Next Article in Journal
Peripheral Cytokine Levels as a Prognostic Indicator in Gastric Cancer: A Review of Existing Literature
Next Article in Special Issue
Metabolic Fatty Liver Disease in Children: A Growing Public Health Problem
Previous Article in Journal
Role of Phytoconstituents as PPAR Agonists: Implications for Neurodegenerative Disorders
Previous Article in Special Issue
Diet and Gut Microbiota Interaction-Derived Metabolites and Intrahepatic Immune Response in NAFLD Development and Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hepatokines and Non-Alcoholic Fatty Liver Disease: Linking Liver Pathophysiology to Metabolism

1
Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women’s University, Seoul 04310, Korea
2
Department of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
3
KNU Researcher Training Program for Developing Anti-Viral Innovative Drugs, Kangwon National University, Chuncheon 24341, Korea
*
Author to whom correspondence should be addressed.
Biomedicines 2021, 9(12), 1903; https://doi.org/10.3390/biomedicines9121903
Submission received: 3 December 2021 / Revised: 12 December 2021 / Accepted: 12 December 2021 / Published: 14 December 2021
(This article belongs to the Special Issue NAFLD: From Mechanisms to Therapeutic Approaches)

Abstract

:
The liver plays a key role in maintaining energy homeostasis by sensing and responding to changes in nutrient status under various metabolic conditions. Recently highlighted as a major endocrine organ, the contribution of the liver to systemic glucose and lipid metabolism is primarily attributed to signaling crosstalk between multiple organs via hepatic hormones, cytokines, and hepatokines. Hepatokines are hormone-like proteins secreted by hepatocytes, and a number of these have been associated with extra-hepatic metabolic regulation. Mounting evidence has revealed that the secretory profiles of hepatokines are significantly altered in non-alcoholic fatty liver disease (NAFLD), the most common hepatic manifestation, which frequently precedes other metabolic disorders, including insulin resistance and type 2 diabetes. Therefore, deciphering the mechanism of hepatokine-mediated inter-organ communication is essential for understanding the complex metabolic network between tissues, as well as for the identification of novel diagnostic and/or therapeutic targets in metabolic disease. In this review, we describe the hepatokine-driven inter-organ crosstalk in the context of liver pathophysiology, with a particular focus on NAFLD progression. Moreover, we summarize key hepatokines and their molecular mechanisms of metabolic control in non-hepatic tissues, discussing their potential as novel biomarkers and therapeutic targets in the treatment of metabolic diseases.

1. Introduction

The global prevalence of obesity has been increasing over several decades, reaching epidemic levels, and thus, raising serious public health concerns [1]. Obesity greatly increases the risk of metabolic syndrome, including insulin resistance, type 2 diabetes, fatty liver disease, cardiovascular disease, and neurodegenerative conditions, thereby significantly contributing to greater morbidity and mortality [1]. Moreover, obesity-induced chronic inflammation, particularly in the liver, adipose tissue, and skeletal muscle, plays a crucial role in the development of local and systemic insulin resistance, simultaneously inducing a range of metabolic disturbances in multiple organs via inter-organ crosstalk [2,3]. Given the concurrent detrimental influence of obesity on various organs, obesity-driven metabolic disorders are largely attributed to dysregulated multi-directional interactions between organ metabolism. In other words, inter-organ communication via autocrine, paracrine, and endocrine signals regulates systemic energy homeostasis, which can be disturbed by a disequilibrium between energy intake and expenditure (e.g., obesity, hepatic steatosis, etc.).
Non-alcoholic fatty liver disease (NAFLD) refers to the spectrum of chronic liver disease progression in the absence of excessive alcohol consumption, ranging from simple hepatic steatosis to severe pathological conditions, including non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma [4,5,6,7]. Hepatic steatosis is characterized by the accumulation of ectopic lipids in hepatocytes by more than 5% of total liver mass, exhibiting mostly benign and/or mild clinical symptoms. However, this becomes problematic after developing NASH with the signs of hepatocellular damage, inflammation and fibrotic changes, favoring its progression towards more debilitating conditions. In parallel, emerging evidence has also revealed that patients with NAFLD have greater susceptibility to various infectious diseases, including bacterial pneumonia, Helicobacter pylori infection, urinary tract infection, Clostridium difficile colitis, and coronavirus disease 2019 (COVID-19) in close association with low-grade chronic inflammation, impaired innate immunity, and/or vitamin D deficiency [8,9]. NAFLD serves as an early hepatic manifestation that is primarily responsible for the progression of a variety of metabolic disorders [4,5,6,7], implying the multifactorial features of NAFLD and its close link with other comorbidities. Given the heterogenous pathogenesis of NAFLD and its increasing prevalence, leading international societies of hepatology have recently proposed new nomenclature ‘metabolic dysfunction-associated fatty liver disease’ (MAFLD) in the replacement of NAFLD, emphasizing more on its function in metabolic dysregulation, in order to better reflect the current knowledge, as well as its diverse etiologies of metabolic liver disease [10,11].
Recent advances in comprehensive genetic, transcriptomic, and proteomic technologies have provided insights into the role of the liver as an endocrine organ central to metabolism. Moreover, growing evidence has revealed that the liver mediates metabolic regulation through the release of various secretory factors, including hepatokines [12,13,14,15]. Hepatokines are hormone-like proteins primarily secreted by hepatocytes, with the hepatokine secretory profile known to be markedly disturbed in NAFLD. In fact, NAFLD frequently precedes dysfunction in other organs during the pathogenesis of systemic metabolic diseases. Therefore, it is highly likely that altered hepatokine secretion at NAFLD onset will significantly impair the inter-organ signaling crosstalk, which may trigger the progression of complex and multifaceted metabolic dysregulation. In this review, we first outline distinctive roles of the liver as a hepatokine-producing organ under normal physiology. Then, we discuss alterations in hepatokine secretion during NAFLD development and their impact on metabolic disease. The current review also summarizes recent discoveries regarding the molecular mechanisms and effects of key hepatokines, in addition to their potential as novel diagnostic and therapeutic targets in metabolic disease treatment.

2. Role of the Liver in Metabolism under Normal Physiology

The liver contributes to the maintenance of systemic metabolism by controlling complex pathways that coordinate nutrient intake and energy expenditure. Blood flows into the liver from the heart through the hepatic artery (~25% of total blood volume) or from the gastrointestinal tract via the portal vein (~75%), with most of the nutrients (e.g., glucose and lipids) absorbed in the intestines which are delivered prior to their entry into systemic circulation. These anatomical and structural features enable the liver to sense and promptly respond to changes in nutrient availability [16,17].
The liver serves as a key regulator of glucose metabolism by orchestrating hepatic glucose production and glycogen storage. In a postprandial state, hepatic glucose uptake is upregulated in response to elevated plasma glucose and insulin levels. Then, the absorbed glucose is stored in the liver as glycogen or is utilized for fatty acid synthesis (i.e., de novo lipogenesis). Under the influence of insulin secreted in response to elevated blood glucose, hepatic glucose production and glycogenolysis are suppressed in order to normalize blood glucose concentrations [13,14]. During the fasting state, the liver upregulates blood glucose levels by stimulating hepatic glucose production and glycogen breakdown via transcriptional and non-transcriptional mechanisms [13,18,19], thus increasing the supply of glucose as a major energy source to non-hepatic peripheral tissues (e.g., brain, adipose tissue, skeletal muscle). The liver also plays a key role in lipid metabolism in response to nutrient availability, as well as insulin. Following a meal, insulin facilitates hepatic fatty acid uptake, the synthesis and storage of triglycerides (TG) via the utilization of dietary fatty acids, and promotes de novo lipogenesis in the liver [20], favoring long-term lipid storage. However, under fasting, wherein glucose availability is low, the liver carries out lipid oxidation and acutely produces ketone bodies, which serve as an alternative fuel source for non-hepatic tissues in order to meet energy demands [20,21]. As the liver encounters repetitive fasting-feeding transitions throughout the life cycle of living organisms, the hepatic regulation of metabolism, including energy production, expenditure, storage, and redistribution, is critical for maintaining systemic energy homeostasis under normal physiology.

3. Overview of Hepatokines

The liver regulates physiology and metabolism via the production and secretion of various plasma proteins, including albumin, coagulation factors, complement factors, transport proteins, and other factors [14]. Although the liver has long been recognized as a secretory organ, recent advances in mass spectrometry-based quantitative proteomics have enabled researchers to identify ~10,200 proteins produced in the human liver [22], with up to 40% of hepatic transcripts encoding secretory proteins [23]. Similarly, quantitative analysis of the mouse liver and plasma proteome identified 7099 and 4727 proteins, respectively, with ~25% of these overlapping with plasma proteins, suggestive of their secretion [12,24]. As mentioned above, the liver is directly connected to systemic circulation, receiving a substantial amount of blood from the heart and gastrointestinal tract, exchanging nutrients and other substances within the sinusoids, and then draining blood via the hepatic vein and inferior vena cava towards the heart for recirculation throughout the body [14]. Considering its secretory capacity as an endocrine organ, it is plausible to expect that the liver plays a fundamental role in inter-organ crosstalk through the release of secretory proteins, including hepatokines.
Analogous to adipose tissue-derived adipokines and skeletal muscle-derived myokines, hepatokines are a class of organokines, meaning a group of secretory proteins that are exclusively produced by the parenchymal cell type of respective tissue. The significance of hepatokines in the regulation of various biological processes in autocrine, paracrine, and endocrine fashion has been recently highlighted [13,25]. Given the functional features of hepatocytes, which constitute ~80% of the volume and ~70% of the total cell number within the whole liver, several studies have shown that normal mouse hepatocytes release more than 500 secretory proteins, which may or may not contain an N-terminal secretory peptide [12,26]. Since emerging evidence has demonstrated that factors secreted from hepatocytes actively mediate metabolic regulation between the liver and other organs, hepatokines have drawn increasing attention due to their capacity for metabolic regulation, making them novel targets for the modulation of energy homeostasis and the treatment of metabolic disorders [12,13,14,15]. In this review, we summarized target organs or cells of important hepatokines and their biological functions (Table 1).

4. Hepatokines and NAFLD

It has been established that NAFLD is strongly associated with other metabolic comorbidities, including obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, colonic diverticulosis, and neurodegenerative conditions [1,4,5,6,7,14,91]. Hepatic steatosis, which refers to a pathological state of the liver characterized by an accumulation of lipid content at over ~5% of the total organ weight, is closely associated with insulin resistance in multiple organs, as supported by several studies demonstrating impaired insulin action in both lean and non-diabetic obese individuals [14,92,93,94]. Moreover, hepatic steatosis usually develops prior to the accumulation of lipid in skeletal muscle, macrophage-driven inflammation, extrahepatic insulin resistance, and hyperglycemia [14,16,95], suggestive of its potential as an early indicator of systemic metabolic dysregulation. Interestingly, similar to adipose tissue or skeletal muscle where the secretion of respective organokines is altered via overnutrition, hepatic gene expression and protein content are also regulated in response to caloric overload, and these changes have been revealed to be strongly associated with the onset of insulin resistance and type 2 diabetes [26,96,97,98]. Many of the liver-secreted proteins that are upregulated in the plasma of subjects with type 2 diabetes were capable of inducing insulin resistance, supportive of the pathophysiological role of hepatokines in metabolic dysregulation [99]. Recent studies have provided valuable insight into the impact of ectopic fat accumulation in hepatocytes on alterations in the hepatic proteome, with suppressed protein synthesis observed in the livers of obese mice [100]. Furthermore, metabolic remodeling was mediated via changes in the translation/secretion processes or post-translational modifications, with minor changes at the transcriptional level [12,26]. In line with this notion, approximately 20% of proteins with an N-terminal signal peptide were differentially secreted from mouse steatotic hepatocytes compared to normal hepatocytes, and some of these proteins were found to induce insulin resistance and pro-inflammatory signaling [26]. The above-described findings indicate that a substantial fraction of the hepatic proteome is heavily distributed during NAFLD progression, which may significantly impair energy homeostasis and systemic metabolism. However, relatively a small number of hepatokines have been identified, with a few of their metabolic functions and related regulatory mechanism(s) in NAFLD progression investigated. Considering the close association between NAFLD and hepatokines in the context of metabolic disease progression, we have summarized recent findings on the function of some key hepatokines in the regulation of metabolism via organ-to-organ crosstalk (Table 1).

4.1. Angiopoietin-Like Proteins (ANGPTLs)

ANGPTLs are a family of glycoproteins primarily secreted from the liver, and all of these share an N-terminal coiled-coil domain and a C-terminal fibrinogen-like domain, except for ANGPTL8, which lacks the latter [40,44,101]. To date, eight ANGPTLs have been identified (ANGPTL1–ANGPTL8), all of which are capable of regulating angiogenesis as angiopoietins [15,27,40,101,102,103,104]. Despite their structural similarity, ANGPTLs were originally regarded as orphan ligands since they do not bind to classical angiopoietin cognate receptors, including tyrosine kinase with immunoglobulin-like and EGF-like domain 1 (Tie1) and TEK receptor tyrosine kinase (Tie2) [101,105,106]. Thereafter, human leukocyte immunoglobulin-like receptor B2 and its mouse orthologue paired immunoglobulin-like receptor were defined as the receptors for ANGPTL1, 2, 5, and 7 [107]. Angiopoietin-like proteins play an important role in lipid metabolism, with ANGPTL3, 4, 6, and 8 involved in lipoprotein metabolism and modulating plasma lipid levels via the regulation of lipoprotein lipase (LPL) and endothelial lipase-dependent TG hydrolysis (Figure 1) [15,27,40,101].
ANGPTL3 is predominantly synthesized and secreted by the liver, while ANGPTL4 and ANGPTL8 are also expressed in other tissues, such as adipose tissue [28]. Activated after feeding, ANGPTL3 inhibits LPL through direct binding via the N-terminal coiled-coil domain, subsequently dissociating active LPL dimers into inactive monomers with the help of ANGPTL8 [15,27,28,29,30], which leads to the increased storage of lipoprotein-derived fatty acids in white adipose tissue [31]. In addition, ANGPTL3 inhibition decreases plasma TG and free fatty acid levels in mice [28], while also reducing VLDL size and lipid content in an endothelial lipase-dependent manner [15,32]. Interestingly, fasting conditions as well as very low-calorie intake markedly decreased serum ANGPTL3 in obese individuals, while ANGPTL4 showed the opposite or a distinct trend [108]. Furthermore, the circulating levels of both may be independent risk factors and potential predictors of coronary atherosclerosis [109]. Recent studies have provided insight into the metabolic role of circulating ANGPTL3 in preclinical models, wherein the treatment with an anti-sense oligonucleotide targeting Angptl3 mRNA in low-density lipoprotein receptor knockout mice resulted in a dramatic reduction of liver TG content, as well as atherosclerosis progression [110]. The ob/ob mice, vaccinated with a peptide targeting ANGPTL3, also exhibited a marked decrease in diet-induced obesity and hepatic steatosis [111], suggestive of the metabolic impact of ANGPTL3 on NAFLD. In addition, the treatment with evinacumab alone or in combination with other lipid-lowering medications, such as statins and PCSK9 inhibitors, showed promising results for ameliorating hyperlipidemia in comparison to general lipid-lowering drugs alone in a phase 3 clinical trial, as well as in rodent models [112,113,114,115]. These findings indicate that hepatokine ANGPTL3 acts as an important regulator of lipoprotein metabolism and has great potential for the treatment of atherosclerosis [28].
ANGPTL4 is the most extensively studied member of the ANGPTL family and is primarily produced from several organs, including adipose tissue and the liver [116,117]. In humans, ANGPTL4 is predominantly expressed in the liver, followed by adipose tissue, while in mice the highest levels of ANGPTL4 are observed in white and brown adipose tissue [118]. ANGPTL4 was originally referred to as fasting-induced adipose factor, with its expression increasing in multiple tissues under fasting or hypoxia [34]. Similarly, Angptl4 transcript levels in the liver are upregulated upon fasting via peroxisome proliferator-activated receptor α (PPARα) signaling and are suppressed after refeeding [15,119]. ANGPTL4 plays a crucial role in various biological and pathophysiological processes, including the maintenance of energy homeostasis, TG metabolism, wound repair, angiogenesis, tumorigenesis, etc. [15,27,34,104,120]. The full-length ANGPTL4 undergoes proteolytic cleavage at the linker region into its N-terminal and C-terminal portions, which exhibit distinct biological functions [34]. Emerging evidence has demonstrated that both full-length ANGPTL4 and N-terminal ANGPTL4 upregulate plasma TG levels via the inhibition of LPL activity and the suppression of TG-containing lipoprotein clearance from plasma [15,34,35,36,37]. Meanwhile, C-terminal ANGPTL4 not only increases adipocyte lipolysis [38], but also regulates several processes beyond lipid metabolism, including the metabolic flexibility of metastatic cancer cells, wound healing in the skin, and pulmonary inflammation [121,122,123]. Given the therapeutic potential of ANGPTL4 in metabolic disorders, a number of studies have sought to validate the functional role of ANGPTL4 in energy metabolism using preclinical animal models, with global ANGPTL4 depletion via neutralizing monoclonal antibodies or germline Angptl4 knockout mice and monkeys exhibiting severe metabolic abnormalities under high-fat diet (HFD) feeding [124,125,126]. In line with these findings, there have been conflicting observations regarding the levels of circulating ANGPTL4 in humans, with several studies reporting that ANGPTL4 plasma levels were elevated in patients with type 2 diabetes and obese non-diabetic subjects [108,127,128,129,130], while the concentration of ANGPTL4 was lower in another cohort of type 2 diabetes patients and showed an inverse correlation with blood glucose levels and the insulin resistance index [39]. These discrepancies highlight the necessity for a more fundamental approach towards understanding the regulatory mechanism of ANGPTL4 in biological and metabolic processes. Interestingly, several studies have found that ANGPTL4 distinctively regulates lipid and glucose metabolism in a tissue-dependent manner under various pathophysiological conditions. Adenovirus-driven Angptl4 overexpression in mice improved systemic glucose tolerance and enhanced the insulin-induced suppression hepatic glucose production. However, mice developed hepatic steatosis and hyperlipidemia, including elevated plasma concentration of TGs, free fatty acids, total, and HDL cholesterol [39]. Transgenic mice with adipose tissue-specific Angptl4 overexpression exhibited glucose intolerance and impaired insulin sensitivity in skeletal muscle, as well as aggravated hepatic steatosis when subjected to long-term HFD feeding [131]. Recent studies revealed that the genetic ablation of Angptl4 in liver or adipose tissue, as well as targeted pharmacological inhibition of ANGPTL4 in hepatocytes significantly reduced circulating TG and cholesterol levels in mice, protecting them against diet-induced obesity, glucose intolerance, liver steatosis, and atherogenesis [124,132]. Taken together, all these findings strongly suggest a complex molecular mechanism of ANGPTL4 function in vivo, highlighting the need for further investigation into the tissue-dependent metabolic roles of ANGPTL4.
ANGPTL6, also referred to as angiopoietin-related growth factor, is another circulating protein predominantly secreted from the liver into the blood, with a key role in the maintenance of glucose and lipid metabolism [133,134]. Mice with genetic ablation of Angptl6 exhibited greater obesity phenotypes, with increased hepatic fat accumulation and insulin resistance, in conjunction with impaired energy utilization [133], while Angptl6 overexpression improved hepatic steatosis and obesity under increased energy expenditure [133]. In addition, several studies have demonstrated that the adenosine monophosphate-activated kinase (AMPK) pathway is activated by ANGPTL6, in parallel to enhanced insulin signaling in skeletal muscle [40]. Furthermore, ANGPTL6 inhibited hepatic gluconeogenesis by suppressing FOXO1 activity downstream of PI3K-Akt [41,42]. The treatment with recombinant ANGPTL6 increased mitochondrial oxygen consumption and PPARα expression via extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase signaling in cultured adipocytes [43]. Although there is limited information on the pathophysiological significance of ANGPTL6 up-/downregulation in humans under different metabolic statuses, emerging evidence has revealed that serum ANGPTL6 levels were increased in individuals suffering from type 2 diabetes, metabolic syndrome, and preeclampsia [135,136,137,138]. In particular, ANGPTL6 levels were upregulated in patients with obesity and type 2 diabetes, which are positively correlated with the fasting serum glucose concentration and preceding the development of representative metabolic syndrome symptoms, including dyslipidemia (low HDL and high plasma TG levels) [135,138]. Interestingly, mice subjected to HFD feeding exhibited increased serum leptin levels in conjunction with hepatic ANPTL6 expression, which were normalized via exercise training in both animals and humans [139]. In particular, the leptin treatment significantly increased hepatic ANGPTL6 expression at both the transcript and protein level in vitro and in vivo, indicating that ANGPTL6 is indeed directly regulated via leptin signaling in the context of metabolic disturbance [139]. These findings strongly support the therapeutic potential of ANGPTL6 against metabolic disorders, necessitating a more comprehensive approach for the elucidation of its molecular mechanism of action in the future.
ANGPTL8, also referred to as lipasin, betatrophin, and RIFL (refeeding induced in fat and liver), has recently been identified as the 8th member of the ANGPTL family, while possessing distinct structural features compared to the rest, as exemplified by the lack of a C-terminal fibrinogen/angiopoietin-like domain [40,44,101]. ANGPTL8 is mainly produced and secreted from the liver and adipose tissue in mice, as well as humans, although the main site of ANGPTL8 expression may vary among species [140]. ANGPTL8 also plays an important role in regulating LPL activity in concert with ANGPTL3 and ANGPTL4, presumably due to the high structural homology of its N-terminal domain with ANGPTL3 and ANGPTL4 [45]. Interestingly, accumulating evidence has demonstrated that ANGPTL8 does not operate alone with regards to LPL regulation, rather forming a complex with ANGPTL3 to elicit a maximal inhibitory effect on LPL activity [31,141]. Conversely, ANGPTL8 binds to ANGPTL4 and impairs its ability to inhibit LPL activity [141], thus acting as an endogenous inhibitor of ANGPTL4.
ANGPTL8 originally drew significant interest based on the initial findings regarding its possible roles in the induction of pancreatic β cell proliferation, where the term ‘betatrophin’ initially came from [44,142]. However, a series of subsequent studies deemed these findings irreproducible [142], reporting a marginal effect of ANGPTL8 on β cell proliferation [44,143,144,145]. Recent studies have provided greater insight into the role of ANGPTL8 in other metabolic processes, with adenovirus-mediated Angptl8 overexpression reducing fasting blood glucose levels and improving glucose tolerance, as well as insulin sensitivity in diabetic mice via the suppression of hepatic gluconeogenic gene expression downstream of Akt [146]. ANGPTL8 has been reported to negatively regulate NF-ĸB, a critical driver of inflammatory signaling, by triggering the autophagic degradation of IKKγ, an inhibitory regulator of IĸBα [44,147]. Of note, the treatment with Angptl8 anti-sense oligonucleotides markedly alleviated HFD-induced hepatic steatosis in rodents [148], presumably due to ANGPTL8 promoting lipogenesis via upregulating nuclear sterol-regulatory element binding protein 1 (SREBP1) [149]. Moreover, the treatment of 3T3-L1 cells or HepG2 cells with recombinant ANGPTL8 resulted in a significant suppression of lipolysis, as indicated by the decreased expression of adipose TG lipase and hormone-sensitive lipase through Akt and mTOR activation [44,46]. Furthermore, the overexpression of ANPTGL8 in the liver improved insulin sensitivity and glucose tolerance, with enhanced Akt phosphorylation and improved insulin signaling in isolated primary hepatocytes [47].
Hepatic ANGPTL8 expression is upregulated in response to a number of nutritional stress conditions, including fasting/refeeding, hyperglycemia, hyperlipidemia, and endoplasmic reticulum stress, many of which are also well-established risk factors for NAFLD progression [44,48,150,151]. While accumulating evidence indicates the potential of ANGPTL8 as a novel biomarker and/or therapeutic target for a myriad of metabolic disorders, there have been conflicting results showing a bidirectional correlation of circulating ANGPTL8 levels with NAFLD, obesity, type 2 diabetes, and dyslipidemia [15,44,152,153,154,155,156], which altogether, warrant a further comprehensive investigation.

4.2. Fetuin-A and Fetuin-B

Fetuins are glycoproteins primarily synthesized from the liver and secreted into the bloodstream. Fetuins belong to the cystatin superfamily of proteins that mediate the transport of multiple cargo substances in the blood as carriers. While serum albumin is the representative and most abundant plasma carrier protein in adults, Fetuins are more enriched in fetal blood, which are identified as the major plasma proteins during fetal life, peaking in concentration shortly after birth, in a transient fashion [157]. Accumulating evidence has revealed that Fetuins, namely Fetuin-A and Fetuin-B, play crucial roles in a variety of cellular processes within the context of metabolic homeostasis.
Fetuin-A, also known as alpha2-HS-glycoprotein, is the first hepatokine discovered to mediate organ-to-organ communication in the regulation of metabolic homeostasis [12,13,14,15]. Fetuin-A is predominantly produced and secreted from the liver, with recent studies describing other sources of circulating Fetuin-A, including visceral and subcutaneous adipose tissue [158,159,160,161]. Fetuin-A initially drew great attention as a natural inhibitor of the insulin receptor tyrosine kinase in the liver, adipose tissue, and skeletal muscle [13,49,50,51]. These findings were further supported by improved insulin signaling, decreased body fat, and protection from diet-induced body weight gain in Fetuin-A gene knockout mice [52]. Furthermore, a positive correlation of Fetuin-A expression level in the liver with the expression of key glucose metabolism enzymes phosphoenolpyruvate kinase 1 and glucose-6-phosphatase has been reported [12,162], suggesting its fundamental role in insulin signaling and energy metabolism. Subsequent studies further demonstrated that circulating Fetuin-A protein levels were increased in NAFLD patients with insulin resistance and obesity, independent of adiposity [162,163,164,165]. In addition, several studies provided valuable insights for understanding Fetuin-A’s mechanism of action in the induction of insulin resistance, with Fetuin-A provoking the inflammatory response by increasing pro-inflammatory cytokine production in monocytes and adipocytes [53]. Furthermore, Fetuin-A serves as an endogenous ligand for toll-like receptor 4, mediating free fatty acid-inflicted TLR4 signaling activation to induce insulin resistance (Figure 2) [12,15,54,55]. In line with this notion, Fetuin-A was found to negatively regulate adiponectin production, thus affecting systemic insulin resistance [15,166,167]. Based on its role in metabolic processes, numerous pharmacological intervention studies for Fetuin-A have been examined in the metabolic disease model and it was found that liraglutide and pioglitazone reduce the circulating levels of Fetuin-A, while salsalate decreases Fetuin-A mRNA levels [115,168,169,170]. Collectively, given the significant contribution of Fetuin-A in inflammatory signaling and metabolic processes, it might represent a potential target for the treatment of metabolic diseases, including insulin resistance and type 2 diabetes.
Fetuin-B is another member of the Fetuin family, which shares ~24% of sequence homology with Fetuin-A in rodents and humans [157,161]. Similar to Fetuin-A, Fetuin-B is primarily produced and secreted by the liver, with its expression also detected in other tissues, yet to a lesser extent [157,161]. Recent cell-based studies revealed that Fetuin-B induces insulin resistance in cultured hepatocytes, as well as myotubes [26] and promotes lipid accumulation in HepG2 cells presumably via its capacity for decreasing AMPK activity, while activating liver-X-receptor α-SREBP-1c signaling (Figure 2) [56]. In line with these observations, Fetuin-B administration to lean mice resulted in glucose intolerance, with a mild effect on insulin resistance [26], while its liver-specific knockdown improved glucose tolerance in mice [26], in addition to lowering TG levels in the liver and plasma [56]. Emerging evidence from human studies has also demonstrated that circulating Fetuin-B levels are upregulated in patients with NAFLD, which are closely associated with insulin resistance [26,171]. Furthermore, the plasma concentration of Fetuin-B significantly correlated with clinical indices of obesity, hepatic steatosis, and homeostatic model assessment for insulin resistance [172]. Given their close relationship, the plasma levels of both Fetuin-A and -B were elevated and exhibited an inverse correlation with liver fibrosis stage in NAFLD patients [173,174]. However, when the association between Fetuin-A and -B was investigated in two independent cohorts, it was described that they may regulate glucose metabolism in slightly different manners, as Fetuin-A seems to modulate insulin signaling, while Fetuin-B may affect glucose effectiveness [175], suggesting that more comprehensive approaches are required to define how Fetuin-B regulates these metabolic processes in concert with Fetuin-A. Nevertheless, based on the findings from a number of studies, Fetuin-B is another potential therapeutic target in the treatment of metabolic diseases, such as obesity and insulin resistance.

4.3. Fibroblast Growth Factor 21 (FGF21)

Based on numerous studies that demonstrate its broad and significant impact on systemic energy metabolism in response to various metabolic challenges, hepatokine FGF21 holds promise as a potential therapeutic for a wide range of metabolic abnormalities, including obesity and type 2 diabetes [12,13,14,15,176]. FGF21 is expressed in various organs, including the liver, white and brown adipose tissue, as well as the pancreas [57,176], but circulating FGF21 is predominantly derived from the liver [177]. Accumulating studies have demonstrated that the plasma levels of FGF21 increase in parallel with the severity of hepatic steatosis in humans [176,178], and are positively associated with TG, fasting insulin, and other insulin resistance indices, while negatively correlated with body mass index and HDL [179,180]. In agreement with this notion, serum FGF21 levels were positively correlated with the steatosis grade and NAFLD activity score in obese children and patients with advanced NASH, respectively [30,181,182]. Taken together, these results strongly suggest that the circulating FGF21 level may reflect the severity of hepatic steatosis and may indicate the early onset of NAFLD progression, thus representing a candidate biomarker of metabolic disorders [12,180]. Interestingly, emerging evidence also suggests that FGF21 positively regulates a variety of cellular processes and improves various metabolic abnormalities, as it markedly promotes glucose uptake in cultured adipocytes [57], enhances pancreatic β cell function and insulin secretion [58], upregulates fatty acid oxidation and insulin sensitivity, and improves hepatic steatosis and glycemic control [58,59,60]. The beneficial roles of FGF21 on metabolism are further validated by several studies using animal models deficient for FGF21 or its cognate receptor, which exhibited increased adiposity, exacerbated hepatic fat accumulation, insulin resistance, and hyperglycemia [14,183,184], suggesting that circulating FGF21 may be increased in patients with NAFLD in order to maintain energy homeostasis under metabolic stress. Of note, FGF21 levels are regulated by various physiological stimuli, such as fasting and exercise. As a representative fasting hormone, the expression of FGF21 is increased following prolonged fasting through the induction of PPARα and mediates its beneficial metabolic effects (Figure 2) [185,186]. In addition, physical training upregulated serum FGF21 levels in humans and rodents, in conjunction with increased FGF21 gene expression in skeletal muscle and liver, as well as enhanced effects of FGF21 in adipose tissue [187,188,189]. Given the promising therapeutic efficacy against obesity and type 2 diabetes in preclinical studies, several FGF21 analogues and mimetics have been developed as pharmacological intervention strategies and are currently undergoing several clinical trials in patients with obesity, type 2 diabetes, and NASH [115,190,191,192,193]. To date, these FGF21 analogues have shown substantial improvement in dyslipidemia, while exerting a marginal effect on glycemic control in patients with obesity and type 2 diabetes [115,190,191,192]. Additionally, these pharmacological interventions ameliorated hepatic steatosis, as well as several biomarkers of liver fibrosis, although the beneficial effects on histology and other clinical outcomes of NASH were not satisfactory [190,191,193], suggesting that more comprehensive and targeted approaches are required to improve the therapeutic efficacy and safety of FGF21 analogues.

4.4. Selenoprotein P

Selenoprotein P was initially identified as a carrier protein containing 10 selenocysteine residues, which are responsible for transporting selenium from the liver to extrahepatic tissues, including the brain and testes [194,195]. Selenoprotein P is a glycoprotein primarily secreted by the liver, and its expression is increased in humans with NAFLD, type 2 diabetes, and cardiovascular diseases, in close association with insulin resistance and hyperlipidemia [61,196,197]. In line with this notion, hepatic selenoprotein P expression is upregulated in animal models of diet-induced obesity and type 2 diabetes [14,61,197]. Moreover, circulating selenoprotein P levels are negatively correlated with adiponectin levels in type 2 diabetes patients, in agreement with observations from selenoprotein P-deficient mice [198], suggesting its involvement in signaling crosstalk with other organokines. Selenoprotein P treatment of cultured primary hepatocytes or immortalized myocytes inhibited insulin signaling, as indicated by the diminished insulin-stimulated phosphorylation of the insulin receptor and Akt, which led to an increase in hepatic glucose production and a decrease in glucose uptake by myotubes [61]. Likewise, in vivo administration of selenoprotein P led to hepatic and peripheral insulin resistance, while liver-specific genetic ablation significantly ameliorated systemic glucose intolerance in association with enhanced insulin signaling in the liver and skeletal muscle [12,61,62,63,64]. Collectively, most of the studies on the role of selenoprotein P in the progression of insulin resistance and type 2 diabetes strongly support its potential as a novel therapeutic target in various metabolic disorders.

4.5. Leukocyte Cell-Derived Chemotaxin 2 (LECT2)

LECT2 is a liver-secreted protein that was initially identified as a neutrophil chemotactic factor promoting the growth of osteoblasts and chondrocytes [199]. LECT2 is primarily expressed and secreted from the liver, and, to a lesser extent, from adipose tissue, neurons, as well as white blood cells [15]. LECT2 is highly sensitive to changes in dietary fat content, as well as to the severity of hepatic steatosis. For example, LECT2 serum levels are rapidly increased prior to body weight gain [200] and exhibit a positive correlation with the extent of NAFLD, insulin resistance, hepatic inflammation, and liver fibrosis [65,200,201,202,203]. Moreover, HFD feeding increased circulating and hepatic LECT2 levels in mice and humans [65,189], while the opposite result was obtained in mice following exercise training or pharmacological inhibition of dipeptidyl peptidase-4, as mediated via augmented AMPK phosphorylation [65,204]. Consistently, recent studies have also demonstrated less M1-type macrophages, as well as a lower M1/M2 ratio in the livers of LECT2-deficient mice, in close association with reduced hepatic inflammation (Figure 2) [66], implicating that increased LECT2 levels may be an early manifestation of NAFLD progression. In line with this notion, LECT2-deficient mice fed an HFD exhibited improved insulin sensitivity in skeletal muscle, as opposed to the mice administered recombinant LECT2 protein, which developed insulin resistance in skeletal muscle [65]. Interestingly, the loss of LECT2 did not affect insulin sensitivity in liver and adipose tissue [65], while LECT2 treatment impaired insulin signaling in both cultured myotubes and differentiated 3T3-L1 cells by inhibiting Akt phosphorylation [65,205], thus implying the existence of a complex, unknown regulatory mechanism of LECT2 in vivo.

4.6. Follistatin

Follistatin is a secretory glycoprotein that is expressed in almost every tissue throughout the body, with the liver serving as the primary organ responsible for the production of circulating follistatin [206]. Follistatin is a member of the TGFβ family and was initially identified as an inhibitor of follicle-stimulating hormone production in the pituitary gland [14,67]. Subsequent evidence revealed its myostatin-antagonizing effect, which suppresses skeletal muscle growth [14,15]. Recently, follistatin has drawn significant attention due to its possible association with energy metabolism. The serum follistatin level is increased in patients with NAFLD and type 2 diabetes [207,208], while individuals with reduced body weight after bariatric surgery exhibited a decrease in serum follistatin level, in conjunction with improved insulin sensitivity and glycemic control [68,209]. Interestingly, circulating follistatin levels are also controlled by other physiological factors, including prolonged fasting and/or exercise, both of which are characterized by a high glucagon-to-insulin ratio due to the increased energy demand [210]. Nevertheless, the physiological role of circulating follistatin remains to be fully characterized, as existing evidence from studies of genetically manipulated animals has been somewhat confusing. Mice with liver-specific overexpression of follistatin exhibited increased hepatic glucose production, as well as aggravated insulin resistance in adipose tissue and skeletal muscle accompanying whole-body glucose intolerance, while follistatin knockdown improved insulin sensitivity [14,15,68,69]. Therefore, follistatin may contribute to metabolic disease progression. On the other hand, it has been reported that increased follistatin levels following exercise promote glucose and free fatty acid uptake in skeletal muscle, inducing the differentiation of brown adipocytes [70]. Furthermore, follistatin promoted thermogenesis by upregulating the uncoupling protein 1 expression in both brown and white adipose tissues [70,71]. Collectively, given the promising role of follistatin in a broad range of biological processes, including energy metabolism, a more comprehensive investigation is necessary to clearly understand its physiological role and therapeutic potential in metabolic disease.

4.7. Hepassocin

Hepassocin, also known as hepatocyte-derived fibrinogen-related protein 1, is upregulated during liver regeneration and is frequently downregulated in hepatocellular carcinoma [211,212]. Its expression is transcriptionally regulated by hepatocyte nuclear factor-1α [212]. The therapeutic effect of recombinant human hepassocin has been reported for fulminant hepatic failure [213]. Hepassocin enhances the proliferation of hepatocytes in vitro and in vivo, reducing hepatocyte apoptosis. Furthermore, the administration of recombinant human hepassocin successfully improved survival in rats with chemically-induced fulminant hepatic failure [213].
Hepassocin contributes to insulin resistance and type 2 diabetes. Plasma hepassocin levels were increased in participants with impaired fasting glucose, impaired glucose tolerance, and newly diagnosed diabetes, when compared to those with normal glucose tolerance [72]. High glucose levels promote hepassocin expression via the STAT3 and PP2A-HNF1 pathways [214]. In patients with hyperglycemic crisis or streptozotocin-induced hyperglycemic mice, the increase in blood or hepatic levels of hepassocin was diminished after the hyperglycemia treatment [214]. Similar results were obtained in HFD-fed mice following the metformin or rosiglitazone treatment [72]. Both the administration of recombinant hepassocin and its hepatic overexpression promoted insulin resistance in mice, whereas hepassocin knockdown had the opposite effect [72]. Mechanistically, ERK1/2 was shown to mediate hepassocin-induced insulin resistance [185]. Another research group also demonstrated that the recombinant hepassocin treatment impaired insulin sensitivity in differentiated C2C12 cells through JNK activation and the suppression of basal AMPK phosphorylation [73]. These results support a role for hepassocin in the development of insulin resistance.
Serum hepassocin levels are elevated in subjects with NAFLD compared to those without NAFLD [74]. The treatment of hepatocytes with palmitate upregulated hepassocin expression through CCAAT/enhancer-binding protein β (C/EBPβ). Overexpression of hepassocin in hepatocytes upregulated lipogenic proteins, such as fatty acid synthase, acetyl-CoA carboxylase, and mature SREBP-1, in an ERK1/2-dependent manner, thereby increasing hepatic TG accumulation [188]. In addition, liver injury and inflammatory cytokines, such as interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNFα), were induced via the hepatic overexpression of hepassocin. Furthermore, the scores for all the histological features comprising the NAFLD activity score (i.e., steatosis, hepatocyte ballooning, and lobular inflammation) were higher in hepassocin-transgenic mice than control GFP-transgenic mice. In contrast, the hepatic knockdown of hepassocin attenuated HFD-induced NAFLD in mice [188]. Taken together, hepassocin plays an important role in hepatic steatosis.
Obesity is closely associated with insulin resistance and NAFLD. Hepassocin levels were significantly upregulated in overweight and obese subjects [215]. Moreover, serum hepassocin was positively correlated with obesity indices, including body mass index, waist circumference, and fat areas (total, visceral, and subcutaneous) in humans [75]. Overexpression of hepassocin in epididymal adipose tissue increased fat weight and the size of adipocytes along with the induction of fatty acid synthase and SREBP-1. Hepassocin enhanced adipogenesis through an ERK1/2-C/EBPβ-dependent pathway [190]. These studies indicate that hepassocin is not only a useful biomarker candidate for obesity and obesity-related metabolic diseases, but that hepassocin also contributes to adipogenesis, insulin resistance, and hepatic steatosis.

4.8. Retinol Binding Protein 4 (RBP4)

Liver is the major organ, where most of the vitamin A in our body is stored in the form of retinyl esters. The liver enzyme hydrolyzes the retinyl ester into retinol, which in turn binds to the member of lipocalin family, RBP4 in the hepatocytes. The primary function of RBP4 is to transport retinol into the circulation [216]. Two RBP4 receptors have been identified, which are responsible for the retinol uptake across the cell membrane [216]. RBP4 also plays an important role in metabolic syndrome. Yang et al. showed that RBP4 promotes insulin resistance in obesity and type 2 diabetes [79]. The insulin tolerance test was evaluated in mice expressing human RBP4 in muscle and mice injected chronically with purified RBP4. Both of the in vivo experiments showed that the elevated serum RBP4 caused insulin resistance. In contrast, insulin action was improved in RBP4 knockout mice or mice treated with fenretinide, a synthetic retinoid with a bulky side chain, which causes renal excretion of RBP4 and lowering serum RBP4 levels. The RBP4 treatment increased PEPCK and glucose production in hepatocytes [79]. RBP4 directly promoted basal lipolysis in adipocytes [76] and also triggered adipose tissue inflammation by increasing adipose tissue macrophage and CD4 T cell infiltration, resulting in systemic insulin resistance [81]. Hepatocytes are the major source of circulating RBP4 [217], but adipocytes also secrete it. Adipocyte-specific overexpression of RBP4 also increased hepatic gluconeogenic gene expression. In addition, it promoted the hepatic uptake of adipose-derived circulating free fatty acids and de novo lipogenesis, and suppressed hepatic free fatty acid oxidation [218]. The RBP4 overexpression contributes to hepatic LCAD hyperacetylation through the suppression of SIRT3 activity [78]. The study using RBP4 transgenic mice also demonstrated that RBP4 stimulated hepatic mitochondrial dysfunction and induced hepatic steatosis [78]. However, a model for liver-specific RBP4 overexpression in mice, established by adeno-associated viruses that express a highly liver-specific promoter (LP1)-RBP4, showed that liver-specific RBP4 overexpression did not induce glucose intolerance and had no effect on energy metabolism [82]. These contradictory results implicate that the tissue-specific RBP4 may have distinct roles in the development of insulin resistance.
Serum and adipose tissue RBP4 expression is subsequently increased in the obese subjects [76], whereas serum RBP4 levels are decreased with exercise and bariatric surgery [77,219]. In addition, those levels are elevated in patients with NAFLD, insulin resistance or type 2 diabetes [220,221,222]. Clinical evidence suggests that RBP4 is associated with hepatic steatosis and insulin resistance.

4.9. SPARC-Related Modular Calcium-Binding Protein 1 (SMOC1)

SMOC1 belongs to the BM-40 family, which is characterized by an extracellular calcium-binding domain and follistatin-like domain [223]. SMOC1 is a glycoprotein, which exhibits a widespread expression in various tissues [224]. It regulates embryonic development and pathophysiological processes, including osteoblast differentiation, ocular and limb development, angiogenesis, aortic valve calcification, and thrombin activation [225,226,227,228,229,230]. Montgomery et al. demonstrated that SMOC1 is a glucose-responsive hepatokine [83]. SMOC1 attenuated cAMP-PKA signaling and hepatic glucose output, and thereby improved glycemic control in lean, obese pre-diabetic, and diabetic db/db mice. Daily injection of SMOC1 recombinant protein for 3 weeks failed to show long-lasting metabolic improvement due to its rapid clearance, but a SMOC1-Fc fusion protein (fused to human immunoglobulin G1 Fc) exhibited the increased stability and therapeutic potential. Interestingly, the SMOC1-Fc treatment improved glycemic control, even better than the first-line diabetes medication metformin. However, it did not change the body weight and food intake. Plasma SMOC1 levels are downregulated in obese, insulin-resistant humans, and its levels are positively correlated with the glucose infusion rate during the hyperinsulinemic-euglycemic clamp and hepatic insulin sensitivity [83]. These results indicate that SMOC1 can be a promising therapeutic strategy in patients with type 2 diabetes.

4.10. Growth Differentiation Factor 15 (GDF15)

GDF15, a member of the TGF-β superfamily, is the ligand for the GFRAL receptor, which is abundantly expressed in neurons of the area postrema and nucleus of the solitary tract in mice and humans [89,231]. When GDF15 binds to GFRAL, it forms a complex with a co-receptor, RET. Then, RET is phosphorylated, which leads to downstream signaling via AKT, ERK1/2, and phospholipase C-γ [87]. Several inflammatory cytokines, such as IL-1β, IL-2, and TNF-α, increase GDF15 expression, and thereby circulating GDF15 levels are elevated in diabetes and cardiovascular diseases [15,232]. Circulating GDF15 levels are also increased in response to metformin, colchicine, AICAR, and cisplatin [233]. The increasing serum GDF15 expression is associated with weight loss in patients with advanced prostate cancer [234]. GDF15 has potent anti-obesity actions [88]. GDF15 increased thermogenesis, lipolysis, and oxidative metabolism [85]. The treatment of human GDF15 decreased food intake, resulted in weight loss, and improved insulin sensitivity, which was due to the increased oxidative metabolism and lipolysis [86,235]. Transgenic mice that ubiquitously overexpress the human GDF15 weighed less, although the food consumption was similar to wild-type mice [236]. Both the GDF15-treated mice and GDF15 transgenic mice exhibited less body weight compared to the respective controls. However, the effect of GDF15 administration and GDF15 transgenic mice on food intake was conflicting. A recent study from Borner et al. provided another evidence that GDF15 has an anorectic action through nausea and emesis [90]. GFRAL is essential for GDF15-induced food intake reduction and weight loss in mice [87]. GFRAL knockout mice exacerbated diet-induced obesity and insulin resistance [87]. These findings demonstrate that targeting GDF15-GFRAL may be an attractive therapeutic approach for obesity and comorbidities or eating disorders.

5. Conclusions and Perspectives

The global prevalence of NAFLD currently stands at approximately 25%, and its incidence is rapidly growing due to the obesity pandemic [237]. The liver as a key nutrient-sensing organ is dynamically exposed to changes in nutrient availability through the gut-liver axis or systemically and regulates the whole-body energy metabolism. This anatomical and physiological characteristic of the liver can partially explain why NAFLD progression frequently precedes other forms of metabolic diseases, such as type 2 diabetes and dyslipidemia. It is now widely appreciated that the impact of NAFLD on whole-body metabolic diseases is primarily attributed to the dysregulated secretome profile of steatotic hepatocytes following metabolic stress.
Hepatokine profiles are greatly affected by the metabolic status (Table 1). The liver communicates with other major metabolic organs to maintain the energy balance by producing and releasing hepatokines, which play an important role in this inter-organ communication network. Emerging evidence has now revealed that a number of hepatokines possess a metabolic capacity to regulate a myriad of biological processes in multiple extrahepatic tissues. In addition, hepatokines mediate the physiological benefit and/or influence in certain circumstances, such as exercise [238] and fasting/refeeding transition [239,240], suggesting that hepatokines can be attractive and important targets for the maintenance of metabolic homeostasis. Hepatokine dysregulation is implicated in the development of NAFLD and insulin resistance. Given the molecular features and metabolic functions of hepatokines, it is highly likely that both the chronic liver disease (namely NAFLD) and metabolic disturbances at the systemic level can be concurrently ameliorated by targeting candidate hepatokine(s) that exhibit autocrine, paracrine, and endocrine roles.
Currently, a liver biopsy is the gold standard for NAFLD and NASH diagnosis. However, several limitations, including the risk of bleeding or infection have restricted its widespread utilization, suggestive of the necessity for the development of reliable and valid non-invasive biomarkers. In NAFLD, the secretion of several hepatokines, such as Fetuin-A, Fetuin-B, FGF21, selenoprotein P, LECT2, follistatin, and hepassocin, is upregulated. This holds promise for the development of multi-hepatokine biomarker panels for NAFLD and NASH. The identification and evaluation of hepatokines, that are dynamically and specifically modulated under various physiological statuses, may be of help in developing new biomarkers, since this would provide more insights in deciphering the chronic disease progression stage and thus, developing the personalized therapy strategies that have multiple metabolic complications (e.g., metabolic syndrome).
Collectively, we summarized and discussed the role of hepatokines in the regulation of lipid and glucose metabolism at molecular, cellular, and systemic levels. In vitro and in vivo studies have shown that hepatokine inhibition, overexpression or recombinant hepatokine treatment have a major influence on NAFLD and/or insulin resistance. Based on these preclinical findings, hepatokine mimetics or inhibitors may represent effective therapeutics for NAFLD and other metabolic diseases.

Author Contributions

Conceptualization and writing of the original draft, T.H.K. and Y.M.Y.; editing figures, D.-G.H.; review, T.H.K., D.-G.H. and Y.M.Y.; supervision, Y.M.Y. All authors have read and agreed to the published version of the manuscript.

Funding

Y.M.Y. was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea Government (MSIT) (Nos. 2021R1A4A3031661 and 2020R1C1C1004185). T.H.K. was supported by Sookmyung Women’s University Research grants (1–2103–2009).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Blüher, M. Obesity: Global epidemiology and pathogenesis. Nat. Rev. Endocrinol. 2019, 15, 288–298. [Google Scholar] [CrossRef] [PubMed]
  2. Lee, Y.S.; Wollam, J.; Olefsky, J.M. An Integrated View of Immunometabolism. Cell 2018, 172, 22–40. [Google Scholar] [CrossRef] [Green Version]
  3. Hotamisligil, G.S. Inflammation, metaflammation and immunometabolic disorders. Nature 2017, 542, 177–185. [Google Scholar] [CrossRef] [PubMed]
  4. Yki-Järvinen, H. Non-alcoholic fatty liver disease as a cause and a consequence of metabolic syndrome. Lancet Diabetes Endocrinol. 2014, 2, 901–910. [Google Scholar] [CrossRef]
  5. Younossi, Z.; Tacke, F.; Arrese, M.; Sharma, B.C.; Mostafa, I.; Bugianesi, E.; Wong, V.W.; Yilmaz, Y.; George, J.; Fan, J.; et al. Global Perspectives on Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis. Hepatology 2019, 69, 2672–2682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Younossi, Z.M. Non-alcoholic fatty liver disease–A global public health perspective. J. Hepatol. 2019, 70, 531–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Younossi, Z.; Anstee, Q.M.; Marietti, M.; Hardy, T.; Henry, L.; Eslam, M.; George, J.; Bugianesi, E. Global burden of NAFLD and NASH: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 11–20. [Google Scholar] [CrossRef] [PubMed]
  8. Adenote, A.; Dumic, I.; Madrid, C.; Barusya, C.; Nordstrom, C.W.; Rueda Prada, L. NAFLD and Infection, a Nuanced Relationship. Can. J. Gastroenterol. Hepatol. 2021, 2021, 5556354. [Google Scholar] [CrossRef]
  9. Drucker, D.J. Diabetes, obesity, metabolism, and SARS-CoV-2 infection: The end of the beginning. Cell Metab. 2021, 33, 479–498. [Google Scholar] [CrossRef]
  10. Eslam, M.; Newsome, P.N.; Sarin, S.K.; Anstee, Q.M.; Targher, G.; Romero-Gomez, M.; Zelber-Sagi, S.; Wai-Sun Wong, V.; Dufour, J.; Schattenberg, J.M.; et al. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J. Hepatol. 2020, 73, 202–209. [Google Scholar] [CrossRef] [PubMed]
  11. Eslam, M.; Sanyal, A.J.; George, J. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology 2020, 158, 1999–2014. [Google Scholar] [CrossRef]
  12. Meex, R.C.R.; Watt, M.J. Hepatokines: Linking nonalcoholic fatty liver disease and insulin resistance. Nat. Rev. Endocrinol. 2017, 13, 508–520. [Google Scholar] [CrossRef] [PubMed]
  13. Stefan, N.; Häring, H. The role of hepatokines in metabolism. Nat. Rev. Endocrinol. 2013, 9, 144–152. [Google Scholar] [CrossRef]
  14. Watt, M.J.; Miotto, P.M.; De Nardo, W.; Montgomery, M.K. The Liver as an Endocrine Organ-Linking NAFLD and Insulin Resistance. Endocr. Rev. 2019, 40, 1367–1393. [Google Scholar] [CrossRef]
  15. Jensen-Cody, S.O.; Potthoff, M.J. Hepatokines and metabolism: Deciphering communication from the liver. Mol. Metab. 2021, 44, 101138. [Google Scholar] [CrossRef] [PubMed]
  16. Turner, N.; Kowalski, G.M.; Leslie, S.J.; Risis, S.; Yang, C.; Lee-Young, R.S.; Babb, J.R.; Meikle, P.J.; Lancaster, G.I.; Henstridge, D.C.; et al. Distinct patterns of tissue-specific lipid accumulation during the induction of insulin resistance in mice by high-fat feeding. Diabetologia 2013, 56, 1638–1648. [Google Scholar] [CrossRef] [PubMed]
  17. Samuel, V.T.; Liu, Z.; Qu, X.; Elder, B.D.; Bilz, S.; Befroy, D.; Romanelli, A.J.; Shulman, G.I. Mechanism of hepatic insulin resistance in non-alcoholic fatty liver disease. J. Biol. Chem. 2004, 279, 32345–32353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Roden, M. Mechanisms of Disease: Hepatic steatosis in type 2 diabetes--pathogenesis and clinical relevance. Nat. Clin. Pract. Endocrinol. Metab. 2006, 2, 335–348. [Google Scholar] [CrossRef]
  19. Ramnanan, C.J.; Saraswathi, V.; Smith, M.S.; Donahue, E.P.; Farmer, B.; Farmer, T.D.; Neal, D.; Williams, P.E.; Lautz, M.; Mari, A.; et al. Brain insulin action augments hepatic glycogen synthesis without suppressing glucose production or gluconeogenesis in dogs. J. Clin. Investig. 2011, 121, 3713–3723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Hodson, L.; Gunn, P.J. The regulation of hepatic fatty acid synthesis and partitioning: The effect of nutritional state. Nat. Rev. Endocrinol. 2019, 15, 689–700. [Google Scholar] [CrossRef] [PubMed]
  21. Geisler, C.E.; Ghimire, S.; Bogan, R.L.; Renquist, B.J. Role of ketone signaling in the hepatic response to fasting. Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 316, G623–G631. [Google Scholar] [CrossRef]
  22. Kim, M.; Pinto, S.M.; Getnet, D.; Nirujogi, R.S.; Manda, S.S.; Chaerkady, R.; Madugundu, A.K.; Kelkar, D.S.; Isserlin, R.; Jain, S.; et al. A draft map of the human proteome. Nature 2014, 509, 575–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A.; et al. Proteomics. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef] [PubMed]
  24. Lai, K.K.Y.; Kolippakkam, D.; Beretta, L. Comprehensive and quantitative proteome profiling of the mouse liver and plasma. Hepatology 2008, 47, 1043–1051. [Google Scholar] [CrossRef]
  25. Choi, K.M. The Impact of Organokines on Insulin Resistance, Inflammation, and Atherosclerosis. Endocrinol. Metab. 2016, 31, 1–6. [Google Scholar] [CrossRef]
  26. Meex, R.C.; Hoy, A.J.; Morris, A.; Brown, R.D.; Lo, J.C.Y.; Burke, M.; Goode, R.J.A.; Kingwell, B.A.; Kraakman, M.J.; Febbraio, M.A.; et al. Fetuin B Is a Secreted Hepatocyte Factor Linking Steatosis to Impaired Glucose Metabolism. Cell Metab. 2015, 22, 1078–1089. [Google Scholar] [CrossRef] [Green Version]
  27. Dijk, W.; Kersten, S. Regulation of lipid metabolism by angiopoietin-like proteins. Curr. Opin. Lipidol. 2016, 27, 249–256. [Google Scholar] [CrossRef] [Green Version]
  28. Kersten, S. Angiopoietin-like 3 in lipoprotein metabolism. Nat. Rev. Endocrinol. 2017, 13, 731–739. [Google Scholar] [CrossRef]
  29. Chi, X.; Britt, E.C.; Shows, H.W.; Hjelmaas, A.J.; Shetty, S.K.; Cushing, E.M.; Li, W.; Dou, A.; Zhang, R.; Davies, B.S.J. ANGPTL8 promotes the ability of ANGPTL3 to bind and inhibit lipoprotein lipase. Mol. Metab. 2017, 6, 1137–1149. [Google Scholar] [CrossRef]
  30. Kucukoglu, O.; Sowa, J.; Mazzolini, G.D.; Syn, W.; Canbay, A. Hepatokines and adipokines in NASH-related hepatocellular carcinoma. J. Hepatol. 2021, 74, 442–457. [Google Scholar] [CrossRef]
  31. Wang, Y.; McNutt, M.C.; Banfi, S.; Levin, M.G.; Holland, W.L.; Gusarova, V.; Gromada, J.; Cohen, J.C.; Hobbs, H.H. Hepatic ANGPTL3 regulates adipose tissue energy homeostasis. Proc. Natl. Acad. Sci. USA 2015, 112, 11630–11635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Adam, R.C.; Mintah, I.J.; Alexa-Braun, C.A.; Shihanian, L.M.; Lee, J.S.; Banerjee, P.; Hamon, S.C.; Kim, H.I.; Cohen, J.C.; Hobbs, H.H.; et al. Angiopoietin-like protein 3 governs LDL-cholesterol levels through endothelial lipase-dependent VLDL clearance. J. Lipid Res. 2020, 61, 1271–1286. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, Y.; Gusarova, V.; Banfi, S.; Gromada, J.; Cohen, J.C.; Hobbs, H.H. Inactivation of ANGPTL3 reduces hepatic VLDL-triglyceride secretion. J. Lipid Res. 2015, 56, 1296–1307. [Google Scholar] [CrossRef] [Green Version]
  34. Zhu, P.; Goh, Y.Y.; Chin, H.F.A.; Kersten, S.; Tan, N.S. Angiopoietin-like 4: A decade of research. Biosci. Rep. 2012, 32, 211–219. [Google Scholar] [CrossRef]
  35. Yoshida, K.; Shimizugawa, T.; Ono, M.; Furukawa, H. Angiopoietin-like protein 4 is a potent hyperlipidemia-inducing factor in mice and inhibitor of lipoprotein lipase. J. Lipid Res. 2002, 43, 1770–1772. [Google Scholar] [CrossRef] [Green Version]
  36. Lichtenstein, L.; Berbée, J.F.P.; van Dijk, S.J.; van Dijk, K.W.; Bensadoun, A.; Kema, I.P.; Voshol, P.J.; Müller, M.; Rensen, P.C.N.; Kersten, S. Angptl4 upregulates cholesterol synthesis in liver via inhibition of LPL- and HL-dependent hepatic cholesterol uptake. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 2420–2427. [Google Scholar] [CrossRef] [Green Version]
  37. Sukonina, V.; Lookene, A.; Olivecrona, T.; Olivecrona, G. Angiopoietin-like protein 4 converts lipoprotein lipase to inactive monomers and modulates lipase activity in adipose tissue. Proc. Natl. Acad. Sci. USA 2006, 103, 17450–17455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. McQueen, A.E.; Kanamaluru, D.; Yan, K.; Gray, N.E.; Wu, L.; Li, M.; Chang, A.; Hasan, A.; Stifler, D.; Koliwad, S.K.; et al. The C-terminal fibrinogen-like domain of angiopoietin-like 4 stimulates adipose tissue lipolysis and promotes energy expenditure. J. Biol. Chem. 2017, 292, 16122–16134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Xu, A.; Lam, M.C.; Chan, K.W.; Wang, Y.; Zhang, J.; Hoo, R.L.C.; Xu, J.Y.; Chen, B.; Chow, W.; Tso, A.W.K.; et al. Angiopoietin-like protein 4 decreases blood glucose and improves glucose tolerance but induces hyperlipidemia and hepatic steatosis in mice. Proc. Natl. Acad. Sci. USA 2005, 102, 6086–6091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Hato, T.; Tabata, M.; Oike, Y. The role of angiopoietin-like proteins in angiogenesis and metabolism. Trends Cardiovasc. Med. 2008, 18, 6–14. [Google Scholar] [CrossRef] [PubMed]
  41. Qaddoumi, M.G.; Alanbaei, M.; Hammad, M.M.; Al Khairi, I.; Cherian, P.; Channanath, A.; Thanaraj, T.A.; Al-Mulla, F.; Abu-Farha, M.; Abubaker, J. Investigating the Role of Myeloperoxidase and Angiopoietin-like Protein 6 in Obesity and Diabetes. Sci. Rep. 2020, 10, 6170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Kitazawa, M.; Ohizumi, Y.; Oike, Y.; Hishinuma, T.; Hashimoto, S. Angiopoietin-related growth factor suppresses gluconeogenesis through the Akt/forkhead box class O1-dependent pathway in hepatocytes. J. Pharmacol. Exp. Ther. 2007, 323, 787–793. [Google Scholar] [CrossRef]
  43. Kang, S.G.; Yi, H.; Choi, M.J.; Ryu, M.J.; Jung, S.; Chung, H.K.; Chang, J.Y.; Kim, Y.K.; Lee, S.E.; Kim, H.; et al. ANGPTL6 expression is coupled with mitochondrial OXPHOS function to regulate adipose FGF21. J. Endocrinol. 2017, 233, 105–118. [Google Scholar] [CrossRef] [Green Version]
  44. Abu-Farha, M.; Ghosh, A.; Al-Khairi, I.; Madiraju, S.R.M.; Abubaker, J.; Prentki, M. The multi-faces of Angptl8 in health and disease: Novel functions beyond lipoprotein lipase modulation. Prog. Lipid Res. 2020, 80, 101067. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, R. Lipasin, a novel nutritionally-regulated liver-enriched factor that regulates serum triglyceride levels. Biochem. Biophys. Res. Commun. 2012, 424, 786–792. [Google Scholar] [CrossRef]
  46. Zhang, Y.; Li, S.; Donelan, W.; Xie, C.; Wang, H.; Wu, Q.; Purich, D.L.; Reeves, W.H.; Tang, D.; Yang, L. Angiopoietin-like protein 8 (betatrophin) is a stress-response protein that down-regulates expression of adipocyte triglyceride lipase. Biochim. Biophys. Acta 2016, 1861, 130–137. [Google Scholar] [CrossRef]
  47. Zhang, Z.; Wu, H.; Dai, L.; Yuan, Y.; Zhu, Y.; Ma, Z.; Ruan, X.; Guo, X. ANGPTL8 enhances insulin sensitivity by directly activating insulin-mediated AKT phosphorylation. Gene 2020, 749, 144707. [Google Scholar] [CrossRef] [PubMed]
  48. Nidhina Haridas, P.A.; Soronen, J.; Sädevirta, S.; Mysore, R.; Quagliarini, F.; Pasternack, A.; Metso, J.; Perttilä, J.; Leivonen, M.; Smas, C.M.; et al. Regulation of Angiopoietin-Like Proteins (ANGPTLs) 3 and 8 by Insulin. J. Clin. Endocrinol. Metab. 2015, 100, 1299. [Google Scholar] [CrossRef]
  49. Goustin, A.; Abou-Samra, A.B. The “thrifty” gene encoding Ahsg/Fetuin-A meets the insulin receptor: Insights into the mechanism of insulin resistance. Cell Signal 2011, 23, 980–990. [Google Scholar] [CrossRef]
  50. Mathews, S.T.; Chellam, N.; Srinivas, P.R.; Cintron, V.J.; Leon, M.A.; Goustin, A.S.; Grunberger, G. Alpha2-HSG, a specific inhibitor of insulin receptor autophosphorylation, interacts with the insulin receptor. Mol. Cell Endocrinol. 2000, 164, 87–98. [Google Scholar] [CrossRef]
  51. Auberger, P.; Falquerho, L.; Contreres, J.O.; Pages, G.; Le Cam, G.; Rossi, B.; Le Cam, A. Characterization of a natural inhibitor of the insulin receptor tyrosine kinase: cDNA cloning, purification, and anti-mitogenic activity. Cell 1989, 58, 631–640. [Google Scholar] [CrossRef]
  52. Mathews, S.T.; Singh, G.P.; Ranalletta, M.; Cintron, V.J.; Qiang, X.; Goustin, A.S.; Jen, K.C.; Charron, M.J.; Jahnen-Dechent, W.; Grunberger, G. Improved insulin sensitivity and resistance to weight gain in mice null for the Ahsg gene. Diabetes 2002, 51, 2450–2458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Mukhopadhyay, S.; Bhattacharya, S. Plasma fetuin-A triggers inflammatory changes in macrophages and adipocytes by acting as an adaptor protein between NEFA and TLR-4. Diabetologia 2016, 59, 859–860. [Google Scholar] [CrossRef] [Green Version]
  54. Pal, D.; Dasgupta, S.; Kundu, R.; Maitra, S.; Das, G.; Mukhopadhyay, S.; Ray, S.; Majumdar, S.S.; Bhattacharya, S. Fetuin-A acts as an endogenous ligand of TLR4 to promote lipid-induced insulin resistance. Nat. Med. 2012, 18, 1279–1285. [Google Scholar] [CrossRef]
  55. Stefan, N.; Häring, H. Circulating fetuin-A and free fatty acids interact to predict insulin resistance in humans. Nat. Med. 2013, 19, 394–395. [Google Scholar] [CrossRef] [PubMed]
  56. Zhou, W.; Yang, J.; Zhu, J.; Wang, Y.; Wu, Y.; Xu, L.; Yang, Y. Fetuin B aggravates liver X receptor-mediated hepatic steatosis through AMPK in HepG2 cells and mice. Am. J. Transl. Res. 2019, 11, 1498–1509. [Google Scholar] [PubMed]
  57. Fon Tacer, K.; Bookout, A.L.; Ding, X.; Kurosu, H.; John, G.B.; Wang, L.; Goetz, R.; Mohammadi, M.; Kuro-o, M.; Mangelsdorf, D.J.; et al. Research resource: Comprehensive expression atlas of the fibroblast growth factor system in adult mouse. Mol. Endocrinol. 2010, 24, 2050–2064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Xu, J.; Lloyd, D.J.; Hale, C.; Stanislaus, S.; Chen, M.; Sivits, G.; Vonderfecht, S.; Hecht, R.; Li, Y.; Lindberg, R.A.; et al. Fibroblast growth factor 21 reverses hepatic steatosis, increases energy expenditure, and improves insulin sensitivity in diet-induced obese mice. Diabetes 2009, 58, 250–259. [Google Scholar] [CrossRef] [Green Version]
  59. Berglund, E.D.; Li, C.Y.; Bina, H.A.; Lynes, S.E.; Michael, M.D.; Shanafelt, A.B.; Kharitonenkov, A.; Wasserman, D.H. Fibroblast growth factor 21 controls glycemia via regulation of hepatic glucose flux and insulin sensitivity. Endocrinology 2009, 150, 4084–4093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Coskun, T.; Bina, H.A.; Schneider, M.A.; Dunbar, J.D.; Hu, C.C.; Chen, Y.; Moller, D.E.; Kharitonenkov, A. Fibroblast growth factor 21 corrects obesity in mice. Endocrinology 2008, 149, 6018–6027. [Google Scholar] [CrossRef] [PubMed]
  61. Misu, H.; Takamura, T.; Takayama, H.; Hayashi, H.; Matsuzawa-Nagata, N.; Kurita, S.; Ishikura, K.; Ando, H.; Takeshita, Y.; Ota, T.; et al. A liver-derived secretory protein, selenoprotein P, causes insulin resistance. Cell Metab. 2010, 12, 483–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Zeng, M.; Li, X.; Liu, Y.; Zhao, H.; Zhou, J.; Li, K.; Huang, J.; Sun, L.; Tang, J.; Xia, X.; et al. A high-selenium diet induces insulin resistance in gestating rats and their offspring. Free Radic. Biol. Med. 2012, 52, 1335–1342. [Google Scholar] [CrossRef] [Green Version]
  63. Hellwege, J.N.; Palmer, N.D.; Ziegler, J.T.; Langefeld, C.D.; Lorenzo, C.; Norris, J.M.; Takamura, T.; Bowden, D.W. Genetic variants in selenoprotein P plasma 1 gene (SEPP1) are associated with fasting insulin and first phase insulin response in Hispanics. Gene 2014, 534, 33–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Ko, B.J.; Kim, S.M.; Park, K.H.; Park, H.S.; Mantzoros, C.S. Levels of circulating selenoprotein P, fibroblast growth factor (FGF) 21 and FGF23 in relation to the metabolic syndrome in young children. Int. J. Obes. 2014, 38, 1497–1502. [Google Scholar] [CrossRef]
  65. Lan, F.; Misu, H.; Chikamoto, K.; Takayama, H.; Kikuchi, A.; Mohri, K.; Takata, N.; Hayashi, H.; Matsuzawa-Nagata, N.; Takeshita, Y.; et al. LECT2 functions as a hepatokine that links obesity to skeletal muscle insulin resistance. Diabetes 2014, 63, 1649–1664. [Google Scholar] [CrossRef] [Green Version]
  66. Takata, N.; Ishii, K.; Takayama, H.; Nagashimada, M.; Kamoshita, K.; Tanaka, T.; Kikuchi, A.; Takeshita, Y.; Matsumoto, Y.; Ota, T.; et al. LECT2 as a hepatokine links liver steatosis to inflammation via activating tissue macrophages in NASH. Sci. Rep. 2021, 11, 555. [Google Scholar] [CrossRef]
  67. Ueno, N.; Ling, N.; Ying, S.Y.; Esch, F.; Shimasaki, S.; Guillemin, R. Isolation and partial characterization of follistatin: A single-chain Mr 35,000 monomeric protein that inhibits the release of follicle-stimulating hormone. Proc. Natl. Acad. Sci. USA 1987, 84, 8282–8286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Tao, R.; Wang, C.; Stöhr, O.; Qiu, W.; Hu, Y.; Miao, J.; Dong, X.C.; Leng, S.; Stefater, M.; Stylopoulos, N.; et al. Inactivating hepatic follistatin alleviates hyperglycemia. Nat. Med. 2018, 24, 1058–1069. [Google Scholar] [CrossRef] [PubMed]
  69. Wu, H.; Ballantyne, C.M. Skeletal muscle inflammation and insulin resistance in obesity. J. Clin. Investig. 2017, 127, 43–54. [Google Scholar] [CrossRef]
  70. Braga, M.; Reddy, S.T.; Vergnes, L.; Pervin, S.; Grijalva, V.; Stout, D.; David, J.; Li, X.; Tomasian, V.; Reid, C.B.; et al. Follistatin promotes adipocyte differentiation, browning, and energy metabolism. J. Lipid Res. 2014, 55, 375–384. [Google Scholar] [CrossRef] [Green Version]
  71. Singh, R.; Braga, M.; Reddy, S.T.; Lee, S.; Parveen, M.; Grijalva, V.; Vergnes, L.; Pervin, S. Follistatin Targets Distinct Pathways to Promote Brown Adipocyte Characteristics in Brown and White Adipose Tissues. Endocrinology 2017, 158, 1217–1230. [Google Scholar] [CrossRef] [PubMed]
  72. Wu, H.; Ou, H.; Hung, H.; Su, Y.; Lu, F.; Wu, J.; Yang, Y.; Wu, C.; Chang, C. A novel hepatokine, HFREP1, plays a crucial role in the development of insulin resistance and type 2 diabetes. Diabetologia 2016, 59, 1732–1742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Jung, T.W.; Chung, Y.H.; Kim, H.; Abd El-Aty, A.M.; Jeong, J.H. Hyperlipidemia-induced hepassocin in the liver contributes to insulin resistance in skeletal muscle. Mol. Cell. Endocrinol. 2018, 470, 26–33. [Google Scholar] [CrossRef] [PubMed]
  74. Wu, H.; Lu, F.; Ou, H.; Su, Y.; Hung, H.; Wu, J.; Yang, Y.; Wu, C.; Chang, C. The role of hepassocin in the development of non-alcoholic fatty liver disease. J. Hepatol. 2013, 59, 1065–1072. [Google Scholar] [CrossRef]
  75. Wu, H.; Chen, S.; Fan, K.; Kuo, C.; Lin, S.; Wang, S.; Chang, C.; Li, H. Targeting fibrinogen-like protein 1 is a novel therapeutic strategy to combat obesity. FASEB J. 2020, 34, 2958–2967. [Google Scholar] [CrossRef]
  76. Kilicarslan, M.; de Weijer, B.A.; Simonyté Sjödin, K.; Aryal, P.; Ter Horst, K.W.; Cakir, H.; Romijn, J.A.; Ackermans, M.T.; Janssen, I.M.; Berends, F.J.; et al. RBP4 increases lipolysis in human adipocytes and is associated with increased lipolysis and hepatic insulin resistance in obese women. FASEB J. 2020, 34, 6099–6110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Graham, T.E.; Yang, Q.; Blüher, M.; Hammarstedt, A.; Ciaraldi, T.P.; Henry, R.R.; Wason, C.J.; Oberbach, A.; Jansson, P.; Smith, U.; et al. Retinol-binding protein 4 and insulin resistance in lean, obese, and diabetic subjects. N. Engl. J. Med. 2006, 354, 2552–2563. [Google Scholar] [CrossRef]
  78. Liu, Y.; Mu, D.; Chen, H.; Li, D.; Song, J.; Zhong, Y.; Xia, M. Retinol-Binding Protein 4 Induces Hepatic Mitochondrial Dysfunction and Promotes Hepatic Steatosis. J. Clin. Endocrinol. Metab. 2016, 101, 4338–4348. [Google Scholar] [CrossRef] [PubMed]
  79. Yang, Q.; Graham, T.E.; Mody, N.; Preitner, F.; Peroni, O.D.; Zabolotny, J.M.; Kotani, K.; Quadro, L.; Kahn, B.B. Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Nature 2005, 436, 356–362. [Google Scholar] [CrossRef] [PubMed]
  80. Norseen, J.; Hosooka, T.; Hammarstedt, A.; Yore, M.M.; Kant, S.; Aryal, P.; Kiernan, U.A.; Phillips, D.A.; Maruyama, H.; Kraus, B.J.; et al. Retinol-binding protein 4 inhibits insulin signaling in adipocytes by inducing proinflammatory cytokines in macrophages through a c-Jun N-terminal kinase- and toll-like receptor 4-dependent and retinol-independent mechanism. Mol. Cell. Biol. 2012, 32, 2010–2019. [Google Scholar] [CrossRef] [Green Version]
  81. Moraes-Vieira, P.M.; Yore, M.M.; Dwyer, P.M.; Syed, I.; Aryal, P.; Kahn, B.B. RBP4 activates antigen-presenting cells, leading to adipose tissue inflammation and systemic insulin resistance. Cell. Metab. 2014, 19, 512–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Fedders, R.; Muenzner, M.; Weber, P.; Sommerfeld, M.; Knauer, M.; Kedziora, S.; Kast, N.; Heidenreich, S.; Raila, J.; Weger, S.; et al. Liver-secreted RBP4 does not impair glucose homeostasis in mice. J. Biol. Chem. 2018, 293, 15269–15276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Montgomery, M.K.; Bayliss, J.; Devereux, C.; Bezawork-Geleta, A.; Roberts, D.; Huang, C.; Schittenhelm, R.B.; Ryan, A.; Townley, S.L.; Selth, L.A.; et al. SMOC1 is a glucose-responsive hepatokine and therapeutic target for glycemic control. Sci. Transl. Med. 2020, 12, eaaz8048. [Google Scholar] [CrossRef] [PubMed]
  84. Baek, S.J.; Eling, T. Growth differentiation factor 15 (GDF15): A survival protein with therapeutic potential in metabolic diseases. Pharmacol. Ther. 2019, 198, 46–58. [Google Scholar] [CrossRef] [PubMed]
  85. Chrysovergis, K.; Wang, X.; Kosak, J.; Lee, S.H.; Kim, J.S.; Foley, J.F.; Travlos, G.; Singh, S.; Baek, S.J.; Eling, T.E. NAG-1/GDF-15 prevents obesity by increasing thermogenesis, lipolysis and oxidative metabolism. Int. J. Obes. 2014, 38, 1555–1564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Chung, H.K.; Ryu, D.; Kim, K.S.; Chang, J.Y.; Kim, Y.K.; Yi, H.; Kang, S.G.; Choi, M.J.; Lee, S.E.; Jung, S.; et al. Growth differentiation factor 15 is a myomitokine governing systemic energy homeostasis. J. Cell Biol. 2017, 216, 149–165. [Google Scholar] [CrossRef] [PubMed]
  87. Mullican, S.E.; Lin-Schmidt, X.; Chin, C.; Chavez, J.A.; Furman, J.L.; Armstrong, A.A.; Beck, S.C.; South, V.J.; Dinh, T.Q.; Cash-Mason, T.D.; et al. GFRAL is the receptor for GDF15 and the ligand promotes weight loss in mice and nonhuman primates. Nat. Med. 2017, 23, 1150–1157. [Google Scholar] [CrossRef]
  88. Yang, L.; Chang, C.; Sun, Z.; Madsen, D.; Zhu, H.; Padkjær, S.B.; Wu, X.; Huang, T.; Hultman, K.; Paulsen, S.J.; et al. GFRAL is the receptor for GDF15 and is required for the anti-obesity effects of the ligand. Nat. Med. 2017, 23, 1158–1166. [Google Scholar] [CrossRef]
  89. Hsu, J.; Crawley, S.; Chen, M.; Ayupova, D.A.; Lindhout, D.A.; Higbee, J.; Kutach, A.; Joo, W.; Gao, Z.; Fu, D.; et al. Non-homeostatic body weight regulation through a brainstem-restricted receptor for GDF15. Nature 2017, 550, 255–259. [Google Scholar] [CrossRef]
  90. Borner, T.; Shaulson, E.D.; Ghidewon, M.Y.; Barnett, A.B.; Horn, C.C.; Doyle, R.P.; Grill, H.J.; Hayes, M.R.; De Jonghe, B.C. GDF15 Induces Anorexia through Nausea and Emesis. Cell. Metab. 2020, 31, 351–362. [Google Scholar] [CrossRef]
  91. Milovanovic, T.; Pantic, I.; Dragasevic, S.; Lugonja, S.; Dumic, I.; Rajilic-Stojanovic, M. The Interrelationship among Non-Alcoholic Fatty Liver Disease, Colonic Diverticulosis and Metabolic Syndrome. J. Gastrointest. Liver Dis. 2021, 30, 274–282. [Google Scholar] [CrossRef] [PubMed]
  92. Seppälä-Lindroos, A.; Vehkavaara, S.; Häkkinen, A.; Goto, T.; Westerbacka, J.; Sovijärvi, A.; Halavaara, J.; Yki-Järvinen, H. Fat accumulation in the liver is associated with defects in insulin suppression of glucose production and serum free fatty acids independent of obesity in normal men. J. Clin. Endocrinol. Metab. 2002, 87, 3023–3028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Korenblat, K.M.; Fabbrini, E.; Mohammed, B.S.; Klein, S. Liver, muscle, and adipose tissue insulin action is directly related to intrahepatic triglyceride content in obese subjects. Gastroenterology 2008, 134, 1369–1375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Bugianesi, E.; Gastaldelli, A.; Vanni, E.; Gambino, R.; Cassader, M.; Baldi, S.; Ponti, V.; Pagano, G.; Ferrannini, E.; Rizzetto, M. Insulin resistance in non-diabetic patients with non-alcoholic fatty liver disease: Sites and mechanisms. Diabetologia 2005, 48, 634–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Xu, H.; Barnes, G.T.; Yang, Q.; Tan, G.; Yang, D.; Chou, C.J.; Sole, J.; Nichols, A.; Ross, J.S.; Tartaglia, L.A.; et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J. Clin. Investig. 2003, 112, 1821–1830. [Google Scholar] [CrossRef] [PubMed]
  96. Kirpich, I.A.; Gobejishvili, L.N.; Bon Homme, M.; Waigel, S.; Cave, M.; Arteel, G.; Barve, S.S.; McClain, C.J.; Deaciuc, I.V. Integrated hepatic transcriptome and proteome analysis of mice with high-fat diet-induced nonalcoholic fatty liver disease. J. Nutr. Biochem. 2011, 22, 38–45. [Google Scholar] [CrossRef] [Green Version]
  97. Zhang, X.; Yang, J.; Guo, Y.; Ye, H.; Yu, C.; Xu, C.; Xu, L.; Wu, S.; Sun, W.; Wei, H.; et al. Functional proteomic analysis of nonalcoholic fatty liver disease in rat models: Enoyl-coenzyme a hydratase down-regulation exacerbates hepatic steatosis. Hepatology 2010, 51, 1190–1199. [Google Scholar] [CrossRef]
  98. Younossi, Z.M.; Baranova, A.; Ziegler, K.; Del Giacco, L.; Schlauch, K.; Born, T.L.; Elariny, H.; Gorreta, F.; VanMeter, A.; Younoszai, A.; et al. A genomic and proteomic study of the spectrum of nonalcoholic fatty liver disease. Hepatology 2005, 42, 665–674. [Google Scholar] [CrossRef] [PubMed]
  99. Kaur, P.; Rizk, N.M.; Ibrahim, S.; Younes, N.; Uppal, A.; Dennis, K.; Karve, T.; Blakeslee, K.; Kwagyan, J.; Zirie, M.; et al. iTRAQ-based quantitative protein expression profiling and MRM verification of markers in type 2 diabetes. J. Proteome Res. 2012, 11, 5527–5539. [Google Scholar] [CrossRef] [PubMed]
  100. Fu, S.; Fan, J.; Blanco, J.; Gimenez-Cassina, A.; Danial, N.N.; Watkins, S.M.; Hotamisligil, G.S. Polysome profiling in liver identifies dynamic regulation of endoplasmic reticulum translatome by obesity and fasting. PLoS Genet. 2012, 8, e1002902. [Google Scholar] [CrossRef] [PubMed]
  101. Santulli, G. Angiopoietin-like proteins: A comprehensive look. Front. Endocrinol. 2014, 5, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Le Jan, S.; Amy, C.; Cazes, A.; Monnot, C.; Lamandé, N.; Favier, J.; Philippe, J.; Sibony, M.; Gasc, J.; Corvol, P.; et al. Angiopoietin-like 4 is a proangiogenic factor produced during ischemia and in conventional renal cell carcinoma. Am. J. Pathol. 2003, 162, 1521–1528. [Google Scholar] [CrossRef] [Green Version]
  103. Camenisch, G.; Pisabarro, M.T.; Sherman, D.; Kowalski, J.; Nagel, M.; Hass, P.; Xie, M.; Gurney, A.; Bodary, S.; Liang, X.H.; et al. ANGPTL3 stimulates endothelial cell adhesion and migration via integrin alpha vbeta 3 and induces blood vessel formation in vivo. J. Biol. Chem. 2002, 277, 17281–17290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Oike, Y.; Yasunaga, K.; Suda, T. Angiopoietin-related/angiopoietin-like proteins regulate angiogenesis. Int. J. Hematol. 2004, 80, 21–28. [Google Scholar] [CrossRef] [PubMed]
  105. Kim, I.; Kwak, H.J.; Ahn, J.E.; So, J.N.; Liu, M.; Koh, K.N.; Koh, G.Y. Molecular cloning and characterization of a novel angiopoietin family protein, angiopoietin-3. FEBS Lett. 1999, 443, 353–356. [Google Scholar] [CrossRef] [Green Version]
  106. Ward, N.L.; Dumont, D.J. The angiopoietins and Tie2/Tek: Adding to the complexity of cardiovascular development. Semin. Cell. Dev. Biol. 2002, 13, 19–27. [Google Scholar] [CrossRef]
  107. Zheng, J.; Umikawa, M.; Cui, C.; Li, J.; Chen, X.; Zhang, C.; Huynh, H.; Hyunh, H.; Kang, X.; Silvany, R.; et al. Inhibitory receptors bind ANGPTLs and support blood stem cells and leukaemia development. Nature 2012, 485, 656–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Cinkajzlová, A.; Mráz, M.; Lacinová, Z.; Kloučková, J.; Kaválková, P.; Kratochvílová, H.; Trachta, P.; Křížová, J.; Haluzíková, D.; Škrha, J.; et al. Angiopoietin-like protein 3 and 4 in obesity, type 2 diabetes mellitus, and malnutrition: The effect of weight reduction and realimentation. Nutr. Diabetes 2018, 8, 21. [Google Scholar] [CrossRef] [PubMed]
  109. Sun, T.; Zhan, W.; Wei, L.; Xu, Z.; Fan, L.; Zhuo, Y.; Wang, C.; Zhang, J. Circulating ANGPTL3 and ANGPTL4 levels predict coronary artery atherosclerosis severity. Lipids Health Dis. 2021, 20, 154. [Google Scholar] [CrossRef] [PubMed]
  110. Graham, M.J.; Lee, R.G.; Brandt, T.A.; Tai, L.; Fu, W.; Peralta, R.; Yu, R.; Hurh, E.; Paz, E.; McEvoy, B.W.; et al. Cardiovascular and Metabolic Effects of ANGPTL3 Antisense Oligonucleotides. N. Engl. J. Med. 2017, 377, 222–232. [Google Scholar] [CrossRef]
  111. Fukami, H.; Morinaga, J.; Nakagami, H.; Hayashi, H.; Okadome, Y.; Matsunaga, E.; Kadomatsu, T.; Horiguchi, H.; Sato, M.; Sugizaki, T.; et al. Vaccine targeting ANGPTL3 ameliorates dyslipidemia and associated diseases in mouse models of obese dyslipidemia and familial hypercholesterolemia. Cell Rep. Med. 2021, 2, 100446. [Google Scholar] [CrossRef]
  112. Hurt-Camejo, E. ANGPTL3, PCSK9, and statin therapy drive remarkable reductions in hyperlipidemia and atherosclerosis in a mouse model. J. Lipid Res. 2020, 61, 272–274. [Google Scholar] [CrossRef] [PubMed]
  113. Raal, F.J.; Rosenson, R.S.; Reeskamp, L.F.; Hovingh, G.K.; Kastelein, J.J.P.; Rubba, P.; Ali, S.; Banerjee, P.; Chan, K.; Gipe, D.A.; et al. Evinacumab for Homozygous Familial Hypercholesterolemia. N. Engl. J. Med. 2020, 383, 711–720. [Google Scholar] [CrossRef] [PubMed]
  114. Ahmad, Z.; Banerjee, P.; Hamon, S.; Chan, K.; Bouzelmat, A.; Sasiela, W.J.; Pordy, R.; Mellis, S.; Dansky, H.; Gipe, D.A.; et al. Inhibition of Angiopoietin-Like Protein 3 with a Monoclonal Antibody Reduces Triglycerides in Hypertriglyceridemia. Circulation 2019, 140, 470–486. [Google Scholar] [CrossRef] [PubMed]
  115. Gastaldelli, A.; Stefan, N.; Häring, H. Liver-targeting drugs and their effect on blood glucose and hepatic lipids. Diabetologia 2021, 64, 1461–1479. [Google Scholar] [CrossRef] [PubMed]
  116. Yoon, J.C.; Chickering, T.W.; Rosen, E.D.; Dussault, B.; Qin, Y.; Soukas, A.; Friedman, J.M.; Holmes, W.E.; Spiegelman, B.M. Peroxisome proliferator-activated receptor gamma target gene encoding a novel angiopoietin-related protein associated with adipose differentiation. Mol. Cell. Biol. 2000, 20, 5343–5349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Kersten, S.; Mandard, S.; Tan, N.S.; Escher, P.; Metzger, D.; Chambon, P.; Gonzalez, F.J.; Desvergne, B.; Wahli, W. Characterization of the fasting-induced adipose factor FIAF, a novel peroxisome proliferator-activated receptor target gene. J. Biol. Chem. 2000, 275, 28488–28493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Zandbergen, F.; Van Dijk, S.; Müller, M.; Kersten, S. Fasting-induced adipose factor/angiopoietin–like protein 4: A potential target for dyslipidemia? Future Lipidol. 2006, 1, 227–236. [Google Scholar] [CrossRef]
  119. Cushing, E.M.; Chi, X.; Sylvers, K.L.; Shetty, S.K.; Potthoff, M.J.; Davies, B.S.J. Angiopoietin-like 4 directs uptake of dietary fat away from adipose during fasting. Mol. Metab. 2017, 6, 809–818. [Google Scholar] [CrossRef]
  120. Sun, Y.; Long, J.; Zhou, Y. Angiopoietin-like 4 promotes melanoma cell invasion and survival through aldolase A. Oncol. Lett. 2014, 8, 211–217. [Google Scholar] [CrossRef] [Green Version]
  121. Li, L.; Chong, H.C.; Ng, S.Y.; Kwok, K.W.; Teo, Z.; Tan, E.H.P.; Choo, C.C.; Seet, J.E.; Choi, H.W.; Buist, M.L.; et al. Angiopoietin-like 4 Increases Pulmonary Tissue Leakiness and Damage during Influenza Pneumonia. Cell Rep. 2015, 10, 654–663. [Google Scholar] [CrossRef] [Green Version]
  122. Tan, M.J.; Teo, Z.; Sng, M.K.; Zhu, P.; Tan, N.S. Emerging roles of angiopoietin-like 4 in human cancer. Mol. Cancer Res. 2012, 10, 677–688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Zhu, P.; Tan, M.J.; Huang, R.; Tan, C.K.; Chong, H.C.; Pal, M.; Lam, C.R.I.; Boukamp, P.; Pan, J.Y.; Tan, S.H.; et al. Angiopoietin-like 4 protein elevates the prosurvival intracellular O2(-):H2O2 ratio and confers anoikis resistance to tumors. Cancer Cell 2011, 19, 401–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Aryal, B.; Singh, A.K.; Zhang, X.; Varela, L.; Rotllan, N.; Goedeke, L.; Chaube, B.; Camporez, J.; Vatner, D.F.; Horvath, T.L.; et al. Absence of ANGPTL4 in adipose tissue improves glucose tolerance and attenuates atherogenesis. JCI Insight 2018, 3, e97918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Lichtenstein, L.; Mattijssen, F.; de Wit, N.J.; Georgiadi, A.; Hooiveld, G.J.; van der Meer, R.; He, Y.; Qi, L.; Köster, A.; Tamsma, J.T.; et al. Angptl4 protects against severe proinflammatory effects of saturated fat by inhibiting fatty acid uptake into mesenteric lymph node macrophages. Cell Metab. 2010, 12, 580–592. [Google Scholar] [CrossRef] [Green Version]
  126. Dewey, F.E.; Gusarova, V.; O’Dushlaine, C.; Gottesman, O.; Trejos, J.; Hunt, C.; Van Hout, C.V.; Habegger, L.; Buckler, D.; Lai, K.V.; et al. Inactivating Variants in ANGPTL4 and Risk of Coronary Artery Disease. N. Engl. J. Med. 2016, 374, 1123–1133. [Google Scholar] [CrossRef]
  127. Tjeerdema, N.; Georgiadi, A.; Jonker, J.T.; van Glabbeek, M.; Alizadeh Dehnavi, R.; Tamsma, J.T.; Smit, J.W.A.; Kersten, S.; Rensen, P.C.N. Inflammation increases plasma angiopoietin-like protein 4 in patients with the metabolic syndrome and type 2 diabetes. BMJ Open Diabetes Res. Care 2014, 2, e000034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Abu-Farha, M.; Al-Khairi, I.; Cherian, P.; Chandy, B.; Sriraman, D.; Alhubail, A.; Al-Refaei, F.; AlTerki, A.; Abubaker, J. Increased ANGPTL3, 4 and ANGPTL8/betatrophin expression levels in obesity and T2D. Lipids Health Dis. 2016, 15, 181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Barja-Fernandez, S.; Moreno-Navarrete, J.M.; Folgueira, C.; Xifra, G.; Sabater, M.; Castelao, C.; FernØ, J.; Leis, R.; Diéguez, C.; Casanueva, F.F.; et al. Plasma ANGPTL-4 is Associated with Obesity and Glucose Tolerance: Cross-Sectional and Longitudinal Findings. Mol. Nutr. Food Res. 2018, 62, e1800060. [Google Scholar] [CrossRef]
  130. Mehta, N.; Qamar, A.; Qu, L.; Qasim, A.N.; Mehta, N.N.; Reilly, M.P.; Rader, D.J. Differential association of plasma angiopoietin-like proteins 3 and 4 with lipid and metabolic traits. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1057–1063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Mandard, S.; Zandbergen, F.; van Straten, E.; Wahli, W.; Kuipers, F.; Müller, M.; Kersten, S. The fasting-induced adipose factor/angiopoietin-like protein 4 is physically associated with lipoproteins and governs plasma lipid levels and adiposity. J. Biol. Chem. 2006, 281, 934–944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Singh, A.K.; Chaube, B.; Zhang, X.; Sun, J.; Citrin, K.M.; Canfrán-Duque, A.; Aryal, B.; Rotllan, N.; Varela, L.; Lee, R.G.; et al. Hepatocyte-specific suppression of ANGPTL4 improves obesity-associated diabetes and mitigates atherosclerosis in mice. J. Clin. Investig. 2021, 131, e140989. [Google Scholar] [CrossRef] [PubMed]
  133. Oike, Y.; Akao, M.; Yasunaga, K.; Yamauchi, T.; Morisada, T.; Ito, Y.; Urano, T.; Kimura, Y.; Kubota, Y.; Maekawa, H.; et al. Angiopoietin-related growth factor antagonizes obesity and insulin resistance. Nat. Med. 2005, 11, 400–408. [Google Scholar] [CrossRef]
  134. Kadomatsu, T.; Tabata, M.; Oike, Y. Angiopoietin-like proteins: Emerging targets for treatment of obesity and related metabolic diseases. FEBS J. 2011, 278, 559–564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Namkung, J.; Sohn, J.H.; Chang, J.S.; Park, S.W.; Kim, J.Y.; Koh, S.B.; Kong, I.D.; Park, K.S. Increased Serum Angiopoietin-Like 6 Ahead of Metabolic Syndrome in a Prospective Cohort Study. Diabetes Metab. J. 2019, 43, 521–529. [Google Scholar] [CrossRef] [PubMed]
  136. Stepan, H.; Ebert, T.; Schrey, S.; Reisenbüchler, C.; Stein, S.; Lossner, U.; Bluher, M.; Stumvoll, M.; Kratzsch, J.; Faber, R.; et al. Serum levels of angiopoietin-related growth factor are increased in preeclampsia. Am. J. Hypertens. 2009, 22, 314–318. [Google Scholar] [CrossRef] [Green Version]
  137. Ebert, T.; Kralisch, S.; Loessner, U.; Jessnitzer, B.; Stumvoll, M.; Fasshauer, M.; Tönjes, A. Relationship between serum levels of angiopoietin-related growth factor and metabolic risk factors. Horm. Metab. Res. 2014, 46, 685–690. [Google Scholar] [CrossRef]
  138. Ebert, T.; Bachmann, A.; Lössner, U.; Kratzsch, J.; Blüher, M.; Stumvoll, M.; Fasshauer, M. Serum levels of angiopoietin-related growth factor in diabetes mellitus and chronic hemodialysis. Metabolism 2009, 58, 547–551. [Google Scholar] [CrossRef]
  139. Kim, M.; Namkung, J.; Chang, J.S.; Kim, S.; Park, K.; Kong, I.D. Leptin regulates the expression of angiopoietin-like 6. Biochem. Biophys. Res. Commun. 2018, 502, 397–402. [Google Scholar] [CrossRef] [PubMed]
  140. Quagliarini, F.; Wang, Y.; Kozlitina, J.; Grishin, N.V.; Hyde, R.; Boerwinkle, E.; Valenzuela, D.M.; Murphy, A.J.; Cohen, J.C.; Hobbs, H.H. Atypical angiopoietin-like protein that regulates ANGPTL3. Proc. Natl. Acad. Sci. USA 2012, 109, 19751–19756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Kovrov, O.; Kristensen, K.K.; Larsson, E.; Ploug, M.; Olivecrona, G. On the mechanism of angiopoietin-like protein 8 for control of lipoprotein lipase activity. J. Lipid Res. 2019, 60, 783–793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Yi, P.; Park, J.; Melton, D.A. Betatrophin: A hormone that controls pancreatic β cell proliferation. Cell 2013, 153, 747–758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Cox, A.R.; Lam, C.J.; Bonnyman, C.W.; Chavez, J.; Rios, J.S.; Kushner, J.A. Angiopoietin-like protein 8 (ANGPTL8)/betatrophin overexpression does not increase beta cell proliferation in mice. Diabetologia 2015, 58, 1523–1531. [Google Scholar] [CrossRef] [Green Version]
  144. Gusarova, V.; Alexa, C.A.; Na, E.; Stevis, P.E.; Xin, Y.; Bonner-Weir, S.; Cohen, J.C.; Hobbs, H.H.; Murphy, A.J.; Yancopoulos, G.D.; et al. ANGPTL8/betatrophin does not control pancreatic beta cell expansion. Cell 2014, 159, 691–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Wang, Y.; Quagliarini, F.; Gusarova, V.; Gromada, J.; Valenzuela, D.M.; Cohen, J.C.; Hobbs, H.H. Mice lacking ANGPTL8 (Betatrophin) manifest disrupted triglyceride metabolism without impaired glucose homeostasis. Proc. Natl. Acad. Sci. USA 2013, 110, 16109–16114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Zhao, Z.; Deng, X.; Jia, J.; Zhao, L.; Wang, C.; Cai, Z.; Guo, C.; Yang, L.; Wang, D.; Ma, S.; et al. Angiopoietin-like protein 8 (betatrophin) inhibits hepatic gluconeogenesis through PI3K/Akt signaling pathway in diabetic mice. Metabolism 2021, 126, 154921. [Google Scholar] [CrossRef]
  147. Zhang, Y.; Guo, X.; Yan, W.; Chen, Y.; Ke, M.; Cheng, C.; Zhu, X.; Xue, W.; Zhou, Q.; Zheng, L.; et al. ANGPTL8 negatively regulates NF-κB activation by facilitating selective autophagic degradation of IKKγ. Nat. Commun. 2017, 8, 2164. [Google Scholar] [CrossRef] [PubMed]
  148. Vatner, D.F.; Goedeke, L.; Camporez, J.G.; Lyu, K.; Nasiri, A.R.; Zhang, D.; Bhanot, S.; Murray, S.F.; Still, C.D.; Gerhard, G.S.; et al. Angptl8 antisense oligonucleotide improves adipose lipid metabolism and prevents diet-induced NAFLD and hepatic insulin resistance in rodents. Diabetologia 2018, 61, 1435–1446. [Google Scholar] [CrossRef] [Green Version]
  149. Wang, C.; Tong, Y.; Wen, Y.; Cai, J.; Guo, H.; Huang, L.; Xu, M.; Feng, M.; Chen, X.; Zhang, J.; et al. Hepatocellular Carcinoma-Associated Protein TD26 Interacts and Enhances Sterol Regulatory Element-Binding Protein 1 Activity to Promote Tumor Cell Proliferation and Growth. Hepatology 2018, 68, 1833–1850. [Google Scholar] [CrossRef] [Green Version]
  150. Li, J.; Li, L.; Guo, D.; Li, S.; Zeng, Y.; Liu, C.; Fu, R.; Huang, M.; Xie, W. Triglyceride metabolism and angiopoietin-like proteins in lipoprotein lipase regulation. Clin. Chim. Acta 2020, 503, 19–34. [Google Scholar] [CrossRef] [PubMed]
  151. Dang, F.; Wu, R.; Wang, P.; Wu, Y.; Azam, M.S.; Xu, Q.; Chen, Y.; Liu, Y. Fasting and Feeding Signals Control the Oscillatory Expression of Angptl8 to Modulate Lipid Metabolism. Sci. Rep. 2016, 6, 36926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Luo, M.; Peng, D. ANGPTL8: An Important Regulator in Metabolic Disorders. Front. Endocrinol. 2018, 9, 169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Gómez-Ambrosi, J.; Pascual, E.; Catalán, V.; Rodríguez, A.; Ramírez, B.; Silva, C.; Gil, M.J.; Salvador, J.; Frühbeck, G. Circulating betatrophin concentrations are decreased in human obesity and type 2 diabetes. J. Clin. Endocrinol. Metab. 2014, 99, 2004. [Google Scholar] [CrossRef] [Green Version]
  154. Barja-Fernández, S.; Folgueira, C.; Seoane, L.M.; Casanueva, F.F.; Dieguez, C.; Castelao, C.; Agüera, Z.; Baños, R.; Botella, C.; de la Torre, R.; et al. Circulating Betatrophin Levels Are Increased in Anorexia and Decreased in Morbidly Obese Women. J. Clin. Endocrinol. Metab. 2015, 100, 1188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Tuhan, H.; Abacı, A.; Anık, A.; Çatlı, G.; Küme, T.; Çalan, Ö.G.; Acar, S.; Böber, E. Circulating betatrophin concentration is negatively correlated with insulin resistance in obese children and adolescents. Diabetes Res. Clin. Pract. 2016, 114, 37–42. [Google Scholar] [CrossRef] [PubMed]
  156. Morinaga, J.; Zhao, J.; Endo, M.; Kadomatsu, T.; Miyata, K.; Sugizaki, T.; Okadome, Y.; Tian, Z.; Horiguchi, H.; Miyashita, K.; et al. Association of circulating ANGPTL 3, 4, and 8 levels with medical status in a population undergoing routine medical checkups: A cross-sectional study. PLoS ONE 2018, 13, e0193731. [Google Scholar] [CrossRef] [PubMed]
  157. Olivier, E.; Soury, E.; Ruminy, P.; Husson, A.; Parmentier, F.; Daveau, M.; Salier, J.P. Fetuin-B, a second member of the fetuin family in mammals. Biochem. J. 2000, 350 Pt 2, 589–597. [Google Scholar] [CrossRef] [PubMed]
  158. Jialal, I.; Pahwa, R. Fetuin-A is also an adipokine. Lipids Health Dis. 2019, 18, 73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Pérez-Sotelo, D.; Roca-Rivada, A.; Larrosa-García, M.; Castelao, C.; Baamonde, I.; Baltar, J.; Crujeiras, A.B.; Seoane, L.M.; Casanueva, F.F.; Pardo, M. Visceral and subcutaneous adipose tissue express and secrete functional alpha2hsglycoprotein (Fetuin A) especially in obesity. Endocrine 2017, 55, 435–446. [Google Scholar] [CrossRef]
  160. Jialal, I.; Devaraj, S.; Bettaieb, A.; Haj, F.; Adams-Huet, B. Increased adipose tissue secretion of Fetuin-A, lipopolysaccharide-binding protein and high-mobility group box protein 1 in metabolic syndrome. Atherosclerosis 2015, 241, 130–137. [Google Scholar] [CrossRef]
  161. Denecke, B.; Gräber, S.; Schäfer, C.; Heiss, A.; Wöltje, M.; Jahnen-Dechent, W. Tissue distribution and activity testing suggest a similar but not identical function of fetuin-B and fetuin-A. Biochem. J. 2003, 376, 135–145. [Google Scholar] [CrossRef] [PubMed]
  162. Haukeland, J.W.; Dahl, T.B.; Yndestad, A.; Gladhaug, I.P.; Løberg, E.M.; Haaland, T.; Konopski, Z.; Wium, C.; Aasheim, E.T.; Johansen, O.E.; et al. Fetuin A in nonalcoholic fatty liver disease: In vivo and in vitro studies. Eur. J. Endocrinol. 2012, 166, 503–510. [Google Scholar] [CrossRef] [PubMed]
  163. Reinehr, T.; Roth, C.L. Fetuin-A and its relation to metabolic syndrome and fatty liver disease in obese children before and after weight loss. J. Clin. Endocrinol. Metab. 2008, 93, 4479–4485. [Google Scholar] [CrossRef] [Green Version]
  164. Stefan, N.; Hennige, A.M.; Staiger, H.; Machann, J.; Schick, F.; Kröber, S.M.; Machicao, F.; Fritsche, A.; Häring, H. Alpha2-Heremans-Schmid glycoprotein/fetuin-A is associated with insulin resistance and fat accumulation in the liver in humans. Diabetes Care 2006, 29, 853–857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Stefan, N.; Fritsche, A.; Weikert, C.; Boeing, H.; Joost, H.; Häring, H.; Schulze, M.B. Plasma fetuin-A levels and the risk of type 2 diabetes. Diabetes 2008, 57, 2762–2767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Hennige, A.M.; Staiger, H.; Wicke, C.; Machicao, F.; Fritsche, A.; Häring, H.; Stefan, N. Fetuin-A induces cytokine expression and suppresses adiponectin production. PLoS ONE 2008, 3, e1765. [Google Scholar] [CrossRef] [Green Version]
  167. Dasgupta, S.; Bhattacharya, S.; Biswas, A.; Majumdar, S.S.; Mukhopadhyay, S.; Ray, S.; Bhattacharya, S. NF-kappaB mediates lipid-induced fetuin-A expression in hepatocytes that impairs adipocyte function effecting insulin resistance. Biochem. J. 2010, 429, 451–462. [Google Scholar] [CrossRef] [Green Version]
  168. Jung, T.W.; Youn, B.; Choi, H.Y.; Lee, S.Y.; Hong, H.C.; Yang, S.J.; Yoo, H.J.; Kim, B.; Baik, S.H.; Choi, K.M. Salsalate and adiponectin ameliorate hepatic steatosis by inhibition of the hepatokine fetuin-A. Biochem. Pharmacol. 2013, 86, 960–969. [Google Scholar] [CrossRef] [PubMed]
  169. Esteghamati, A.; Afarideh, M.; Feyzi, S.; Noshad, S.; Nakhjavani, M. Comparative effects of metformin and pioglitazone on fetuin-A and osteoprotegerin concentrations in patients with newly diagnosed diabetes: A randomized clinical trial. Diabetes Metab. Syndr. 2015, 9, 258–265. [Google Scholar] [CrossRef]
  170. Zhang, L.; Qu, X.; Sun, Z.; Zhang, Y. Effect of liraglutide therapy on serum fetuin A in patients with type 2 diabetes and non-alcoholic fatty liver disease. Clin. Res. Hepatol. Gastroenterol. 2020, 44, 674–680. [Google Scholar] [CrossRef]
  171. Zhu, J.; Wan, X.; Wang, Y.; Zhu, K.; Li, C.; Yu, C.; Li, Y. Serum fetuin B level increased in subjects of nonalcoholic fatty liver disease: A case-control study. Endocrine 2017, 56, 208–211. [Google Scholar] [CrossRef]
  172. Li, L.; Spranger, L.; Stobäus, N.; Beer, F.; Decker, A.; Wernicke, C.; Brachs, S.; Brachs, M.; Spranger, J.; Mai, K. Fetuin-B, a potential link of liver-adipose tissue cross talk during diet-induced weight loss-weight maintenance. Nutr. Diabetes 2021, 11, 31. [Google Scholar] [CrossRef] [PubMed]
  173. Ebert, T.; Linder, N.; Schaudinn, A.; Busse, H.; Berger, J.; Lichtinghagen, R.; Keim, V.; Wiegand, J.; Karlas, T. Association of fetuin B with markers of liver fibrosis in nonalcoholic fatty liver disease. Endocrine 2017, 58, 246–252. [Google Scholar] [CrossRef] [PubMed]
  174. Sato, M.; Kamada, Y.; Takeda, Y.; Kida, S.; Ohara, Y.; Fujii, H.; Akita, M.; Mizutani, K.; Yoshida, Y.; Yamada, M.; et al. Fetuin-A negatively correlates with liver and vascular fibrosis in nonalcoholic fatty liver disease subjects. Liver Int. 2015, 35, 925–935. [Google Scholar] [CrossRef]
  175. Peter, A.; Kovarova, M.; Staiger, H.; Machann, J.; Schick, F.; Königsrainer, A.; Königsrainer, I.; Schleicher, E.; Fritsche, A.; Häring, H.; et al. The hepatokines fetuin-A and fetuin-B are upregulated in the state of hepatic steatosis and may differently impact on glucose homeostasis in humans. Am. J. Physiol. Endocrinol. Metab. 2018, 314, E266–E273. [Google Scholar] [CrossRef] [PubMed]
  176. Staiger, H.; Keuper, M.; Berti, L.; Hrabe de Angelis, M.; Häring, H. Fibroblast Growth Factor 21-Metabolic Role in Mice and Men. Endocr. Rev. 2017, 38, 468–488. [Google Scholar] [CrossRef]
  177. Markan, K.R.; Naber, M.C.; Ameka, M.K.; Anderegg, M.D.; Mangelsdorf, D.J.; Kliewer, S.A.; Mohammadi, M.; Potthoff, M.J. Circulating FGF21 is liver derived and enhances glucose uptake during refeeding and overfeeding. Diabetes 2014, 63, 4057–4063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Mutanen, A.; Heikkilä, P.; Lohi, J.; Raivio, T.; Jalanko, H.; Pakarinen, M.P. Serum FGF21 increases with hepatic fat accumulation in pediatric onset intestinal failure. J. Hepatol. 2014, 60, 183–190. [Google Scholar] [CrossRef]
  179. Chavez, A.O.; Molina-Carrion, M.; Abdul-Ghani, M.A.; Folli, F.; Defronzo, R.A.; Tripathy, D. Circulating fibroblast growth factor-21 is elevated in impaired glucose tolerance and type 2 diabetes and correlates with muscle and hepatic insulin resistance. Diabetes Care 2009, 32, 1542–1546. [Google Scholar] [CrossRef] [Green Version]
  180. Zhang, X.; Yeung, D.C.Y.; Karpisek, M.; Stejskal, D.; Zhou, Z.; Liu, F.; Wong, R.L.C.; Chow, W.; Tso, A.W.K.; Lam, K.S.L.; et al. Serum FGF21 levels are increased in obesity and are independently associated with the metabolic syndrome in humans. Diabetes 2008, 57, 1246–1253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Flisiak-Jackiewicz, M.; Bobrus-Chociej, A.; Wasilewska, N.; Tarasow, E.; Wojtkowska, M.; Lebensztejn, D.M. Can hepatokines be regarded as novel non-invasive serum biomarkers of intrahepatic lipid content in obese children? Adv. Med. Sci. 2019, 64, 280–284. [Google Scholar] [CrossRef] [PubMed]
  182. Yang, M.; Xu, D.; Liu, Y.; Guo, X.; Li, W.; Guo, C.; Zhang, H.; Gao, Y.; Mao, Y.; Zhao, J. Combined Serum Biomarkers in Non-Invasive Diagnosis of Non-Alcoholic Steatohepatitis. PLoS ONE 2015, 10, e0131664. [Google Scholar] [CrossRef] [Green Version]
  183. Camporez, J.P.G.; Asrih, M.; Zhang, D.; Kahn, M.; Samuel, V.T.; Jurczak, M.J.; Jornayvaz, F.R. Hepatic insulin resistance and increased hepatic glucose production in mice lacking Fgf21. J. Endocrinol. 2015, 226, 207–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Ding, X.; Boney-Montoya, J.; Owen, B.M.; Bookout, A.L.; Coate, K.C.; Mangelsdorf, D.J.; Kliewer, S.A. βKlotho is required for fibroblast growth factor 21 effects on growth and metabolism. Cell Metab. 2012, 16, 387–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Inagaki, T.; Dutchak, P.; Zhao, G.; Ding, X.; Gautron, L.; Parameswara, V.; Li, Y.; Goetz, R.; Mohammadi, M.; Esser, V.; et al. Endocrine regulation of the fasting response by PPARalpha-mediated induction of fibroblast growth factor 21. Cell Metab. 2007, 5, 415–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Badman, M.K.; Pissios, P.; Kennedy, A.R.; Koukos, G.; Flier, J.S.; Maratos-Flier, E. Hepatic fibroblast growth factor 21 is regulated by PPARalpha and is a key mediator of hepatic lipid metabolism in ketotic states. Cell Metab. 2007, 5, 426–437. [Google Scholar] [CrossRef] [Green Version]
  187. Geng, L.; Liao, B.; Jin, L.; Huang, Z.; Triggle, C.R.; Ding, H.; Zhang, J.; Huang, Y.; Lin, Z.; Xu, A. Exercise Alleviates Obesity-Induced Metabolic Dysfunction via Enhancing FGF21 Sensitivity in Adipose Tissues. Cell Rep. 2019, 26, 2738–2752. [Google Scholar] [CrossRef] [Green Version]
  188. Kruse, R.; Vienberg, S.G.; Vind, B.F.; Andersen, B.; Højlund, K. Effects of insulin and exercise training on FGF21, its receptors and target genes in obesity and type 2 diabetes. Diabetologia 2017, 60, 2042–2051. [Google Scholar] [CrossRef] [Green Version]
  189. Tanimura, Y.; Aoi, W.; Takanami, Y.; Kawai, Y.; Mizushima, K.; Naito, Y.; Yoshikawa, T. Acute exercise increases fibroblast growth factor 21 in metabolic organs and circulation. Physiol. Rep. 2016, 4, e12828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Geng, L.; Lam, K.S.L.; Xu, A. The therapeutic potential of FGF21 in metabolic diseases: From bench to clinic. Nat. Rev. Endocrinol. 2020, 16, 654–667. [Google Scholar] [CrossRef]
  191. Kliewer, S.A.; Mangelsdorf, D.J. A Dozen Years of Discovery: Insights into the Physiology and Pharmacology of FGF21. Cell Metab. 2019, 29, 246–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Kaufman, A.; Abuqayyas, L.; Denney, W.S.; Tillman, E.J.; Rolph, T. AKR-001, an Fc-FGF21 Analog, Showed Sustained Pharmacodynamic Effects on Insulin Sensitivity and Lipid Metabolism in Type 2 Diabetes Patients. Cell Rep. Med. 2020, 1, 100057. [Google Scholar] [CrossRef]
  193. Harrison, S.A.; Ruane, P.J.; Freilich, B.L.; Neff, G.; Patil, R.; Behling, C.A.; Hu, C.; Fong, E.; de Temple, B.; Tillman, E.J.; et al. Efruxifermin in non-alcoholic steatohepatitis: A randomized, double-blind, placebo-controlled, phase 2a trial. Nat. Med. 2021, 27, 1262–1271. [Google Scholar] [CrossRef]
  194. Burk, R.F.; Hill, K.E. Selenoprotein P: An extracellular protein with unique physical characteristics and a role in selenium homeostasis. Annu. Rev. Nutr. 2005, 25, 215–235. [Google Scholar] [CrossRef] [PubMed]
  195. Hariharan, S.; Dharmaraj, S. Selenium and selenoproteins: It’s role in regulation of inflammation. Inflammopharmacology 2020, 28, 667–695. [Google Scholar] [CrossRef] [PubMed]
  196. Yang, S.J.; Hwang, S.Y.; Choi, H.Y.; Yoo, H.J.; Seo, J.A.; Kim, S.G.; Kim, N.H.; Baik, S.H.; Choi, D.S.; Choi, K.M. Serum selenoprotein P levels in patients with type 2 diabetes and prediabetes: Implications for insulin resistance, inflammation, and atherosclerosis. J. Clin. Endocrinol. Metab. 2011, 96, 1325. [Google Scholar] [CrossRef] [PubMed]
  197. Choi, H.Y.; Hwang, S.Y.; Lee, C.H.; Hong, H.C.; Yang, S.J.; Yoo, H.J.; Seo, J.A.; Kim, S.G.; Kim, N.H.; Baik, S.H.; et al. Increased selenoprotein p levels in subjects with visceral obesity and nonalcoholic Fatty liver disease. Diabetes Metab. J. 2013, 37, 63–71. [Google Scholar] [CrossRef] [Green Version]
  198. Misu, H.; Ishikura, K.; Kurita, S.; Takeshita, Y.; Ota, T.; Saito, Y.; Takahashi, K.; Kaneko, S.; Takamura, T. Inverse correlation between serum levels of selenoprotein P and adiponectin in patients with type 2 diabetes. PLoS ONE 2012, 7, e34952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Yamagoe, S.; Yamakawa, Y.; Matsuo, Y.; Minowada, J.; Mizuno, S.; Suzuki, K. Purification and primary amino acid sequence of a novel neutrophil chemotactic factor LECT2. Immunol. Lett. 1996, 52, 9–13. [Google Scholar] [CrossRef]
  200. Chikamoto, K.; Misu, H.; Takayama, H.; Kikuchi, A.; Ishii, K.; Lan, F.; Takata, N.; Tajima-Shirasaki, N.; Takeshita, Y.; Tsugane, H.; et al. Rapid response of the steatosis-sensing hepatokine LECT2 during diet-induced weight cycling in mice. Biochem. Biophys. Res. Commun. 2016, 478, 1310–1316. [Google Scholar] [CrossRef]
  201. Okumura, A.; Unoki-Kubota, H.; Matsushita, Y.; Shiga, T.; Moriyoshi, Y.; Yamagoe, S.; Kaburagi, Y. Increased serum leukocyte cell-derived chemotaxin 2 (LECT2) levels in obesity and fatty liver. Biosci. Trends 2013, 7, 276–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Yoo, H.J.; Hwang, S.Y.; Choi, J.; Lee, H.J.; Chung, H.S.; Seo, J.; Kim, S.G.; Kim, N.H.; Baik, S.H.; Choi, D.S.; et al. Association of leukocyte cell-derived chemotaxin 2 (LECT2) with NAFLD, metabolic syndrome, and atherosclerosis. PLoS ONE 2017, 12, e0174717. [Google Scholar] [CrossRef]
  203. Xu, M.; Xu, H.; Lin, Y.; Sun, X.; Wang, L.; Fang, Z.; Su, X.; Liang, X.; Hu, Y.; Liu, Z.; et al. LECT2, a Ligand for Tie1, Plays a Crucial Role in Liver Fibrogenesis. Cell 2019, 178, 1478–1492.e20. [Google Scholar] [CrossRef] [PubMed]
  204. Hwang, H.; Jung, T.W.; Kim, B.; Hong, H.C.; Seo, J.A.; Kim, S.G.; Kim, N.H.; Choi, K.M.; Choi, D.S.; Baik, S.H.; et al. A dipeptidyl peptidase-IV inhibitor improves hepatic steatosis and insulin resistance by AMPK-dependent and JNK-dependent inhibition of LECT2 expression. Biochem. Pharmacol. 2015, 98, 157–166. [Google Scholar] [CrossRef]
  205. Jung, T.W.; Chung, Y.H.; Kim, H.; Abd El-Aty, A.M.; Jeong, J.H. LECT2 promotes inflammation and insulin resistance in adipocytes via P38 pathways. J. Mol. Endocrinol. 2018, 61, 37–45. [Google Scholar] [CrossRef] [PubMed]
  206. Hansen, J.S.; Plomgaard, P. Circulating follistatin in relation to energy metabolism. Mol. Cell Endocrinol. 2016, 433, 87–93. [Google Scholar] [CrossRef]
  207. Yndestad, A.; Haukeland, J.W.; Dahl, T.B.; Bjøro, K.; Gladhaug, I.P.; Berge, C.; Damås, J.K.; Haaland, T.; Løberg, E.M.; Linnestad, P.; et al. A complex role of activin A in non-alcoholic fatty liver disease. Am. J. Gastroenterol. 2009, 104, 2196–2205. [Google Scholar] [CrossRef] [PubMed]
  208. Hansen, J.S.; Pedersen, B.K.; Xu, G.; Lehmann, R.; Weigert, C.; Plomgaard, P. Exercise-Induced Secretion of FGF21 and Follistatin Are Blocked by Pancreatic Clamp and Impaired in Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2016, 101, 2816–2825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Perakakis, N.; Kokkinos, A.; Peradze, N.; Tentolouris, N.; Ghaly, W.; Tsilingiris, D.; Alexandrou, A.; Mantzoros, C.S. Follistatins in glucose regulation in healthy and obese individuals. Diabetes Obes. Metab. 2019, 21, 683–690. [Google Scholar] [CrossRef]
  210. Hansen, J.S.; Rutti, S.; Arous, C.; Clemmesen, J.O.; Secher, N.H.; Drescher, A.; Gonelle-Gispert, C.; Halban, P.A.; Pedersen, B.K.; Weigert, C.; et al. Circulating Follistatin Is Liver-Derived and Regulated by the Glucagon-to-Insulin Ratio. J. Clin. Endocrinol. Metab. 2016, 101, 550–560. [Google Scholar] [CrossRef] [PubMed]
  211. Hara, H.; Uchida, S.; Yoshimura, H.; Aoki, M.; Toyoda, Y.; Sakai, Y.; Morimoto, S.; Fukamachi, H.; Shiokawa, K.; Hanada, K. Isolation and characterization of a novel liver-specific gene, hepassocin, upregulated during liver regeneration. Biochim. Biophys. Acta 2000, 1492, 31–44. [Google Scholar] [CrossRef]
  212. Yu, H.; Yu, M.; Li, C.; Zhan, Y.; Xu, W.; Li, Y.; Li, W.; Wang, Z.; Ge, C.; Yang, X. Specific expression and regulation of hepassocin in the liver and down-regulation of the correlation of HNF1alpha with decreased levels of hepassocin in human hepatocellular carcinoma. J. Biol. Chem. 2009, 284, 13335–13347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Li, C.; Cao, C.; Xu, W.; Cao, M.; Yang, F.; Dong, L.; Yu, M.; Zhan, Y.; Gao, Y.; Li, W.; et al. Recombinant human hepassocin stimulates proliferation of hepatocytes in vivo and improves survival in rats with fulminant hepatic failure. Gut 2010, 59, 817–826. [Google Scholar] [CrossRef]
  214. Ou, H.; Wu, H.; Lin, C.; Du, Y.; Hu, C.; Hung, H.; Wu, P.; Li, H.; Wang, S.; Chang, C. The Hepatic Protection Effects of Hepassocin in Hyperglycemic Crisis. J. Clin. Endocrinol. Metab. 2017, 102, 2407–2415. [Google Scholar] [CrossRef] [PubMed]
  215. Huang, R.; Li, C.; Du, Y.; Cheng, K.; Lin, C.; Hu, C.; Wu, J.S.; Chang, C.; Wu, H.; Ou, H. Discovery of a role of the novel hepatokine, hepassocin, in obesity. Biofactors 2020, 46, 100–105. [Google Scholar] [CrossRef] [PubMed]
  216. Steinhoff, J.S.; Lass, A.; Schupp, M. Biological Functions of RBP4 and Its Relevance for Human Diseases. Front. Physiol. 2021, 12, 659977. [Google Scholar] [CrossRef] [PubMed]
  217. Thompson, S.J.; Sargsyan, A.; Lee, S.; Yuen, J.J.; Cai, J.; Smalling, R.; Ghyselinck, N.; Mark, M.; Blaner, W.S.; Graham, T.E. Hepatocytes Are the Principal Source of Circulating RBP4 in Mice. Diabetes 2017, 66, 58–63. [Google Scholar] [CrossRef] [Green Version]
  218. Lee, S.; Yuen, J.J.; Jiang, H.; Kahn, B.B.; Blaner, W.S. Adipocyte-specific overexpression of retinol-binding protein 4 causes hepatic steatosis in mice. Hepatology 2016, 64, 1534–1546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Haider, D.G.; Schindler, K.; Prager, G.; Bohdjalian, A.; Luger, A.; Wolzt, M.; Ludvik, B. Serum retinol-binding protein 4 is reduced after weight loss in morbidly obese subjects. J. Clin. Endocrinol. Metab. 2007, 92, 1168–1171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Cai, H.; Lu, S.; Chen, Y.; Das Mbbs Mrcog, S.; Niu, Z.; Zhuo, G.; Lai, L.; Zhang, Z. Serum retinol binding protein 4 and galectin-3 binding protein as novel markers for postmenopausal nonalcoholic fatty liver disease. Clin. Biochem. 2018, 56, 95–101. [Google Scholar] [CrossRef]
  221. Wu, H.; Jia, W.; Bao, Y.; Lu, J.; Zhu, J.; Wang, R.; Chen, Y.; Xiang, K. Serum retinol binding protein 4 and nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus. Diabetes Res. Clin. Pract. 2008, 79, 185–190. [Google Scholar] [CrossRef]
  222. Perseghin, G.; Lattuada, G.; De Cobelli, F.; Esposito, A.; Belloni, E.; Canu, T.; Ragogna, F.; Scifo, P.; Del Maschio, A.; Luzi, L. Serum retinol-binding protein-4, leptin, and adiponectin concentrations are related to ectopic fat accumulation. J. Clin. Endocrinol. Metab. 2007, 92, 4883–4888. [Google Scholar] [CrossRef] [PubMed]
  223. Gersdorff, N.; Müller, M.; Schall, A.; Miosge, N. Secreted modular calcium-binding protein-1 localization during mouse embryogenesis. Histochem. Cell Biol. 2006, 126, 705–712. [Google Scholar] [CrossRef]
  224. Vannahme, C.; Smyth, N.; Miosge, N.; Gösling, S.; Frie, C.; Paulsson, M.; Maurer, P.; Hartmann, U. Characterization of SMOC-1, a novel modular calcium-binding protein in basement membranes. J. Biol. Chem. 2002, 277, 37977–37986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Choi, Y.; Lim, J.; Kim, K.M.; Acharya, B.; Cho, J.; Bae, Y.; Shin, H.; Kim, S.; Park, E.K. Secretome analysis of human BMSCs and identification of SMOC1 as an important ECM protein in osteoblast differentiation. J. Proteome Res. 2010, 9, 2946–2956. [Google Scholar] [CrossRef] [PubMed]
  226. Okada, I.; Hamanoue, H.; Terada, K.; Tohma, T.; Megarbane, A.; Chouery, E.; Abou-Ghoch, J.; Jalkh, N.; Cogulu, O.; Ozkinay, F.; et al. SMOC1 is essential for ocular and limb development in humans and mice. Am. J. Hum. Genet. 2011, 88, 30–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Awwad, K.; Hu, J.; Shi, L.; Mangels, N.; Abdel Malik, R.; Zippel, N.; Fisslthaler, B.; Eble, J.A.; Pfeilschifter, J.; Popp, R.; et al. Role of secreted modular calcium-binding protein 1 (SMOC1) in transforming growth factor β signalling and angiogenesis. Cardiovasc. Res. 2015, 106, 284–294. [Google Scholar] [CrossRef] [Green Version]
  228. Gao, Q.; Mok, H.; Zhuang, J. Secreted modular calcium-binding proteins in pathophysiological processes and embryonic development. Chin. Med. J. 2019, 132, 2476–2484. [Google Scholar] [CrossRef]
  229. Wang, Y.; Gu, J.; Du, A.; Zhang, S.; Deng, M.; Zhao, R.; Lu, Y.; Ji, Y.; Shao, Y.; Sun, W.; et al. SPARC-related modular calcium binding 1 regulates aortic valve calcification by disrupting BMPR-II/p-p38 signalling. Cardiovasc. Res. 2021, cvab107. [Google Scholar] [CrossRef]
  230. Delgado Lagos, F.; Elgheznawy, A.; Kyselova, A.; Meyer Zu Heringdorf, D.; Ratiu, C.; Randriamboavonjy, V.; Mann, A.W.; Fisslthaler, B.; Siragusa, M.; Fleming, I. Secreted modular calcium-binding protein 1 binds and activates thrombin to account for platelet hyperreactivity in diabetes. Blood 2021, 137, 1641–1651. [Google Scholar] [CrossRef] [PubMed]
  231. Emmerson, P.J.; Wang, F.; Du, Y.; Liu, Q.; Pickard, R.T.; Gonciarz, M.D.; Coskun, T.; Hamang, M.J.; Sindelar, D.K.; Ballman, K.K.; et al. The metabolic effects of GDF15 are mediated by the orphan receptor GFRAL. Nat. Med. 2017, 23, 1215–1219. [Google Scholar] [CrossRef] [PubMed]
  232. Adela, R.; Banerjee, S.K. GDF-15 as a Target and Biomarker for Diabetes and Cardiovascular Diseases: A Translational Prospective. J. Diabetes Res. 2015, 2015, 490842. [Google Scholar] [CrossRef] [PubMed]
  233. Klein, A.B.; Kleinert, M.; Richter, E.A.; Clemmensen, C. GDF15 in Appetite and Exercise: Essential Player or Coincidental Bystander? Endocrinology 2021, bqab242. [Google Scholar] [CrossRef] [PubMed]
  234. Johnen, H.; Lin, S.; Kuffner, T.; Brown, D.A.; Tsai, V.W.; Bauskin, A.R.; Wu, L.; Pankhurst, G.; Jiang, L.; Junankar, S.; et al. Tumor-induced anorexia and weight loss are mediated by the TGF-beta superfamily cytokine MIC-1. Nat. Med. 2007, 13, 1333–1340. [Google Scholar] [CrossRef]
  235. Tsai, V.W.; Manandhar, R.; Jørgensen, S.B.; Lee-Ng, K.K.M.; Zhang, H.P.; Marquis, C.P.; Jiang, L.; Husaini, Y.; Lin, S.; Sainsbury, A.; et al. The anorectic actions of the TGFβ cytokine MIC-1/GDF15 require an intact brainstem area postrema and nucleus of the solitary tract. PLoS ONE 2014, 9, e100370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Baek, S.J.; Okazaki, R.; Lee, S.; Martinez, J.; Kim, J.; Yamaguchi, K.; Mishina, Y.; Martin, D.W.; Shoieb, A.; McEntee, M.F.; et al. Nonsteroidal anti-inflammatory drug-activated gene-1 over expression in transgenic mice suppresses intestinal neoplasia. Gastroenterology 2006, 131, 1553–1560. [Google Scholar] [CrossRef]
  237. Maurice, J.; Manousou, P. Non-alcoholic fatty liver disease. Clin. Med. 2018, 18, 245–250. [Google Scholar] [CrossRef] [PubMed]
  238. Ennequin, G.; Sirvent, P.; Whitham, M. Role of exercise-induced hepatokines in metabolic disorders. Am. J. Physiol. Endocrinol. Metab. 2019, 317, E11–E24. [Google Scholar] [CrossRef] [PubMed]
  239. Lin, J.; Jiang, X.; Dong, M.; Liu, X.; Shen, Q.; Huang, Y.; Zhang, H.; Ye, R.; Zhou, H.; Yan, C.; et al. Hepatokine Pregnancy Zone Protein Governs the Diet-Induced Thermogenesis Through Activating Brown Adipose Tissue. Adv. Sci. 2021, 8, e2101991. [Google Scholar] [CrossRef]
  240. Wu, T.; Liu, Q.; Li, Y.; Li, H.; Chen, L.; Yang, X.; Tang, Q.; Pu, S.; Kuang, J.; Li, R.; et al. Feeding-induced hepatokine, Manf, ameliorates diet-induced obesity by promoting adipose browning via p38 MAPK pathway. J. Exp. Med. 2021, 218, e20201203. [Google Scholar] [CrossRef]
Figure 1. The role of ANGPTL family proteins in lipid metabolism in the liver and non-hepatic tissues. ANGPTL family proteins are hepatokines primarily produced and secreted by the liver, regulating various cellular processes, including lipid metabolism, within as well as outside the liver. The upper panel presents the transcription factors that regulate the gene expression of ANGPTL members under fasting or feeding/obesity/type 2 diabetes (T2DM) conditions. The transcription of ANGPTL3, ANGPTL8, and ANGPTL6 is upregulated by liver-X-receptor (LXR) activation or leptin signaling following feeding or under metabolic disorders, such as obesity and type 2 diabetes mellitus (T2DM), while peroxisome proliferator-activated receptor (PPAR) and glucocorticoid receptor (GR) suppress the transactivation of ANGPTL3 and ANGPTL8 under fasting conditions, respectively. Circulating ANGPTL proteins then serve as lipoprotein lipase (LPL) inhibitors in non-hepatic tissues, while liver-derived ANGPTL4 inhibits hepatic lipase, resulting in decreased triglyceride clearance. Therefore, ANGPTL proteins contribute to the development of hepatic steatosis by regulating lipogenesis and/or the inflammatory response, as depicted above. ACC: Acetyl-CoA carboxylase; CHOL: Cholesterol; FAS: Fatty acid synthase; FFA: Free fatty acid; IL6: Interleukin-6; NF-ĸB: Nuclear factor kappa-light-chain-enhancer of activated B cells; SREBP: Sterol regulatory-element binding protein; TF: Transcription factor; TG: Triglyceride; TNF: Tumor necrosis factor.
Figure 1. The role of ANGPTL family proteins in lipid metabolism in the liver and non-hepatic tissues. ANGPTL family proteins are hepatokines primarily produced and secreted by the liver, regulating various cellular processes, including lipid metabolism, within as well as outside the liver. The upper panel presents the transcription factors that regulate the gene expression of ANGPTL members under fasting or feeding/obesity/type 2 diabetes (T2DM) conditions. The transcription of ANGPTL3, ANGPTL8, and ANGPTL6 is upregulated by liver-X-receptor (LXR) activation or leptin signaling following feeding or under metabolic disorders, such as obesity and type 2 diabetes mellitus (T2DM), while peroxisome proliferator-activated receptor (PPAR) and glucocorticoid receptor (GR) suppress the transactivation of ANGPTL3 and ANGPTL8 under fasting conditions, respectively. Circulating ANGPTL proteins then serve as lipoprotein lipase (LPL) inhibitors in non-hepatic tissues, while liver-derived ANGPTL4 inhibits hepatic lipase, resulting in decreased triglyceride clearance. Therefore, ANGPTL proteins contribute to the development of hepatic steatosis by regulating lipogenesis and/or the inflammatory response, as depicted above. ACC: Acetyl-CoA carboxylase; CHOL: Cholesterol; FAS: Fatty acid synthase; FFA: Free fatty acid; IL6: Interleukin-6; NF-ĸB: Nuclear factor kappa-light-chain-enhancer of activated B cells; SREBP: Sterol regulatory-element binding protein; TF: Transcription factor; TG: Triglyceride; TNF: Tumor necrosis factor.
Biomedicines 09 01903 g001
Figure 2. The impact of hepatokines on NAFLD progression and their associated signaling pathways. Most of the hepatokine expression is upregulated under HFD feeding and/or hyperglycemia. FGF21 has been reported to decrease or increase under HFD feeding. Hepatokines, except for FGF21, impair insulin signaling in hepatocytes, as well as non-hepatic tissues in an endocrine manner, with Fetuin-A and LECT2 promoting the macrophage inflammatory response by upregulating pro-inflammatory cytokine production. Fetuins, LECT2, and Hepassocin promote hepatic steatosis via the upregulation of lipogenic genes, whereas FGF21 suppressed it. The regulatory mechanism of follistatin on lipogenesis is unknown.
Figure 2. The impact of hepatokines on NAFLD progression and their associated signaling pathways. Most of the hepatokine expression is upregulated under HFD feeding and/or hyperglycemia. FGF21 has been reported to decrease or increase under HFD feeding. Hepatokines, except for FGF21, impair insulin signaling in hepatocytes, as well as non-hepatic tissues in an endocrine manner, with Fetuin-A and LECT2 promoting the macrophage inflammatory response by upregulating pro-inflammatory cytokine production. Fetuins, LECT2, and Hepassocin promote hepatic steatosis via the upregulation of lipogenic genes, whereas FGF21 suppressed it. The regulatory mechanism of follistatin on lipogenesis is unknown.
Biomedicines 09 01903 g002
Table 1. Target organs or cells of hepatokines and their biological functions.
Table 1. Target organs or cells of hepatokines and their biological functions.
HepatokinesTarget Organs or CellsBiological FunctionsReference
ANGPTL3WAT, muscle, liverSuppressed LPL and endothelial lipase
Increased plasma TG and FFA
Increased VLDL-TG secretion (liver)
Increased uptake of VLDL-TGs (WAT)
Decreased glucose uptake (WAT)
Promoted lipogenesis and inflammatory response (liver)
[15,27,28,29,30,31,32,33]
ANGPTL4WAT, vascular endothelial cellsInhibited LPL activity
Increased plasma TG levels
NAFLDIncreased adipocyte lipolysis
Suppressed hepatic glucose production
[15,34,35,36,37,38,39]
ANGPTL6skeletal muscle, WAT, liverEnhanced insulin signaling (skeletal muscle)
Inhibited gluconeogenic pathway (liver)
Increased mitochondrial oxygen consumption (WAT)
[40,41,42,43]
ANGPTL8hepatocytes, adipocytesImproved insulin signaling and suppressed gluconeogenic gene expression (liver)
Suppressed lipolysis (hepatocyte, adipocyte)
Promoted lipogenesis (liver)
[44,45,46,47,48]
Fetuin-Aliver, WAT, skeletal muscle, monocytesBlocked insulin signaling through inhibition of insulin receptor tyrosine kinase (liver, WAT, skeletal muscle)
Provoked inflammatory response (monocytes, adipocytes)
Inhibited adiponectin production
[13,15,49,50,51,52,53,54,55]
Fetuin-Bhepatocytes, myotubesInduced insulin resistance (hepatocytes, myotubes)
Promoted lipogenesis (hepatocytes)
[26,56]
FGF21WAT/BAT, liver, skeletal muscle, pancreas, CNSPromoted glucose uptake (adipocytes)
Stimulated thermogenesis (BAT)
Enhanced insulin secretion (pancreatic β cells)
Increased fatty acid oxidation and insulin sensitivity (liver, skeletal muscle)
Reduced NAFLD
Decreased VLDL uptake and lipogenesis (liver)
Decreased alcohol and sugar intake
Increased energy expenditure and decreased body weight (CNS)
[57,58,59,60]
Selenoprotein Pliver, skeletal muscleInhibited hepatic glucose production
Decreased glucose uptake (skeletal muscle)
[12,61,62,63,64]
LECT2liver, skeletal muscleIncreased M1/M2 ratio and hepatic inflammation (liver)
Development of insulin resistance (skeletal muscle)
Promoted lipid accumulation (liver)
[65,66]
Follistatinpituitary, skeletal muscle, liver, skeletal muscle, WAT, BATInhibition of FSH production (pituitary)
Suppressed skeletal muscle growth via antagonizing myostatin
Promoted insulin resistance (liver, skeletal muscle, WAT)
Increased glucose and FFA uptake after exercise training (skeletal muscle)
Induced differentiation of brown adipocytes
Promoted thermogenesis (BAT)
[14,15,67,68,69,70,71]
Hepassocinliver, skeletal muscle, WATPromoted insulin resistance
NAFLD
Adipogenesis (WAT)
[72,73,74,75]
RBP4various peripheral tissues including retinaIncreased lipolysis in adipocytes
Promoted hepatic mitochondrial dysfunction and hepatic steatosisSerum RBP4 levels were associated with insulin resistance and components of metabolic syndrome in humans
Depending on the source of RBP4 (hepatocytes or adipocytes), the effect of RBP4 is controversial.
- RBP4 treatment increased PEPCK (liver) and impaired insulin signaling (muscle and adipocytes).
- No effect of liver-secreted RBP4 on glucose homeostasis in mice
[76,77,78,79,80,81,82]
SMOC1liver, skeletal muscle, etc.Improved glycemic control via inhibiting gluconeogenesis and glucose output (liver)[83]
GDF15adipose tissue, skeletal muscle, liver, brain, heart, kidneyAnorexia
Increased energy metabolism (liver, muscle, adipose tissue) and lowered body weight
Stimulated thermogenic and lipolytic genes (BAT, WAT)
Improved glucose tolerance and insulin sensitivity
Prevented liver steatosis in HFD-fed mice
[84,85,86,87,88,89,90]
ANGPTL: Antiopoietin-like proteins; BAT: Brown adipose tissue; CNS: Central nervous system; FFA: Free fatty acid; FSH: Follicle-stimulating hormone; GDF15: Growth differentiation factor 15; HFD: High fat diet; LECT2: Leukocyte cell-derived chemotaxin 2; LPL: Lipoprotein lipase; RBP4: Retinol binding protein 4; SMOC1: SPARC-related modular calcium-binding protein 1; TG: Triglyceride; VLDL: Very low-density lipoprotein; WAT: White adipose tissue.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kim, T.H.; Hong, D.-G.; Yang, Y.M. Hepatokines and Non-Alcoholic Fatty Liver Disease: Linking Liver Pathophysiology to Metabolism. Biomedicines 2021, 9, 1903. https://doi.org/10.3390/biomedicines9121903

AMA Style

Kim TH, Hong D-G, Yang YM. Hepatokines and Non-Alcoholic Fatty Liver Disease: Linking Liver Pathophysiology to Metabolism. Biomedicines. 2021; 9(12):1903. https://doi.org/10.3390/biomedicines9121903

Chicago/Turabian Style

Kim, Tae Hyun, Dong-Gyun Hong, and Yoon Mee Yang. 2021. "Hepatokines and Non-Alcoholic Fatty Liver Disease: Linking Liver Pathophysiology to Metabolism" Biomedicines 9, no. 12: 1903. https://doi.org/10.3390/biomedicines9121903

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