of Crataegus against isoproterenol-induced myocardially infarcted rats. Journal of Medicinal Food 8, 400404.CrossRefGoogle ScholarPubMed
Kuloglu, T et al. (2014) Irisin: a potentially candidate marker for myocardial infarction. Peptides 55, 8591.CrossRefGoogle ScholarPubMed
Aydin, S et al. (2017) The effect of iloprost and sildenafil, alone and in combination, on myocardial ischaemia and nitric oxide and irisin levels. Cardiovascular Journal of Africa 28, 389396.10.5830/CVJA-2017-025CrossRefGoogle ScholarPubMed
Aydin, S et al. (2015) Effect of carnosine supplementation on apoptosis and irisin, total oxidant and antioxidants levels in the serum, liver and lung tissues in rats exposed to formaldehyde inhalation. Peptides 64, 1423.CrossRefGoogle ScholarPubMed
Erdogan, MA and Yalcin, A (2020) Protective effects of benfotiamine on irisin activity in methotrexate-induced liver injury in rats. Archives of Medical Science: AMS 16, 205211.10.5114/aoms.2018.80002CrossRefGoogle ScholarPubMed
Unsal, V, Cicek, M and Sabancilar, I (2021) Toxicity of carbon tetrachloride, free radicals and role of antioxidants. Reviews on Environmental Health 36, 279295.10.1515/reveh-2020-0048CrossRefGoogle ScholarPubMed
Zhao, TM et al. (2020) Bicyclol attenuates acute liver injury by activating autophagy, anti-oxidative and anti-inflammatory capabilities in mice. Frontiers in Pharmacology 11, 463.10.3389/fphar.2020.00463CrossRefGoogle ScholarPubMed
Rabey, E et al. , HA (2021) Green coffee methanolic extract and silymarin protect against CCl4-induced hepatotoxicity in albino male rats. BMC Complementary Medicine and Therapies 21, 19.CrossRefGoogle ScholarPubMed
Pahlavani, N et al. (2020) Molecular and cellular mechanisms of the effects of propolis in inflammation, oxidative stress and glycemic control in chronic diseases. Nutrition and Metabolism 17, 65.10.1186/s12986-020-00485-5CrossRefGoogle ScholarPubMed
Lu, L et al. (2016) Innate immune regulations and liver ischemia-reperfusion injury. Transplantation 100, 26012610.10.1097/TP.0000000000001411CrossRefGoogle ScholarPubMed
Bi, J et al. (2019) Irisin alleviates liver ischemia-reperfusion injury by inhibiting excessive mitochondrial fission, promoting mitochondrial biogenesis and decreasing oxidative stress. Redox Biology 20, 296306.10.1016/j.redox.2018.10.019CrossRefGoogle ScholarPubMed
Youle, RJ and van der Bliek, AM (2012) Mitochondrial fission, fusion, and stress. Science 337, 10621065.CrossRefGoogle Scholar
Zhang, J et al. (2020) Involvement of kindlin-2 in irisin's protection against ischaemia reperfusion-induced liver injury in high-fat diet-fed mice. Journal of Cellular and Molecular Medicine 24, 1308113092.10.1111/jcmm.15910CrossRefGoogle ScholarPubMed
Jing, HR et al. (2018) Fish oil alleviates liver injury induced by intestinal ischemia/reperfusion via AMPK/SIRT-1/autophagy pathway. World Journal of Gastroenterology 24, 833843.10.3748/wjg.v24.i7.833CrossRefGoogle ScholarPubMed
Saidi, SA et al. (2017) Liver injury following small intestinal ischemia reperfusion in rats is attenuated by Pistacia lentiscus oil: antioxidant and anti-inflammatory effects. Archives of Physiology and Biochemistry 123, 199205.CrossRefGoogle ScholarPubMed
Fan, X et al. (2019) Irisin contributes to the hepatoprotection of dexmedetomidine during intestinal ischemia/reperfusion. Oxidative Medicine and Cellular Longevity 2019, 7857082.10.1155/2019/7857082CrossRefGoogle Scholar
Bi, J et al. (2020) Irisin improves autophagy of aged hepatocytes via increasing telomerase activity in liver injury. Oxidative Medicine and Cellular Longevity 2020, 6946037.CrossRefGoogle ScholarPubMed
Cheng, Z et al. (2019) Circulating histones are major mediators of multiple organ dysfunction syndrome in acute critical illnesses. Critical Care Medicine 47, e677ee84.CrossRefGoogle ScholarPubMed
Tan, C et al. (2020) Early systemic inflammatory response syndrome duration predicts infected pancreatic necrosis. Journal of Gastrointestinal Surgery 24, 590597.CrossRefGoogle ScholarPubMed
Ren, YF et al. (2019) Irisin attenuates intestinal injury, oxidative and endoplasmic reticulum stress in mice with L-arginine-induced acute pancreatitis. World Journal of Gastroenterology 25, 66536667.CrossRefGoogle ScholarPubMed
Rhee, C et al. (2017) Incidence and trends of sepsis in US hospitals using clinical vs claims data, 2009–2014. JAMA 318, 12411249.10.1001/jama.2017.13836CrossRefGoogle Scholar
Kobashi, H, Toshimori, J and Yamamoto, K (2013) Sepsis-associated liver injury: incidence, classification and the clinical significance. Hepatology Research 43, 255266.CrossRefGoogle ScholarPubMed
Mao, L et al. (2020) The emerging role of ferroptosis in non-cancer liver diseases: hype or increasing hope? Cell Death & Disease 11, 518.10.1038/s41419-020-2732-5CrossRefGoogle ScholarPubMed
Knorr, J, Wree, A and Feldstein, AE (2021) Pyroptosis in steatohepatitis and liver diseases. Journal of Molecular Biology 434, 167271.10.1016/j.jmb.2021.167271CrossRefGoogle ScholarPubMed
Wei, S et al. (2020) Serum irisin levels are decreased in patients with sepsis, and exogenous irisin suppresses ferroptosis in the liver of septic mice. Clinical and Translational Medicine 10, e173.10.1002/ctm2.173CrossRefGoogle ScholarPubMed
Li, Q et al. (2021) Irisin alleviates LPS-induced liver injury and inflammation through inhibition of NLRP3 inflammasome and NF-kappaB signaling. Journal of Receptor and Signal Transduction Research 41, 294303.CrossRefGoogle ScholarPubMed
Erstad, DJ and Tanabe, KK (2017) Hepatocellular carcinoma: early-stage management challenges. The Journal of Hepatocellular Carcinoma 4, 8192.10.2147/JHC.S107370CrossRefGoogle ScholarPubMed
Gowhari Shabgah, A et al. (2021) Shedding more light on the role of Midkine in hepatocellular carcinoma: new perspectives on diagnosis and therapy. IUBMB Life 73, 659669.CrossRefGoogle ScholarPubMed
Yang, S et al. (2021) Role of forkhead box O proteins in hepatocellular carcinoma biology and progression (review). Frontiers in Oncology 11, 667730.CrossRefGoogle Scholar
Wen, Y et al. (2016) Role of osteopontin in liver diseases. International Journal of Biological Sciences 12, 11211128.CrossRefGoogle ScholarPubMed
Shi, G et al. (2017) Irisin stimulates cell proliferation and invasion by targeting the PI3K/AKT pathway in human hepatocellular carcinoma. Biochemical and Biophysical Research Communications 493, 585591.CrossRefGoogle ScholarPubMed
Sun, EJ et al. (2021) Targeting the PI3K/Akt/mTOR pathway in hepatocellular carcinoma. Biomedicines 9, 1639.CrossRefGoogle ScholarPubMed
Pathria, P, Louis, TL and Varner, JA (2019) Targeting tumor-associated macrophages in cancer. Trends in Immunology 40, 310327.10.1016/j.it.2019.02.003CrossRefGoogle ScholarPubMed
Arvanitakis, K et al. (2022) Tumor-associated macrophages in hepatocellular carcinoma pathogenesis, prognosis and therapy. Cancers 14, 226.CrossRefGoogle Scholar
Liu, H et al. (2021) FNDC5 induces M2 macrophage polarization and promotes hepatocellular carcinoma cell growth by affecting the PPARgamma/NF-kappaB/NLRP3 pathway. Biochemical and Biophysical Research Communications 582, 7785.CrossRefGoogle ScholarPubMed
Aydin, S et al. (2016) Irisin immunohistochemistry in gastrointestinal system cancers. Biotechnic and Histochemistry 91, 242250.10.3109/10520295.2015.1136988CrossRefGoogle ScholarPubMed
Gaggini, M et al. (2017) Increased FNDC5/irisin expression in human hepatocellular carcinoma. Peptides 88, 6266.CrossRefGoogle ScholarPubMed
Varela-Rodriguez, BM et al. (2016) FNDC5 Expression and circulating irisin levels are modified by diet and hormonal conditions in hypothalamus, adipose tissue and muscle. Scientific Reports 6, 29898.CrossRefGoogle Scholar
Zhang, J et al. (2019) Serum irisin predicts posthepatectomy complications in patients with hepatocellular carcinoma. Disease Markers 2019, 9850191.CrossRefGoogle ScholarPubMed
Pazgan-Simon, M et al. (2020) Serum betatrophin and irisin levels in hepatocellular carcinoma. Journal of Physiology and Pharmacology 71, 113123.Google ScholarPubMed
Kim, SH et al. (2019) Serum biomarkers for predicting overall survival and early mortality in older patients with metastatic solid tumors. Journal of Geriatric Oncology 10, 749756.CrossRefGoogle ScholarPubMed
Williamson, C and Geenes, V (2014) Intrahepatic cholestasis of pregnancy. Obstetrics & Gynecology 124, 120133.CrossRefGoogle ScholarPubMed
Geenes, V and Williamson, C (2009) Intrahepatic cholestasis of pregnancy. World Journal of Gastroenterology 15, 20492066.CrossRefGoogle ScholarPubMed
Menzyk, T et al. (2018) The role of metabolic disorders in the pathogenesis of intrahepatic cholestasis of pregnancy. Clinical and Experimental Hepatology 4, 217223.CrossRefGoogle ScholarPubMed
Kirbas, A et al. (2016) Maternal circulating levels of irisin in intrahepatic cholestasis of pregnancy. The Journal of Maternal-Fetal & Neonatal Medicine 29, 34833487.Google ScholarPubMed
Chen, J, Li, Q and Ma, J (2021) Maternal serum, placental, and umbilical venous blood irisin levels in intrahepatic cholestasis of pregnancy. The Journal of Maternal-Fetal & Neonatal Medicine 34, 24032410.CrossRefGoogle ScholarPubMed
Cao, RY et al. (2019) FNDC5: a novel player in metabolism and metabolic syndrome. Biochimie 158, 111116.CrossRefGoogle ScholarPubMed
Zhao, J et al. (2022) Antioxidant effects of irisin in liver diseases: mechanistic insights. Oxidative Medicine and Cellular Longevity 2022, 3563518.10.1155/2022/3563518CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Schematic diagram depicting the pathogenic role and therapeutic potential of FNDC5/irisin in experimental NAFLD models. FNDC5, fibronectin type III domain-containing protein 5; HFD, high-fat diet; MCD, methionine- and choline-deficient; SIRT, sirtuin; UCP1, uncoupling protein 1; PA, palmitic acid; AMPK, adenosine monophosphate-activated protein kinase; mTORC1, mammalian target of rapamycin complex 1; PPARα, peroxisome proliferator-activated receptor α; TLR4, Toll-like receptor 4; MD2, myeloid differentiation factor 2; PGC1α, proliferator-activated receptor-γ co-activator 1α; NAD+, nicotinamide adenine dinucleotide+; MyD88, myeloid differentiation factor 88; MAPK, mitogen-activated protein kinase.

Figure 1

Table 1. The pathogenic role and therapeutic potential of FNDC5/irisin in experimental NAFLD models

Figure 2

Fig. 2. Schematic diagram depicting the pathogenic role and therapeutic potential of FNDC5/irisin in experimental fibrosis models. FNDC5, fibronectin type III domain-containing protein 5; HSC, hepatic stellate cell; ECM, extracellular matrix; AMPK, adenosine monophosphate-activated protein kinase; α-SMA, α-smooth muscle actin; TGF-β, transforming growth factor-β; IL-, interleukin-; HFD, high-fat diet; CCl4, carbon tetrachloride; LPS, lipopolysaccharide; PERK, PKR-like ER kinase; HNRNPA1, heterogeneous nuclear ribonucleoprotein A1; oxLDL, oxidised low density lipoprotein.