Next Article in Journal
Unlocking the Hidden Potential of Agave tequilana for the Green Synthesis of Antibacterial ZnO Nanomaterials: A Waste-to-Value Nanotechnology Approach
Next Article in Special Issue
Role of Acetaldehyde and Dysregulated Mitophagic Lysosomal Processing in Chronic-Binge Ethanol-Induced Liver Injury
Previous Article in Journal
Traditional Health Practices May Promote Nrf2 Activation Similar to Exercise
Previous Article in Special Issue
Hepatic Zonation in MASLD: Old Question, New Challenge in the Era of Spatial Omics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Organokine-Mediated Crosstalk: A Systems Biology Perspective on the Pathogenesis of MASLD—A Narrative Review

by
Sandra Maria Barbalho
1,2,3,4,5,*,†,
Lucas Fornari Laurindo
6,*,
Vitor Engracia Valenti
7,
Nahum Méndez-Sánchez
8,9,
Mariana M. Ramírez-Mejía
8,10 and
Ricardo de Alvares Goulart
2,†
1
Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
2
Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
3
Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, SP, Brazil
4
Department of Research, Research Coordination Center, UNIMAR Charitable Hospital, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
5
Laboratory for Systematic Investigations of Diseases, Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
6
Division of Cellular Growth, Hemodynamic, and Homeostasis Disorders, Graduate Program in Medical Sciences, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo 01246-903, SP, Brazil
7
Faculty of Philosophy and Sciences, Universidade Estadual Paulista (UNESP), Campus Marília, Marília 17525-900, SP, Brazil
8
Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico
9
Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
10
Plan of Combined Studies in Medicine (PECEM-MD/PhD), Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(23), 11547; https://doi.org/10.3390/ijms262311547
Submission received: 17 September 2025 / Revised: 16 November 2025 / Accepted: 21 November 2025 / Published: 28 November 2025
(This article belongs to the Special Issue Molecular Advances and Insights into Liver Diseases)

Abstract

Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent chronic condition with a complex pathophysiology involving multiple organs. Organokines, including hepatokines, myokines, cardiokines, renokines, osteokines, and adipokines, play central roles in lipid metabolism, glucose homeostasis, inflammation, and fibrosis. Dysregulation of these signaling molecules contributes to the progression of MASLD and its systemic complications. This review examines the role of organokine-mediated crosstalk between the liver and peripheral organs (e.g., muscle, heart, kidneys, bone, and adipose tissue) in the pathogenesis of MASLD. Key molecules, such as myostatin, FGF-21, IL-6, and adiponectin, influence insulin sensitivity, lipid metabolism, and inflammation. Some organokines have protective effects (e.g., FGF-21, irisin, and klotho), while others, such as myostatin and fetuin-A, exacerbate insulin resistance and fibrosis. These findings suggest that targeting organokines could provide potential biomarkers and therapeutic strategies for MASLD. Future research should focus on elucidating the molecular mechanisms and assessing the role of organokines in the prevention and treatment of MASLD.

1. Introduction

People’s life expectations have increased thanks to the many benefits of modern society. However, an inadequate lifestyle can have a profound impact on people’s health and quality of life. Increased consumption of fats and sugar and lack of physical exercise can be related to overweight/obesity, insulin resistance/diabetes, dyslipidemia, metabolic syndrome, and cardiovascular diseases (CVD). On the other hand, these conditions are associated with increased inflammatory processes, oxidative stress, and several other factors that can exacerbate existing diseases or lead to the development of others, such as liver diseases and sarcopenia [1,2,3,4,5,6]. In 1986, the medical term non-alcoholic fatty liver disease, also known as NAFLD, was coined for a condition that would become the most prevalent liver disorder worldwide. However, over the last 3–4 years, specialists from around the world have come together to remove NAFLD from the medical dictionary and propose a new nomenclature for the disease, given the global rise in the development of cardiometabolic risk factors [7].
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent liver condition worldwide, affecting over 38.0% of adults globally; however, it is projected to reach approximately 56% in the next decade [8,9,10]. It is the second-leading cause for liver transplantation (and already the leading cause in women) worldwide, and significantly affects the population’s health [11,12,13]. The diagnosis includes liver steatosis, mainly associated with obesity, type 2 diabetes, dyslipidemia, and high blood pressure [14,15,16,17,18,19]. The definition of MASLD was proposed to include steatosis in the role scenario of the patient’s condition [20]. MASLD encompasses hepatic steatosis and more advanced features, including inflammation, hepatocyte ballooning, and lipoapoptotic damage. This condition can evolve from simple steatosis to cirrhosis and hepatocellular carcinoma [21].
Moreover, MASLD has emerged as the most important cause of chronic liver disease on the globe and, as pointed out above, has been driven mainly by the global epidemic of type 2 diabetes mellitus and obesity [6]. Insulin resistance and abdominal fat are potent factors in the development of MASLD. Since lipid metabolism is affected, there is an increase in inflammatory processes, oxidative stress, and mitochondrial dysfunction, which interfere with lipid metabolism and promote hepatic fat deposition [16,22,23,24].
Unfortunately, MASLD is not limited to Western countries. Studies have shown that it is also increasing in regions regularly associated with viral hepatitis and leading to a dual burden of MASLD and hepatitis B virus infection, contributing to the progression of increased risk for liver diseases and liver cancer [6,15,25,26].
A way to reduce the burden of MASLD requires a multidisciplinary approach involving public health professionals, policymakers, medical specialists, and other relevant experts [27]. This underscores the importance of raising awareness about risk factors, promoting early diagnosis, and advancing innovative public health policies. Properly trained physicians play a critical role in the early identification of liver diseases and in initiating effective therapeutic interventions. When supported by well-structured public policies, such efforts can mitigate the progression of disease and reduce the incidence of severe complications, including cirrhosis and hepatocellular carcinoma [28,29,30,31,32]. These actions could reduce public and private costs with the specific and expensive treatment of these conditions. Other factors that may be related to the burden on health systems are premature mortality and the inability to work [27].
Understanding the mechanisms behind MASLD requires a multifaceted approach, with significant contributions from both preventive and therapeutic research. A key area of focus is the crosstalk between the liver and other organs, which provides insight into how interorgan interactions can either exacerbate or alleviate the condition. The liver engages with skeletal and cardiac muscles, kidneys, adipose tissue, and bone through various chemical mediators known as organokines. These signaling molecules play a crucial role in modulating the progression of MASLD. In this review, we first summarize foundational literature on organokine-mediated communication, followed by recent advances in the field that shed light on their role in MASLD pathogenesis. We then identify critical gaps in current knowledge and highlight the potential for targeting these pathways therapeutically. Specifically, we focus on how hepatokines, myokines, adipokines, cardiokines, renokines, and osteokines influence key processes, including insulin sensitivity, lipid metabolism, inflammation, and fibrosis. We also discuss emerging biomarkers and intervention strategies based on this interorgan network, which could offer new diagnostic and therapeutic opportunities. The methodology of this review is described in Table A1 in Appendix A.

2. Exploring the Liver-Muscle Crosstalk (Hepatokines and Myokines) in MASLD

Many authors have demonstrated the association between hepatic and muscular metabolism [33,34,35,36]. Depending on the secretory pattern, this liver-muscle axis has been shown to have numerous metabolic repercussions and exacerbate liver diseases [16,37].
Skeletal muscle plays a fundamental role not only in locomotion and postural control but also in systemic metabolic regulation. It is a significant site for glucose uptake and utilization, thereby contributing to the maintenance of normoglycemia, and serves as a critical reservoir of amino acids during fasting. Consequently, metabolic and structural alterations in skeletal muscle can disrupt insulin signaling, promote chronic low-grade inflammation, and increase oxidative stress, all of which are closely associated with the pathogenesis of MASLD [38,39]. Furthermore, muscle has been considered a proper endocrine organ, releasing myokines that can have systemic effects. These myokines can have diverse effects and are related to antioxidant and anti-inflammatory properties when muscle homeostasis is maintained; however, they can also be associated with harmful impacts when muscle mass undergoes undesirable changes [40,41].
Myokines and hepatokines engage in crosstalk through the endocrine, paracrine, and autocrine pathways. Notwithstanding, they have specific associations regarding obesity, glucose metabolism, insulin resistance, diabetes mellitus, and metabolic syndrome [42,43,44]. Therefore, these biomarkers have been proposed to aid in the diagnosis and prognosis of liver, muscle, and systemic conditions, thereby innovating therapeutic approaches [45,46,47]. Table 1 presents the primary hepatokines and myokines associated with MASLD.
An essential consequence of metabolic impairment in muscle can result in sarcopenia, a progressive disorder that has increased worldwide. Following the European Working Group on Sarcopenia in Older People (EWGSOP), the prevalence of this condition is 8–36% in subjects under 60 years old and 10–27% in those aged 60 years and above [95,96,97]. It affects skeletal muscles, resulting in loss of muscle mass, strength, and function [98,99,100,101,102]. This condition is commonly associated with the aging process, hospitalization, oxidative stress, inflammation, diabetes/insulin resistance, as well as mitochondrial dysfunction, and liver dysfunction, such as MASLD [103,104], leading to physical dysfunction, impaired or severely diminished quality of life, increased morbidity, and mortality [105,106].
The physiopathological mechanisms related to MASLD and sarcopenia remain incompletely understood. As already mentioned, insulin resistance is a significant risk factor for MASLD and plays a crucial role in the muscle-liver crosstalk. People with reduced muscle strength have elevated inflammatory markers such as Interleukin (IL)-1β, IL-6, Tumor Necrosis Factor-α (TNF-α), and high-sensitivity C-reactive Protein (hs-CRP), suggesting the close relationship between muscle mass impairment and liver diseases [107,108,109,110,111]. A notable example is the release of TNF-α, which is related to the physiopathology of MASLD due to the pro-inflammatory macrophages that develop during adipocyte hypertrophy, leading to lipolysis (and an increase in free fatty acids) and favoring insulin resistance [22,112]. Insulin resistance, driven by elevated levels of pro-inflammatory cytokines, contributes to skeletal muscle catabolism. Additional shared risk factors linking muscular and hepatic dysfunction include dyslipidemia, obesity, type 2 diabetes mellitus, and metabolic syndrome. All of which play central roles in the pathophysiology of both sarcopenia and metabolic-associated liver diseases [113,114,115,116].
Furthermore, it is relevant to consider the secretory pattern of the skeletal muscle. Myokines have essential roles in the balance of glucose and lipid metabolism. Impaired muscle mass can also impair the release of myokines [35,71,117,118,119,120]. On the other hand, patients with MASLD can alter myokine levels, exposing muscles to increased ammonia concentrations resulting from hepatocyte dysfunction and an imbalanced urea cycle [110,121]. In addition, fibroblast growth factor-21 (FGF-21) levels are higher in decompensated cirrhosis patients presenting sarcopenia. FGF-21 functions as a liver-muscle crosstalk, imbalances muscle mass regeneration by inhibiting the PI3K/Akt (Phosphoinositide 3-kinase/Protein kinase b) pathways [16,110,122]. The liver and muscle share many metabolic and physiological pathways; thus, MASLD and muscle impairment share the consequences of hepatokines, myokines, insulin resistance, oxidative stress, and chronic inflammation. For these reasons, it is possible to conclude that one disease contributes to the development of the other. Apart from the physiopathological relationship, it is essential to understand the clinical connection between MASLD and sarcopenia to identify high-risk patients and provide effective clinical management. The early recognition of the relationship between muscle and liver diseases can improve the patient’s prognosis [14,107,123,124].
In a meta-analysis, the authors demonstrated that sarcopenia increases the risk of MASLD and is associated with an elevated risk of liver fibrosis [125]. For those mentioned reasons, sarcopenia is not only considered an aging-related syndrome, but it is acknowledged to be secondarily related to chronic diseases such as MASLD [105,120,126,127,128,129,130,131,132].
The role of hepatokines (that directly modulate hepatic homeostasis) in the context of MASLD includes Angiopoietin-Like 3 (ANGPTL3), that impairs lipid clearance, being associated with dyslipidemia and steatosis; activin protects against fatty acid influx into the liver (beneficial regulator); fetuin-A promotes insulin resistance and hepatic lipid accumulation; FGF-21 (hepatic) increases fatty acid oxidation, with a protective role in MASLD. Additionally, selenoprotein P promotes insulin resistance and liver inflammation. Ultimately, lower levels of sex hormone-binding globulin (SHBG) increase the risk of MASLD [50,51,53,54,55,133,134,135].
In summary, hepatokines such as FGF-21, fetuin-A, ANGPTL3, and others play key roles in maintaining hepatic and systemic metabolic homeostasis. Their dysregulation contributes to lipotoxicity, insulin resistance, and inflammation, central events in the pathogenesis of MASLD. A deeper understanding of hepatokine signaling provides new insights into the systemic nature of the disease, highlighting these molecules as potential diagnostic biomarkers and therapeutic targets.
On the other hand, myokines serve as essential mediators of muscle-liver crosstalk, especially in the context of physical activity and metabolic health. Alterations in molecules such as irisin and IL-6 can either exacerbate or attenuate hepatic steatosis, depending on the physiological context. These findings underscore the protective effects of skeletal muscle homeostasis and supportive exercise as a non-pharmacological strategy in the prevention and management of MASLD.

3. Exploring the Liver-Heart Crosstalk (Hepatokines and Cardiokines) in MASLD

As previously noted, clinical and pathophysiological evidence support a causal association between MASLD and CVD [136,137]. MASLD plays a pivotal role in the development and progression of CVD, often preceding hepatic complications such as cirrhosis or hepatocellular carcinoma. The heart actively contributes to this pathophysiological interplay through the secretion of bioactive molecules collectively referred to as cardiokines [11,138].
In MASLD, the steatotic scenario and an environment of lipotoxicity impair liver regulatory functions [139], leading to the accumulation of triglycerides within hepatocytes and the release of atherogenic lipoproteins into the circulation. The increase in liver lipids leads to the release of pro-inflammatory cytokines, such as IL-6 and TNF-α, and the production of reactive oxygen species (ROS), contributing to systemic inflammation, vascular damage, and accelerated atherosclerosis [140,141]. Endothelial dysfunction, a hallmark of early CVD, is commonly observed in MASLD and is exacerbated by both hyperinsulinemia and oxidative stress [142]. This dysfunction impairs nitric oxide bioavailability, leading to increased vascular tone, platelet aggregation, and ultimately, the formation and instability of plaques. Additionally, MASLD is associated with coronary artery calcification, a validated surrogate of coronary atherosclerosis and predictor of major cardiovascular events [143,144,145].
In a prospective five-year study, the authors have demonstrated that the increased risk for CVD in type-2 diabetic participants was partly associated with metabolic syndrome, but significantly linked to NAFLD [146]. A meta-analysis with 34,043 participants demonstrated that patients with NAFLD have an elevated risk of fatal and non-fatal cardiovascular events [147,148]. Moreover, liver inflammation, histologic severity, and the MASLD activity score were correlated with the severity of inflammation in adipose tissue, characterized by increased pro-inflammatory macrophages, TNF-α, and IL-6 in the visceral adipose tissue [11,138].
Some authors have demonstrated that myocardial ischemia, especially acute myocardial infarction, exacerbates liver injury in metabolic-associated fatty liver disease (MAFLD) through two central and interrelated mechanisms. The first is related to the elevation of circulating inflammatory monocytes and their accelerated recruitment to the liver via the MCP-1 (monocyte chemoattractant protein-1)/CCR2 (MCP-1 receptor) axis, and the second involves the synthesis and release of periostin, an extracellular matrix protein re-expressed by cardiac fibroblasts during cardiac injury [149,150,151].
In summary, cardiokines can profoundly influence liver metabolism and vice versa. Atrial Natriuretic Peptide (ANP) regulates liver glycogen metabolism and glucose homeostasis. B-Type Natriuretic Peptide (BNP) is a biomarker of volume overload and is increased in cirrhosis. Growth Differentiation Factor 15 (GDF-15) is involved in the heart-liver crosstalk in cardiac pathogenesis. IL-33 can reduce the inflammatory process as well as hepatic fibrosis in early stages. Cardiac myostatin is associated with muscle atrophy and hepatic fibrosis. On the other hand, natriuretic peptides improve lipid metabolism and may play a protective role against MASLD [54,66,67,71,72,73]. Table 1 summarizes the effects of some cardiokines related to MASLD, and Figure 1 shows the relationship between the liver and the heart.
In conclusion, cardiokines represent a crucial link in the bidirectional communication between the liver and the cardiovascular system. In MASLD, these molecules may contribute to hepatic fibrogenesis and insulin resistance, while the progression of liver disease can, in turn, exacerbate cardiac remodeling and endothelial dysfunction. This reciprocal interaction highlights the significance of cardiovascular risk assessment in patients with MASLD, suggesting cardiokines as potential biomarkers and therapeutic targets for early intervention and risk stratification.

4. Exploring the Liver-Kidney Crosstalk (Hepatokines and Renokines) in MASLD

The kidneys and liver maintain complementary functions, thus playing a central role in maintaining homeostasis and detoxification. Specifically in the kidneys, the regulation of blood volume/pressure, electrolyte and acid-base balance, secretion of erythropoietin and calcitriol, prostaglandin synthesis, and control of the renin–angiotensin–aldosterone system (RAAS) [152]. Failure of these mechanisms increases morbidity and mortality, triggering multisystem dysfunctions with a significant impact on the liver [153,154,155,156].
The regulation of renal function involves a complex network of signaling molecules, known as renokines, which influence systemic homeostasis and the progression of kidney disease [157]. These may include Bone Morphogenetic Proteins (BMPs), particularly BMP-7, which antagonizes the pro-fibrotic TGF-β pathway, inhibiting epithelial–mesenchymal transition and attenuating renal fibrosis and inflammation [158,159].
Erythropoietin (EPO) is a glycoprotein hormone primarily synthesized in the kidneys, which is essential for erythropoiesis. In chronic kidney disease (CKD), its reduced production leads to normocytic, normochromic anemia, exacerbating fatigue and cardiovascular dysfunction [160].
Osteocytes produce Fibroblast Growth Factor 23 (FGF-23) in response to hyperphosphatemia. In early CKD, it promotes renal phosphate excretion, but its sustained elevation contributes to ventricular hypertrophy, cardiovascular calcifications, and kidney disease progression [161,162].
GDF-15, a member of the transforming growth factor-beta (TGF-β) superfamily, is stimulated by cellular stress, including stress in mitochondria, endoplasmic reticulum, and liver. It is also released with exercises, smoking, and age [157]. Klotho is a transmembrane protein with anti-aging action, expressed in renal tubules. It acts as a co-receptor for FGF-23, regulating phosphate and vitamin D metabolism. Its depletion in CKD is associated with vascular calcification, renal fibrosis, and increased mortality [163,164].
Lipocalin-2 is a biomarker of acute kidney injury with two functions: at physiological levels, it promotes tissue repair; when persistently elevated in CKD, it exacerbates tubular inflammation and cellular apoptosis [165].
The RAAS is involved in the chronic activation of renin and angiotensin II, which induces vasoconstriction, glomerular inflammation, and interstitial fibrosis, thereby worsening hypertension and kidney injury [166,167].
TNF-Related Inducer of Apoptosis is a multifunctional cytokine that, at high concentrations, activates inflammatory pathways such as Nuclear Factor Kappa B (NF-κB) and promotes glomerular fibrosis, correlating with the progression of CKD. TNF-α is also produced by T cells and resident kidney cells (such as mesangial cells, renal epithelial cells, and vascular endothelial cells) [168]. It can also induce a natriuretic effect and stimulate hypertensive response and renal injury, which is also toxic to glomerular epithelial cells. IL-1 is produced by hematopoietic cells and resident kidney cell lineages. IL-1α and IL-1β bind to the same receptor for IL-1. The NLRP3 inflammasome mediates the conversion of pro-IL-1β to its active form. The activation of its receptor leads to the recruitment of Myd88 and many IL-1 receptor-associated kinases, inducing the translocation of NF-κB’s p65 component to the nucleus, which drives the transcription of genes related to inflammatory proteins, such as TNF [156,169].
Kidney-liver crosstalk is essential in normal and some pathological conditions. Renal-induced liver harm and liver-induced kidney diseases can occur due to the interaction between these organs [170]. This phenomenon is associated with cytokine synthesis and the activation of pro-inflammatory pathways, as well as oxidative stress, ischemia and reperfusion, metabolic acidosis, and modifications in enzyme activity. These metabolic pathways provide the foundation for the interaction between the kidneys and liver [156].
Regarding cytokine production, acute renal failure triggers a low-grade systemic inflammatory response characterized by elevated levels of cytokines, such as IL-6, IL-17A, and TNF-α. These mediators promote endothelial dysfunction and increased hepatic vascular permeability, facilitating immune cell infiltration and liver injury. This scenario impairs cytochrome P450 (CYP) enzyme activity, mainly CYP3A4, CYP3A1/2, and CYP2E1, thereby compromising hepatic drug metabolism. Notwithstanding, indoxyl sulfate accumulation and increased IL-10 levels further suppress CYP function. IL-6 also induces the synthesis of acute-phase proteins while downregulating hepatic proteins such as albumin [156,171].
In the case of oxidative stress, studies have shown that both ischemic and non-ischemic acute renal failure trigger oxidative stress, contributing to liver injury. The early oxidative activation leads to leukocyte infiltration, hepatocyte apoptosis, and depletion of hepatic antioxidants, such as superoxide dismutase and catalase, while increasing lipid peroxidation markers, including malondialdehyde [172,173].
The liver and kidneys are crucial modulators of acid-base balance. In chronic renal failure, the balancing mechanism can be disrupted, leading to metabolic acidosis [174]. Acute acidosis profoundly interferes with the body. One consequence can be a disturbance in glutamine metabolism, resulting in an acute increase in blood glutamine concentrations and a decline in total hepato-splanchnic glutamine concentrations. Some authors have demonstrated that chronic acidosis reduces glutamine extraction and albumin synthesis in the liver, while increasing renal glutamine and ammonia production [175]. Furthermore, acidosis is linked to an increased rate of systemic protein turnover (both protein synthesis and proteolysis) [176,177,178].
On the other hand, in liver failure, for example, in cirrhosis state, its multisystem complications include acute kidney injury in part of the hospitalized patients [179,180]. Hepatorenal syndrome, a severe form of acute kidney disease in advanced cirrhotic patients, is characterized by renal vasoconstriction and decreased glomerular filtration rate. Its mechanism involves portal hypertension, splanchnic vasodilation, neurohormonal activation (RAAS/sympathetic), and renal hypoperfusion [181]. Biglycan (a protein released by the cirrhotic liver) promotes renal inflammation via TLR2/TLR4 and autophagy [49,182]. Cirrhotic cardiomyopathy and adrenal insufficiency exacerbate renal hypoperfusion, and relative adrenal deficiency further exacerbates renal hypoperfusion [57,183,184].
In summary, the role of renin influences hormonal regulation and renal-hepatic stress. Some examples include erythropoietin, which, among other functions, modulates insulin sensitivity and may reduce liver steatosis. Klotho exhibits anti-inflammatory and antioxidant properties, which contribute to a protective effect in MASLD. Neutrophil gelatinase-associated lipocalin (NGAL or lipocalin 2) is a biomarker of renal stress and correlates with the severity of liver injury. Finally, renin is associated with the activation of the RAAS axis and worsening of steatohepatitis [74,76,77,78,79].
In conclusion, renokines play a significant role in the systemic manifestations of MASLD. Some of them are related to modulating oxidative stress, vascular tone, and hepatic function. In metabolic disease states, kidney dysfunction can lead to the accumulation of uremic toxins and reduced clearance of inflammatory mediators, contributing to hepatic injury and fibrosis. Furthermore, MASLD itself may exacerbate renal damage through systemic inflammation and altered lipid metabolism, establishing a vicious cycle. Recognizing the liver-kidney axis in MASLD pathophysiology supports the development of integrated therapeutic strategies aimed at preserving renal function and mitigating hepatic progression. Figure 2 summarizes the relationship between the liver and the kidney. Table 1 presents the primary renokines associated with MASLD.

5. Exploring the Liver-Adipose Tissue Crosstalk (Hepatokines and Adipokines) in MASLD

Adipokines are biomarkers produced by adipose tissue that regulate glucose and lipid metabolism, influencing liver diseases, appetite, insulin sensitivity, and fibrogenesis [185]. Under normal physiological conditions, adipokines exert regulatory effects on multiple organs, including the brain, liver, skeletal muscle, bone, and the endocrine pancreas [186,187].
In overnutrition, adipose tissue undergoes structural and functional remodeling, resulting in alterations in the secretion of adipokines and inflammatory mediators. This uncontrolled pattern, especially involving adipokines with metabolic and immunoregulatory functions, contributes to adipose tissue dysfunction [188]. This imbalance alters both the secretion profiles and biological actions of adipokines, promoting an increase in ROS, leading to endoplasmic reticulum stress and mitochondrial dysfunction [189,190]. The consequences lead to a state of chronic low-grade inflammation, exacerbating metabolic dysregulation and insulin resistance [191]. In addition to the increase in ROS, adipose tissue also leads to an imbalance of pro-inflammatory adipocytokines, such as IL-6, TNF-α, and resistin [192,193]. In this scenario, there is an amplification of reactive species production, inflammation, and metabolic injury. Another consequence is that the lipid storage capacity of adipose tissue is compromised, favoring the diversion of excess fatty acids to other tissues such as the liver, where their ectopic deposition promotes lipotoxicity and metabolic disorders [188,190,191,194].
Emerging data indicates a sophisticated, bidirectional communication axis between adipose tissue and the liver, coordinating systemic energy homeostasis. This interplay, mediated by both hepatokines and adipokines, plays a central role in the onset and progression of MASLD [193,195].
Among adipokines, adiponectin exerts protective effects by activating the AMPK and PPAR-α pathways, thereby reducing hepatic steatosis, inflammation, and fibrosis; notably, its levels decline as MASLD progresses. In contrast, leptin plays a role in early metabolic regulation; however, chronic elevations and leptin resistance contribute to the worsening of hepatic inflammation and disease progression [196,197]. In obese individuals, adipose tissue hypertrophy leads to a state of chronic low-grade inflammation, increases the attraction of chemotactic molecules, and decreases adiponectin levels. Furthermore, this inflammatory state significantly contributes to insulin and leptin resistance [42,198].
The FGF-21-adiponectin axis serves as a key mediator of crosstalk between the liver and adipose tissue, exerting beneficial effects on maintaining energy consumption homeostasis [199,200].
RBP4 (Retinol-binding protein 4) is an adipokine that is secreted primarily by hepatic and adipose cells and has a role in transporting retinol from the liver to peripheral tissues. Some studies have shown that the levels of this adipokine are associated with an increased risk of metabolic diseases, including obesity, type 2 diabetes, and MASLD [201,202].
Other adipokines, such as resistin, visfatin, and chemerin, also modulate hepatic lipid handling and inflammatory responses, although their roles are less clearly defined. This bidirectional liver-adipose axis forms a feedback loop where hepatokine/adipokine imbalances exacerbate insulin resistance and hepatic lipid deposition [203,204,205,206,207].
Ultimately, the effects of numerous adipokines have been known for a long time. In summary, adiponectin plays a significant anti-inflammatory role, in addition to improving insulin sensitivity. It also has protective effects against steatosis and consequent fibrosis [208,209]. Leptin acts on the hypothalamic satiety center and is associated with decreased inflammation and steatosis. However, obese individuals may develop hyperleptinemia and hepatic fibrosis, thus exhibiting unbeneficial effects. Leptin is also increased in MASLD. Resistin, as its name suggests, induces insulin resistance and is associated with the development of steatosis. Another molecule associated with insulin resistance is visfatin. It stimulates NF-κB, consequently increasing pro-inflammatory cytokines such as TNF-α, IL-6, and IL-1β. It is associated with the worsening of steatosis. RBP4 levels are related to increased risk of metabolic diseases, including obesity, type 2 diabetes, and MASLD. The FGF-21-adiponectin axis is a key mediator of crosstalk between the liver and adipose tissue, exerting beneficial effects [85,200,210,211,212,213,214,215]. Table 1 illustrates the impact of some adipokines, and Figure 3 summarizes the relationship between liver and white adipose tissue.
Ultimately, adipose tissue, particularly in its dysfunctional and inflamed state, acts as a critical endocrine organ through the secretion of adipokines that modulate hepatic lipid metabolism and inflammation. The imbalance between pro-inflammatory and anti-inflammatory adipokines is closely associated with MASLD progression. These insights underscore the significance of adipose-liver communication and reinforce the therapeutic potential of targeting adipokine pathways in the treatment of metabolic liver disease.

6. Exploring the Liver-Bone Tissue Crosstalk (Hepatokines and Osteokines) in MASLD

Although a link between bone and liver seems unlikely, a bidirectional liver-bone axis exists, in which bone-derived osteokines regulate hepatic metabolism and contribute to the pathogenesis of MASLD [216]. For these reasons, bone is also recognized as an endocrine organ, secreting osteokines such as FGF-23, NGAL, osteopontin, and osteocalcin that interact with the liver. These osteokines can predict the incidence and progression of metabolic disorders and liver diseases [217,218].
Protective osteokines include osteocalcin, which reduces liver steatosis. Conversely, other osteokines, such as osteopontin, which is elevated in MASLD, promote inflammation and fibrosis. FGF-23, which is associated with increased insulin resistance, and sclerostin, which reduces insulin sensitivity, exacerbate the progression of MASLD and its consequences [219,220].
In summary, osteokines such as FGF-23 correlate with vascular calcification and endothelial dysfunction, in addition to being associated with an increased risk of steatosis and portal hypertension. Osteocalcin promotes hepatoprotective effects. In animal models, its administration activates the Nuclear factor erythroid 2-related factor 2 (Nrf2) pathway [209], increases the expression of antioxidant enzymes such as superoxide dismutase, catalase, and glutathione peroxidase, and inhibits cJun N-terminal kinase (JNK) activation, resulting in reduced steatosis and liver injury [221]. Furthermore, it regulates fatty acid uptake via CD36 through the AMPK/FOXO1/BCL6 pathway [222]. It is related to increased insulin release and reduced fibrosis in MASLD [223,224,225,226]. Osteopontin promotes hepatic inflammation and fibrosis, and its levels are increased in MASLD/hepatic steatosis. Its levels are elevated in patients with Nonalcoholic steatohepatitis (NASH), and promote macrophage recruitment and activation of fibrogenic pathways in the liver [227,228]. Finally, sclerostin, produced by osteocytes, interferes with Wnt/β-catenin signaling, being associated with metabolic dysfunction, insulin resistance, and MASLD [93]. NGAL upregulation may work as a protective osteokine, reducing obesity-induced glucose intolerance. The regulatory importance of it may be related to the stimulation of PPARγ (peroxisome proliferator-activated receptor gamma), which mediates liver adipogenesis and lipogenesis [90,210]. Table 1 shows some osteokines and their effects. Figure 4 summarizes the relationship between the liver and bones.
In conclusion, although traditionally associated with bone metabolism, osteokines such as osteocalcin and osteopontin have emerged as influential regulators of energy metabolism and liver health. Their involvement in insulin sensitivity, inflammation, and lipid handling reveals a novel endocrine axis between bone and liver. Future studies are essential to clarify their exact role and therapeutic potential in MASLD, especially considering the frequent overlap between hepatic and skeletal complications in metabolic disorders.

7. Conclusions

Tissue-specific organokines play crucial roles in modulating MASLD, acting on interorgan axes (muscle-liver, bone-liver, kidney-liver, heart-liver, and adipose tissue-liver). While some (such as FGF-21, irisin, adiponectin, klotho, and osteocalcin) have protective effects, others (such as myostatin, fetuin-A, visfatin, and ANGPTL3) exacerbate insulin resistance, inflammation, and fibrosis. Dysfunction in these pathways reflects the systemic nature of MASLD, highlighting potential therapeutic targets for this condition. However, some cytokines, such as FGF-21 and myostatin, are produced in multiple tissues with distinct contextual effects, and there are context-dependent molecules, including IL-6 and IL-15. Dysregulation of these pathways reinforces the systemic nature of MASLD, highlighting the need for multiorgan therapeutic targets.

8. Future Perspectives

Future studies should aim to elucidate further the molecular pathways by which organokines, including hepatokines, adipokines, myokines, cardiokines, and osteokines, modulate the development and progression of MASLD. A detailed understanding of the intracellular signaling cascades involved in inflammation, fibrosis, steatosis, and systemic metabolic dysfunction will enable the identification of precise molecular targets for therapeutic intervention. These organokines could work as noninvasive biomarkers for early diagnosis, disease stratification, and treatment monitoring in MASLD. In the therapeutic approach, selective inhibition of pro-inflammatory organokines could mitigate hepatic inflammation and fibrosis. However, clinical trials are necessary to evaluate the safety, efficacy, and long-term outcomes of such interventions.
Another critical area for further developing MASLD research is the interaction between the gut microbiota and the liver. The gut microbiota, composed of bacteria, fungi, viruses, and archaea, along with its metabolites, can profoundly impact liver health, leading to the amelioration or exacerbation of liver diseases. Unhealthy changes to the gut microbiota can lead to intestinal barrier dysfunction and the displacement of microbial components, further leading to the development of MASLD [229]. The gut microbiota is also related to hepatocyte lipid metabolism and immune regulation, and recent research has linked interventions with gut microbiota to the brain, kidney, heart, adipose tissue, and muscle health. In this sense, future research endeavors must elucidate how new therapeutic avenues, including microbial-targeted therapies, dietary interventions, and metabolic surgery, can be used to treat and intervene against MASLD effectively [230,231,232].
Finally, given the multisystemic nature of MASLD, future therapeutic approaches should adopt a comprehensive and personalized strategy that includes modulation of organokine signaling, pharmacological interventions, lifestyle modification, and management of associated comorbidities. This integrative vision will be crucial in preventing disease progression and reducing long-term complications in individuals affected by the disease. However, clinical trials need to be performed to open the way for using these molecules as therapies for MASLD and other metabolic conditions.

Author Contributions

Conceptualization, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G.; methodology, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G.; software, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G.; validation, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G.; formal analysis, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G.; investigation, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G.; resources, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G.; data curation, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G.; writing—original draft preparation, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G.; writing—review and editing, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G.; visualization, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G.; supervision, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G.; project administration, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G.; funding acquisition, S.M.B., L.F.L., V.E.V., N.M.-S., M.M.R.-M. and R.d.A.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We would like to express our sincere gratitude to BioRender (https://BioRender.com) for providing the medical figures used to create the images for this article. Their high-quality illustrations significantly enhanced the clarity and visual appeal of our work. BioRender is licensed under an active Creative Commons license, allowing its use with proper attribution. We appreciate their support and contribution to the advancement of scientific communication.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Appendix A

Table A1. Methodology for Selecting the Studies Included in this Review.
Table A1. Methodology for Selecting the Studies Included in this Review.
Scope and ObjectivesThis narrative review aims to synthesize current evidence on the mechanisms of interorgan crosstalk in MASLD, with an emphasis on emerging mechanistic and therapeutic perspectives.
Search StrategyPubMed, Scopus, Web of Science, ScienceDirect, and SpringerLink were searched. Keywords included “Organokines” and “MASLD”. No time restrictions were imposed. Inclusion comprised articles that delve into hepatokines, osteokines, renokines, myokines, cardiokines, and adipokines in the context of MASLD development and progression, as well as the interorgan crosstalk between the liver, bones, kidneys, muscles, heart, and adipose tissue producers in MASLD.
Data Extraction and SynthesisThe results were synthesized thematically, highlighting three main aspects: “Tissue-Derived Cytokine,” “Condition/Stimulus for Release,” and “Relations with MASLD.”

References

  1. Zhang, J.Z.; Liu, C.H.; Shen, Y.L.; Song, X.N.; Tang, H.; Li, H. Sarcopenia in trauma patients: A systematic review and meta-analysis. Ageing Res. Rev. 2024, 104, 102628. [Google Scholar] [CrossRef]
  2. Mellen, R.H.; Girotto, O.S.; Marques, E.B.; Laurindo, L.F.; Grippa, P.C.; Mendes, C.G.; Garcia, L.N.H.; Bechara, M.D.; Barbalho, S.M.; Sinatora, R.V.; et al. Insights into Pathogenesis, Nutritional and Drug Approach in Sarcopenia: A Systematic Review. Biomedicines 2023, 11, 136. [Google Scholar] [CrossRef]
  3. Laurindo, L.F.; Rodrigues, V.D.; Laurindo, L.F.; Cherain, L.M.A.; de Lima, E.P.; Boaro, B.L.; da Silva Camarinha Oliveira, J.; Chagas, E.F.B.; Catharin, V.C.S.; Dos Santos Haber, J.F.; et al. Targeting AMPK with Irisin: Implications for metabolic disorders, cardiovascular health, and inflammatory conditions—A systematic review. Life Sci. 2025, 360, 123230. [Google Scholar] [CrossRef]
  4. Direito, R.; Barbalho, S.M.; Sepodes, B.; Figueira, M.E. Plant-Derived Bioactive Compounds: Exploring Neuroprotective, Metabolic, and Hepatoprotective Effects for Health Promotion and Disease Prevention. Pharmaceutics 2024, 16, 577. [Google Scholar] [CrossRef]
  5. Morales-Galicia, A.E.; Ramírez-Mejía, M.M.; Ponciano-Rodriguez, G.; Méndez-Sánchez, N. Revolutionizing the understanding of liver disease: Metabolism, function and future. World J. Hepatol. 2024, 16, 1365–1370. [Google Scholar] [CrossRef] [PubMed]
  6. Zhou, X.D.; Chen, Q.F.; Targher, G.; Byrne, C.D.; Mantzoros, C.S.; Zhang, H.; Lonardo, A.; Lip, G.Y.H.; Porta, G.; Misra, A.; et al. Global burden of disease attributable to metabolic risk factors in adolescents and young adults aged 15–39, 1990–2021. Clin. Nutr. 2024, 43, 391–404. [Google Scholar] [CrossRef] [PubMed]
  7. Portincasa, P.; Baffy, G. Metabolic dysfunction-associated steatotic liver disease: Evolution of the final terminology. Eur. J. Intern. Med. 2024, 124, 35–39. [Google Scholar] [CrossRef]
  8. Huang, D.Q.; El-Serag, H.B.; Loomba, R. Global epidemiology of NAFLD-related HCC: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 223–238. [Google Scholar] [CrossRef]
  9. Barbalho, S.M.; Méndez-Sánchez, N.; Fornari Laurindo, L. AdipoRon and ADP355, adiponectin receptor agonists, in Metabolic-associated Fatty Liver Disease (MAFLD) and Nonalcoholic Steatohepatitis (NASH): A systematic review. Biochem. Pharmacol. 2023, 218, 115871. [Google Scholar] [CrossRef] [PubMed]
  10. Carpi, R.Z.; Barbalho, S.M.; Sloan, K.P.; Laurindo, L.F.; Gonzaga, H.F.; Grippa, P.C.; Zutin, T.L.M.; Girio, R.J.S.; Repetti, C.S.F.; Detregiachi, C.R.P.; et al. The Effects of Probiotics, Prebiotics and Synbiotics in Non-Alcoholic Fat Liver Disease (NAFLD) and Non-Alcoholic Steatohepatitis (NASH): A Systematic Review. Int. J. Mol. Sci. 2022, 23, 8805. [Google Scholar] [CrossRef]
  11. Sebastiani, G.; Raggi, P.; Guaraldi, G. Integrating the care of Metabolic Dysfunction-associated Steatotic Liver Disease into Cardiac Rehabilitation: A Multisystem Approach. Can. J. Cardiol. 2025. [Google Scholar] [CrossRef]
  12. Devarbhavi, H.; Asrani, S.K.; Arab, J.P.; Nartey, Y.A.; Pose, E.; Kamath, P.S. Global burden of liver disease: 2023 update. J. Hepatol. 2023, 79, 516–537. [Google Scholar] [CrossRef] [PubMed]
  13. Noureddin, M.; Vipani, A.; Bresee, C.; Todo, T.; Kim, I.K.; Alkhouri, N.; Setiawan, V.W.; Tran, T.; Ayoub, W.S.; Lu, S.C. NASH leading cause of liver transplant in women: Updated analysis of indications for liver transplant and ethnic and gender variances. Off. J. Am. Coll. Gastroenterol.|ACG 2018, 113, 1649–1659. [Google Scholar] [CrossRef] [PubMed]
  14. Viswanath, A.; Fouda, S.; Fernandez, C.J.; Pappachan, J.M. Metabolic-associated fatty liver disease and sarcopenia: A double whammy. World J. Hepatol. 2024, 16, 152–163. [Google Scholar] [CrossRef] [PubMed]
  15. Zhang, H.; Targher, G.; Byrne, C.D.; Kim, S.U.; Wong, V.W.; Valenti, L.; Glickman, M.; Ponce, J.; Mantzoros, C.S.; Crespo, J.; et al. A global survey on the use of the international classification of diseases codes for metabolic dysfunction-associated fatty liver disease. Hepatol. Int. 2024, 18, 1178–1201. [Google Scholar] [CrossRef]
  16. De Cól, J.P.; de Lima, E.P.; Pompeu, F.M.; Cressoni Araújo, A.; de Alvares Goulart, R.; Bechara, M.D.; Laurindo, L.F.; Méndez-Sánchez, N.; Barbalho, S.M. Underlying Mechanisms behind the Brain-Gut-Liver Axis and Metabolic-Associated Fatty Liver Disease (MAFLD): An Update. Int. J. Mol. Sci. 2024, 25, 3694. [Google Scholar] [CrossRef]
  17. Zeng, X.F.; Varady, K.A.; Wang, X.D.; Targher, G.; Byrne, C.D.; Tayyem, R.; Latella, G.; Bergheim, I.; Valenzuela, R.; George, J.; et al. The role of dietary modification in the prevention and management of metabolic dysfunction-associated fatty liver disease: An international multidisciplinary expert consensus. Metab. Clin. Exp. 2024, 161, 156028. [Google Scholar] [CrossRef]
  18. Tauil, R.B.; Golono, P.T.; de Lima, E.P.; de Alvares Goulart, R.; Guiguer, E.L.; Bechara, M.D.; Nicolau, C.C.T.; Yanaguizawa Junior, J.L.; Fiorini, A.M.R.; Méndez-Sánchez, N.; et al. Metabolic-Associated Fatty Liver Disease: The Influence of Oxidative Stress, Inflammation, Mitochondrial Dysfunctions, and the Role of Polyphenols. Pharmaceuticals 2024, 17, 1354. [Google Scholar] [CrossRef]
  19. El-Shoukrofy, M.S.; Ismail, A.; Elhamammy, R.H.; Abdelhady, S.A.; Nassra, R.; Makkar, M.S.; Agami, M.A.; Wahid, A.; Nematalla, H.A.; Sai, M.; et al. Novel thiazolones for the simultaneous modulation of PPARγ, COX-2 and 15-LOX to address metabolic disease-associated portal inflammation. Eur. J. Med. Chem. 2025, 289, 117415. [Google Scholar] [CrossRef]
  20. Alabdul Razzak, I.; Fares, A.; Stine, J.G.; Trivedi, H.D. The Role of Exercise in Steatotic Liver Diseases: An Updated Perspective. Liver Int. Off. J. Int. Assoc. Study Liver 2025, 45, e16220. [Google Scholar] [CrossRef]
  21. An, H.; Jang, Y.; Choi, J.; Hur, J.; Kim, S.; Kwon, Y. New Insights into AMPK, as a Potential Therapeutic Target in Metabolic Dysfunction-Associated Steatotic Liver Disease and Hepatic Fibrosis. Biomol. Ther. 2025, 33, 18–38. [Google Scholar] [CrossRef] [PubMed]
  22. Sakurai, Y.; Kubota, N.; Yamauchi, T.; Kadowaki, T. Role of Insulin Resistance in MAFLD. Int. J. Mol. Sci. 2021, 22, 4156. [Google Scholar] [CrossRef]
  23. Gao, J.; Cang, X.; Liu, L.; Lin, J.; Zhu, S.; Liu, L.; Liu, X.; Zhu, J.; Xu, C. Farrerol alleviates insulin resistance and hepatic steatosis of metabolic associated fatty liver disease by targeting PTPN1. J. Cell. Mol. Med. 2024, 28, e70096. [Google Scholar] [CrossRef]
  24. de Lima, E.P.; Moretti, R.C., Jr.; Torres Pomini, K.; Laurindo, L.F.; Sloan, K.P.; Sloan, L.A.; Castro, M.V.M.; Baldi, E., Jr.; Ferraz, B.F.R.; de Souza Bastos Mazuqueli Pereira, E.; et al. Glycolipid Metabolic Disorders, Metainflammation, Oxidative Stress, and Cardiovascular Diseases: Unraveling Pathways. Biology 2024, 13, 519. [Google Scholar] [CrossRef]
  25. Wu, C.; Targher, G.; Byrne, C.D.; Mao, Y.; Cheung, T.T.; Yilmaz, Y.; Valenti, L.; Méndez-Sánchez, N.; Sookoian, S.; Chan, W.K.; et al. Global, regional, and national burden of primary liver cancer attributable to metabolic risks: An analysis of the Global Burden of Disease Study 1990–2021. Am. J. Gastroenterol. 2025, 120, 2280–2290. [Google Scholar] [CrossRef]
  26. Kim, D.; Loomba, R.; Ahmed, A. Current Burden of MASLD, MetALD, and Hepatic Fibrosis among US Adults with Prediabetes and Diabetes, 2017–2023. Clin. Mol. Hepatol. 2024, 31, e235–e238. [Google Scholar] [CrossRef]
  27. Younossi, Z.M.; Razavi, H.; Sherman, M.; Allen, A.M.; Anstee, Q.M.; Cusi, K.; Friedman, S.L.; Lawitz, E.; Lazarus, J.V.; Schuppan, D.; et al. Addressing the High and Rising Global Burden of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) and Metabolic Dysfunction-Associated Steatohepatitis (MASH): From the Growing Prevalence to Payors’ Perspective. Aliment. Pharmacol. Ther. 2025, 61, 1467–1478. [Google Scholar] [CrossRef]
  28. Mitsinikos, T.; Aw, M.M.; Bandsma, R.; Godoy, M.; Ibrahim, S.H.; Mann, J.P.; Memon, I.; Mohan, N.; Mouane, N.; Porta, G.; et al. FISPGHAN statement on the global public health impact of metabolic dysfunction-associated steatotic liver disease. J. Pediatr. Gastroenterol. Nutr. 2025, 80, 397–407. [Google Scholar] [CrossRef]
  29. Unalp-Arida, A.; Ruhl, C.E. Prevalence of metabolic dysfunction-associated steatotic liver disease and fibrosis defined by liver elastography in the United States using National Health and Nutrition examination survey 2017-march 2020 and august 2021-august 2023 data. Hepatology 2025, 82, 1256–1273. [Google Scholar] [CrossRef] [PubMed]
  30. Hwang, S.Y.; Danpanichkul, P.; Agopian, V.; Mehta, N.; Parikh, N.D.; Abou-Alfa, G.K.; Singal, A.G.; Yang, J.D. Hepatocellular carcinoma: Updates on epidemiology, surveillance, diagnosis and treatment. Clin. Mol. Hepatol. 2025, 31, S228–S254. [Google Scholar] [CrossRef] [PubMed]
  31. Wu, Y.; Yu, J.; Song, H.; Lu, D.; Li, Z.; Li, X.; Ding, Z.; Zhou, L.; Ma, J.; Zhang, Y.; et al. Profiling of RNA N6-Methyladenosine methylation reveals the critical role of m6A in betaine alleviating hepatic steatosis. Sci. Rep. 2025, 15, 7298. [Google Scholar] [CrossRef]
  32. Alkhouri, N.; Charlton, M.; Gray, M.; Noureddin, M. The pleiotropic effects of glucagon-like peptide-1 receptor agonists in patients with metabolic dysfunction-associated steatohepatitis: A review for gastroenterologists. Expert Opin. Investig. Drugs 2025, 34, 169–195. [Google Scholar] [CrossRef]
  33. Li, Q.; Xiao, N.; Zhang, H.; Liang, G.; Lin, Y.; Qian, Z.; Yang, X.; Yang, J.; Fu, Y.; Zhang, C.; et al. Systemic aging and aging-related diseases. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2025, 39, e70430. [Google Scholar] [CrossRef]
  34. Kumbaroğlu, B.F.; Balaban, Y.H.; Düger, T. Muscle Strength and Cardiovascular Health in MASLD: A Prospective Study. Medicina 2025, 61, 247. [Google Scholar] [CrossRef]
  35. Barbalho, S.M.; Minniti, G.; Miola, V.F.B.; Haber, J.; Bueno, P.; de Argollo Haber, L.S.; Girio, R.S.J.; Detregiachi, C.R.P.; Dall’Antonia, C.T.; Rodrigues, V.D.; et al. Organokines in COVID-19: A Systematic Review. Cells 2023, 12, 1349. [Google Scholar] [CrossRef]
  36. Laurindo, L.F.; de Maio, M.C.; Barbalho, S.M.; Guiguer, E.L.; Araújo, A.C.; de Alvares Goulart, R.; Flato, U.A.P.; Júnior, E.B.; Detregiachi, C.R.P.; Dos Santos Haber, J.F.; et al. Organokines in Rheumatoid Arthritis: A Critical Review. Int. J. Mol. Sci. 2022, 23, 6193. [Google Scholar] [CrossRef]
  37. Jia, Z.; Wang, Z.; Pan, H.; Zhang, J.; Wang, Q.; Zhou, C.; Liu, J. Crosstalk between fat tissue and muscle, brain, liver, and heart in obesity: Cellular and molecular perspectives. Eur. J. Med. Res. 2024, 29, 637. [Google Scholar] [CrossRef]
  38. Tatsuta, M.; Masaki, T.; Kimura, S.; Sato, Y.; Tomida, A.; Ishikawa, I.; Nakamura, Y.; Takuma, K.; Nakahara, M.; Oura, K.; et al. Efficiency of Skeletal Muscle Mass/Weight Measurement for Distinguishing Metabolic Dysfunction-Associated Steatotic Liver Disease: A Prospective Analysis Using InBody Bioimpedance Devices. Nutrients 2024, 16, 4422. [Google Scholar] [CrossRef] [PubMed]
  39. Real Martinez, Y.; Fernandez-Garcia, C.E.; Fuertes-Yebra, E.; Calvo Soto, M.; Berlana, A.; Barrios, V.; Caldas, M.; Gonzalez Moreno, L.; Garcia-Buey, L.; Molina Baena, B.; et al. Assessment of skeletal muscle alterations and circulating myokines in metabolic dysfunction-associated steatotic liver disease: A cross-sectional study. World J. Gastroenterol. 2025, 31, 100039. [Google Scholar] [CrossRef] [PubMed]
  40. Crişan, D.; Avram, L.; Morariu-Barb, A.; Grapa, C.; Hirişcau, I.; Crăciun, R.; Donca, V.; Nemeş, A. Sarcopenia in MASLD-Eat to Beat Steatosis, Move to Prove Strength. Nutrients 2025, 17, 178. [Google Scholar] [CrossRef] [PubMed]
  41. Zhao, Y.; Li, J.; Ma, A.; Wang, Z.; Ni, Y.; Wu, D.; Zhou, Y.; Zhang, N.; Zhang, L.; Chang, Y.; et al. Irisin alleviates hepatic steatosis by activating the autophagic SIRT3 pathway. Chin. Med. J. 2025. [Google Scholar] [CrossRef] [PubMed]
  42. Faramia, J.; Ostinelli, G.; Drolet-Labelle, V.; Picard, F.; Tchernof, A. Metabolic adaptations after bariatric surgery: Adipokines, myokines and hepatokines. Curr. Opin. Pharmacol. 2020, 52, 67–74. [Google Scholar] [CrossRef]
  43. Sayharman, S.; Belviranlı, M.; Okudan, N. The association between circulating irisin, osteocalcin and FGF21 levels with anthropometric characteristics and blood lipid profile in young obese male subjects. Adv. Med. Sci. 2025, 70, 117–123. [Google Scholar] [CrossRef] [PubMed]
  44. Laurindo, L.F.; Lima, E.P.; Araújo, A.C.; Dogani Rodrigues, V.; Dias, J.A.; Barbosa Tavares Filho, M.; Zuccari, D.; Fornari Laurindo, L.; Miglino, M.A.; Chagas, E.F.B.; et al. Targeting Muscle Regeneration with Small Extracellular Vesicles from Adipose Tissue-Derived Stem Cells-A Review. Cells 2025, 14, 683. [Google Scholar] [CrossRef]
  45. Gonzalez-Gil, A.M.; Elizondo-Montemayor, L. The Role of Exercise in the Interplay between Myokines, Hepatokines, Osteokines, Adipokines, and Modulation of Inflammation for Energy Substrate Redistribution and Fat Mass Loss: A Review. Nutrients 2020, 12, 1899. [Google Scholar] [CrossRef] [PubMed]
  46. de Oliveira Dos Santos, A.R.; de Oliveira Zanuso, B.; Miola, V.F.B.; Barbalho, S.M.; Santos Bueno, P.C.; Flato, U.A.P.; Detregiachi, C.R.P.; Buchaim, D.V.; Buchaim, R.L.; Tofano, R.J.; et al. Adipokines, Myokines, and Hepatokines: Crosstalk and Metabolic Repercussions. Int. J. Mol. Sci. 2021, 22, 2639. [Google Scholar] [CrossRef]
  47. Lu, X.; Chen, Y.; Shi, Y.; Shi, Y.; Su, X.; Chen, P.; Wu, D.; Shi, H. Exercise and exerkines: Mechanisms and roles in anti-aging and disease prevention. Exp. Gerontol. 2025, 200, 112685. [Google Scholar] [CrossRef]
  48. Zuo, Y.; Zhang, C.; Zhou, Y.; Li, H.; Xiao, W.; Herzog, R.W.; Xu, J.; Zhang, J.; Chen, Y.E.; Han, R. Liver-specific in vivo base editing of Angptl3 via AAV delivery efficiently lowers blood lipid levels in mice. Cell Biosci. 2023, 13, 109. [Google Scholar] [CrossRef]
  49. Park, S.Y.; Cho, Y.; Son, S.M.; Hur, J.H.; Kim, Y.; Oh, H.; Lee, H.Y.; Jung, S.; Park, S.; Kim, I.Y.; et al. Activin E is a new guardian protecting against hepatic steatosis via inhibiting lipolysis in white adipose tissue. Exp. Mol. Med. 2025, 57, 466–477. [Google Scholar] [CrossRef]
  50. Zhang, P.; Qi, Z.; Xu, H.; Zhou, L.; Zhao, X.; Zhong, H.; Zhou, W.; Fan, B.; Wu, H.; Ge, J. Fetuin-A increases thrombosis risk in non-alcoholic fatty liver disease by binding to TLR-4 on platelets. Cardiovasc. Res. 2025, 121, 1091–1107. [Google Scholar] [CrossRef]
  51. Tanaka, M.; Tanaka, S.; Kobayashi, R.; Murai, R.; Takahashi, S. Analytical and Clinical Validation of a Plasma Fibroblast Growth Factor 21 ELISA Kit Using an Automated Platform in Steatotic Liver Disease. Biomolecules 2025, 15, 877. [Google Scholar] [CrossRef] [PubMed]
  52. Ye, X.; Toyama, T.; Taguchi, K.; Arisawa, K.; Kaneko, T.; Tsutsumi, R.; Yamamoto, M.; Saito, Y. Sulforaphane decreases serum selenoprotein P levels through enhancement of lysosomal degradation independent of Nrf2. Commun. Biol. 2023, 6, 1060. [Google Scholar] [CrossRef] [PubMed]
  53. Ichimura-Shimizu, M.; Kurrey, K.; Miyata, M.; Dezawa, T.; Tsuneyama, K.; Kojima, M. Emerging Insights into the Role of BDNF on Health and Disease in Periphery. Biomolecules 2024, 14, 444. [Google Scholar] [CrossRef]
  54. Nishikawa, H.; Kim, S.K.; Asai, A. The role of Myokines in liver diseases. Int. J. Mol. Sci. 2025, 26, 1043. [Google Scholar] [CrossRef]
  55. Nagayama, I.; Kamimura, K.; Owaki, T.; Ko, M.; Nagoya, T.; Tanaka, Y.; Ohkoshi, M.; Setsu, T.; Sakamaki, A.; Yokoo, T. Complementary role of peripheral and central autonomic nervous system on insulin-like growth factor-1 activation to prevent fatty liver disease. Hepatol. Int. 2024, 18, 155–167, Correction in Hepatol. Int. 2024, 18, 1067–1069. [Google Scholar] [CrossRef]
  56. Shen, W.; Yang, M.; Chen, H.; He, C.; Li, H.; Yang, X.; Zhuo, J.; Lin, Z.; Hu, Z.; Lu, D. FGF21-mediated autophagy: Remodeling the homeostasis in response to stress in liver diseases. Genes. Dis. 2024, 11, 101027. [Google Scholar] [CrossRef]
  57. Ndakotsu, A.; Nduka, T.C.; Agrawal, S.; Asuka, E. Cirrhotic cardiomyopathy: Comprehensive insights into pathophysiology, diagnosis, and management. Heart Fail. Rev. 2025, 30, 739–748. [Google Scholar] [CrossRef]
  58. Ciałowicz, M.; Woźniewski, M.; Murawska-Ciałowicz, E.; Dzięgiel, P. The Influence of Irisin on Selected Organs-The Liver, Kidneys, and Lungs: The Role of Physical Exercise. Cells 2025, 14, 1228. [Google Scholar] [CrossRef] [PubMed]
  59. Ringleb, M.; Javelle, F.; Haunhorst, S.; Bloch, W.; Fennen, L.; Baumgart, S.; Drube, S.; Reuken, P.A.; Pletz, M.W.; Wagner, H. Beyond muscles: Investigating immunoregulatory myokines in acute resistance exercise–A systematic review and meta-analysis. FASEB J. 2024, 38, e23596. [Google Scholar] [CrossRef]
  60. Pedersen, B.K.; Febbraio, M.A. Muscle as an endocrine organ: Focus on muscle-derived interleukin-6. Physiol. Rev. 2008, 88, 1379–1406. [Google Scholar] [CrossRef]
  61. Taru, V.; Szabo, G.; Mehal, W.; Reiberger, T. Inflammasomes in chronic liver disease: Hepatic injury, fibrosis progression and systemic inflammation. J. Hepatol. 2024, 81, 895–910. [Google Scholar] [CrossRef]
  62. Cepero-Donates, Y.; Lacraz, G.; Ghobadi, F.; Rakotoarivelo, V.; Orkhis, S.; Mayhue, M.; Chen, Y.G.; Rola-Pleszczynski, M.; Menendez, A.; Ilangumaran, S.; et al. Interleukin-15-mediated inflammation promotes non-alcoholic fatty liver disease. Cytokine 2016, 82, 102–111. [Google Scholar] [CrossRef]
  63. Sheptulina, A.F.; Mamutova, E.M.; Elkina, A.Y.; Timofeev, Y.S.; Metelskaya, V.A.; Kiselev, A.R.; Drapkina, O.M. Serum irisin, myostatin, and myonectin correlate with metabolic health markers, liver disease progression, and blood pressure in patients with metabolic dysfunction-associated fatty liver disease and hypertension. Metabolites 2024, 14, 584. [Google Scholar] [CrossRef] [PubMed]
  64. Tanigawa, R.; Nakajima, A.; Eguchi, Y.; Takahashi, H.; Loomba, R.; Suganami, H.; Tanahashi, M.; Arai, H. Effects of Pemafibrate on Serum Carnitine and Plasma Myostatin in Patients With Metabolic Dysfunction Associated Steatotic Liver Disease. JCSM Commun. 2025, 8, e70002. [Google Scholar] [CrossRef]
  65. Li, W.; Zhang, X.; Zhou, Z.; Guo, W.; Wang, M.; Zhou, T.; Liu, M.; Wu, Q.; Dong, N. Cardiac corin and atrial natriuretic peptide regulate liver glycogen metabolism and glucose homeostasis. Cardiovasc. Diabetol. 2024, 23, 383. [Google Scholar] [CrossRef]
  66. Sakae, H.; Tanoue, S.; Mawatari, S.; Oda, K.; Taniyama, O.; Toyodome, A.; Ijuin, S.; Tabu, K.; Kumagai, K.; Ido, A. Fontan-Associated Liver Disease: Predictors of Elevated Liver Stiffness and the Role of Transient Elastography in Long-Term Follow-Up. Cureus 2025, 17, e85336. [Google Scholar] [CrossRef]
  67. Risteska, M.; Vladimirova-Kitova, L.; Andonov, V. Serum NT-ProBNP potential marker of cirrhotic cardiomyopathy. Folia Med. 2022, 64, 740–745. [Google Scholar] [CrossRef]
  68. Mosca, A.; Braghini, M.R.; Andolina, G.; De Stefanis, C.; Cesarini, L.; Pastore, A.; Comparcola, D.; Monti, L.; Francalanci, P.; Balsano, C.; et al. Levels of Growth Differentiation Factor 15 Correlated with Metabolic Dysfunction-Associated Steatotic Liver Disease in Children. Int. J. Mol. Sci. 2025, 26, 6486. [Google Scholar] [CrossRef]
  69. Debroy, P.; Pike, F.; Gawrieh, S.; Corey, K.E.; Hartig, S.; Balasubramanyam, A.; Ailstock, K.; Funderburg, N.; Lake, J.E. Circulating fibroblast growth factor 21 and growth differentiation factor 15 are associated with severity of hepatic steatosis in people with HIV. HIV Med. 2025, 26, 1457–1462. [Google Scholar] [CrossRef]
  70. Huang, M.; Wang, D.; Huang, J.; Bae, A.N.; Xia, Y.; Zhao, X.; Mortaja, M.; Azin, M.; Collier, M.R.; Semenov, Y.R.; et al. Hepatitis B virus promotes liver cancer by modulating the immune response to environmental carcinogens. Nat. Commun. 2025, 16, 5360. [Google Scholar] [CrossRef] [PubMed]
  71. Bucarey, J.L.; Trujillo-González, I.; Paules, E.M.; Espinosa, A. Myokines and Their Potential Protective Role Against Oxidative Stress in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Antioxidants 2024, 13, 1363. [Google Scholar] [CrossRef]
  72. Badmus, O.O.; da Silva, A.A.; Li, X.; Taylor, L.C.; Greer, J.R.; Wasson, A.R.; McGowan, K.E.; Patel, P.R.; Stec, D.E. Cardiac lipotoxicity and fibrosis underlie impaired contractility in a mouse model of metabolic dysfunction-associated steatotic liver disease. FASEB Bioadv. 2024, 6, 131–142. [Google Scholar] [CrossRef]
  73. Shi, Y.; Perez-Bonilla, P.; Chen, X.; Tam, K.; Marshall, M.; Morin, J.; LaViolette, B.; Kue, N.R.; Damilano, F.; Miller, R.A.; et al. Metabolic Syndrome Nonalcoholic Steatohepatitis Male Mouse With Adeno-Associated Viral Renin as a Novel Model for Heart Failure with Preserved Ejection Fraction. J. Am. Heart Assoc. 2024, 13, e035894. [Google Scholar] [CrossRef]
  74. Martin, L.; Maric, D.; Idriss, S.; Delamare, M.; Le Roy, A.; Maaziz, N.; Caillaud, A.; Si-Tayeb, K.; Robriquet, F.; Lenglet, M.; et al. Identification of Hepatic-like EPO as a Cause of Polycythemia. N. Engl. J. Med. 2025, 392, 1684–1697. [Google Scholar] [CrossRef]
  75. Hong, T.; Ge, Z.; Meng, R.; Wang, H.; Zhang, P.; Tang, S.; Lu, J.; Gu, T.; Zhu, D.; Bi, Y. Erythropoietin alleviates hepatic steatosis by activating SIRT1-mediated autophagy. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2018, 1863, 595–603. [Google Scholar] [CrossRef]
  76. Wei, K.; Zhang, Q.; Chen, C.; Yang, Y.; Sun, S.; Wang, L.; Chen, X.; Luo, X.; Chen, Q. Higher Circulating Alpha-klotho Levels Increase All-cause Mortality Through Mediation Effects of Liver Fibrosis: A Second Analysis of National Health and Nutrition Examination Survey. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2025, 80, glaf109. [Google Scholar] [CrossRef]
  77. Meroni, M.; Dongiovanni, P.; Tiano, F.; Piciotti, R.; Alisi, A.; Panera, N. β-Klotho as novel therapeutic target in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): A narrative review. Biomed. Pharmacother. 2024, 180, 117608. [Google Scholar] [CrossRef]
  78. Harris, R.; Cunningham, T.; Yates, A.R.; Phelps, C.; Zepeda-Orozco, D.; Beckman, B.; Kistler, I.; Alexander, R.; Krawczeski, C.D.; Bi, J. Identifying kidney injury via urinary biomarkers after the comprehensive stage II palliation and bidirectional Glenn procedure: A pilot study. Cardiol. Young 2025, 1–9. [Google Scholar] [CrossRef]
  79. Kojima, Y.; Tsuchiya, A.; Mito, M.; Watanabe, Y.; Kawata, Y.; Tominaga, K.; Terai, S. Urinary NGAL in gastrointestinal diseases can be used as an indicator of early infection in addition to acute kidney injury marker. JGH Open 2024, 8, e70009. [Google Scholar] [CrossRef]
  80. Johnson, R.; Shabalala, S.; Mabasa, L.; Kotzé-Hörstmann, L.; Sangweni, N.; Ramharack, P.; Sharma, J.; Pheiffer, C.; Arowolo, A.; Sadie-Van Gijsen, H. Integrated profiling of adiponectin and cytokine signaling pathways in high-fat diet-induced MASLD reveals early markers of disease progression. Sci. Rep. 2025, 15, 19700. [Google Scholar] [CrossRef]
  81. Chen, Q.C.; Cai, W.F.; Ni, Q.; Lin, S.X.; Jiang, C.P.; Yi, Y.K.; Liu, L.; Liu, Q.; Shen, C.Y. A New Perspective on Signaling Pathways and Structure-Activity Relationships of Natural Active Ingredients in Metabolic Crosstalk Between Liver and Brown Adipose Tissue: A Narrative Review. Phytother. Res. PTR 2025, 39, 3062–3090. [Google Scholar] [CrossRef]
  82. Madureira, A.R.; Martins, V.V.P.; Cardoso, J.C.; Torezani-Sales, S.; Miranda, K.O.; Salgado, B.S.; Monteiro, L.N.; Nogueira, B.V.; Leopoldo, A.S.; Lima-Leopoldo, A.P. High-Fat Diet Induces MASLD and Adipose Tissue Changes in Obesity-Resistant Rats. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2025, 59, 434–452. [Google Scholar] [CrossRef]
  83. Casillas-Ramírez, A.; Maroto-Serrat, C.; Sanus, F.; Micó-Carnero, M.; Rojano-Alfonso, C.; Cabrer, M.; Peralta, C. Regulation of Adiponectin and Resistin in Liver Transplantation Protects Grafts from Extended-Criteria Donors. Am. J. Pathol. 2025, 195, 494–527. [Google Scholar] [CrossRef]
  84. Nouri, S.; Navari, M.; Shafiee, R.; Mahmoudi, T.; Rezamand, G.; Asadi, A.; Nobakht, H.; Dabiri, R.; Farahani, H.; Tabaeian, S.P. The rs1862513 promoter variant of resistin gene influences susceptibility to nonalcoholic fatty liver disease. Rev. Assoc. Med. Bras. 2024, 70, e20231537. [Google Scholar] [CrossRef]
  85. Chen, S.; Wu, K.; Ke, Y.; Chen, S.; He, R.; Zhang, Q.; Shen, C.; Li, Q.; Ruan, Y.; Zhu, Y.; et al. Association of circulating visfatin level and metabolic fatty liver disease: An updated meta-analysis and systematic review. Medicine 2024, 103, e39613. [Google Scholar] [CrossRef]
  86. Heo, Y.J.; Choi, S.E.; Lee, N.; Jeon, J.Y.; Han, S.J.; Kim, D.J.; Kang, Y.; Lee, K.W.; Kim, H.J. Visfatin exacerbates hepatic inflammation and fibrosis in a methionine-choline-deficient diet mouse model. J. Gastroenterol. Hepatol. 2021, 36, 2592–2600. [Google Scholar] [CrossRef]
  87. He, X.; Shen, Y.; Ma, X.; Ying, L.; Peng, J.; Pan, X.; Bao, Y.; Zhou, J. The association of serum FGF23 and non-alcoholic fatty liver disease is independent of vitamin D in type 2 diabetes patients. Clin. Exp. Pharmacol. Physiol. 2018, 45, 668–674. [Google Scholar] [CrossRef]
  88. Cao, W.; Xu, Y.; Shen, Y.; Wang, Y.; Ma, X.; Bao, Y. Serum Fibroblast Growth Factor 23 Level and Liver Fat Content in MAFLD: A Community-Based Cohort. Diabetes Metab. Syndr. Obes. 2021, 14, 4135–4143. [Google Scholar] [CrossRef]
  89. Ocker, M. Fibroblast growth factor signaling in non-alcoholic fatty liver disease and non-alcoholic steatohepatitis: Paving the way to hepatocellular carcinoma. World J. Gastroenterol. 2020, 26, 279–290. [Google Scholar] [CrossRef]
  90. Martiniakova, M.; Mondockova, V.; Kovacova, V.; Babikova, M.; Zemanova, N.; Biro, R.; Penzes, N.; Omelka, R. Interrelationships among metabolic syndrome, bone-derived cytokines, and the most common metabolic syndrome-related diseases negatively affecting bone quality. Diabetol. Metab. Syndr. 2024, 16, 217. [Google Scholar] [CrossRef]
  91. Gao, H.; Peng, X.; Li, N.; Gou, L.; Xu, T.; Wang, Y.; Qin, J.; Liang, H.; Ma, P.; Li, S.; et al. Emerging role of liver-bone axis in osteoporosis. J. Orthop. Transl. 2024, 48, 217–231. [Google Scholar] [CrossRef]
  92. Liu, L.; Niu, K.; Yang, Z.; Song, J.; Wei, D.; Zhang, R.; Tao, K. Osteopontin: An indispensable component in common liver, pancreatic, and biliary related disease. J. Cancer Res. Clin. Oncol. 2024, 150, 508. [Google Scholar] [CrossRef]
  93. Nowicki, J.K.; Jakubowska-Pietkiewicz, E. Osteocalcin: Beyond Bones. Endocrinol. Metab. 2024, 39, 399–406. [Google Scholar] [CrossRef]
  94. Jiang, H.; Li, D.; Han, Y.; Li, N.; Tao, X.; Liu, J.; Zhang, Z.; Yu, Y.; Wang, L.; Yu, S.; et al. The role of sclerostin in lipid and glucose metabolism disorders. Biochem. Pharmacol. 2023, 215, 115694. [Google Scholar] [CrossRef]
  95. Han, X.; Zhang, G.S.; Li, Q.R.; Zhang, Z. Current approach to the diagnosis of sarcopenia in cardiovascular diseases. Front. Nutr. 2024, 11, 1422663. [Google Scholar] [CrossRef]
  96. Petermann-Rocha, F.; Balntzi, V.; Gray, S.R.; Lara, J.; Ho, F.K.; Pell, J.P.; Celis-Morales, C. Global prevalence of sarcopenia and severe sarcopenia: A systematic review and meta-analysis. J. Cachexia Sarcopenia Muscle 2022, 13, 86–99. [Google Scholar] [CrossRef]
  97. Cruz-Jentoft, A.J.; Baeyens, J.P.; Bauer, J.M.; Boirie, Y.; Cederholm, T.; Landi, F.; Martin, F.C.; Michel, J.P.; Rolland, Y.; Schneider, S.M.; et al. Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing 2010, 39, 412–423. [Google Scholar] [CrossRef]
  98. Sayer, A.A.; Cruz-Jentoft, A. Sarcopenia definition, diagnosis and treatment: Consensus is growing. Age Ageing 2022, 51, afac220. [Google Scholar] [CrossRef]
  99. Benz, E.; Pinel, A.; Guillet, C.; Capel, F.; Pereira, B.; De Antonio, M.; Pouget, M.; Cruz-Jentoft, A.J.; Eglseer, D.; Topinkova, E.; et al. Sarcopenia and Sarcopenic Obesity and Mortality Among Older People. JAMA Netw. Open 2024, 7, e243604. [Google Scholar] [CrossRef]
  100. Cho, M.R.; Lee, S.; Song, S.K. A Review of Sarcopenia Pathophysiology, Diagnosis, Treatment and Future Direction. J. Korean Med. Sci. 2022, 37, e146. [Google Scholar] [CrossRef]
  101. Lian, R.; Liu, Q.; Jiang, G.; Zhang, X.; Tang, H.; Lu, J.; Yang, M. Blood biomarkers for sarcopenia: A systematic review and meta-analysis of diagnostic test accuracy studies. Ageing Res. Rev. 2024, 93, 102148. [Google Scholar] [CrossRef]
  102. Goisser, S.; Kob, R.; Sieber, C.C.; Bauer, J.M. Diagnosis and therapy of sarcopenia-an update. Der Internist 2019, 60, 141–148. [Google Scholar] [CrossRef]
  103. Affourtit, C.; Carré, J.E. Mitochondrial involvement in sarcopenia. Acta Physiol. 2024, 240, e14107. [Google Scholar] [CrossRef]
  104. Jimenez-Gutierrez, G.E.; Martínez-Gómez, L.E.; Martínez-Armenta, C.; Pineda, C.; Martínez-Nava, G.A.; Lopez-Reyes, A. Molecular Mechanisms of Inflammation in Sarcopenia: Diagnosis and Therapeutic Update. Cells 2022, 11, 2359. [Google Scholar] [CrossRef]
  105. Evans, W.J.; Guralnik, J.; Cawthon, P.; Appleby, J.; Landi, F.; Clarke, L.; Vellas, B.; Ferrucci, L.; Roubenoff, R. Sarcopenia: No consensus, no diagnostic criteria, and no approved indication-How did we get here? GeroScience 2024, 46, 183–190. [Google Scholar] [CrossRef]
  106. Wang, J.; Jia, D.; Zhang, Z.; Wang, D. Exerkines and Sarcopenia: Unveiling the Mechanism Behind Exercise-Induced Mitochondrial Homeostasis. Metabolites 2025, 15, 59. [Google Scholar] [CrossRef]
  107. Feng, Z.; Zhao, F.; Wang, Z.; Tang, X.; Xie, Y.; Qiu, L. The relationship between sarcopenia and metabolic dysfunction-associated fatty liver disease among the young and middle-aged populations. BMC Gastroenterol. 2024, 24, 111. [Google Scholar] [CrossRef]
  108. Musio, A.; Perazza, F.; Leoni, L.; Stefanini, B.; Dajti, E.; Menozzi, R.; Petroni, M.L.; Colecchia, A.; Ravaioli, F. Osteosarcopenia in NAFLD/MAFLD: An Underappreciated Clinical Problem in Chronic Liver Disease. Int. J. Mol. Sci. 2023, 24, 7517. [Google Scholar] [CrossRef]
  109. Hao, X.Y.; Zhang, K.; Huang, X.Y.; Yang, F.; Sun, S.Y. Muscle strength and non-alcoholic fatty liver disease/metabolic-associated fatty liver disease. World J. Gastroenterol. 2024, 30, 636–643. [Google Scholar] [CrossRef]
  110. Zhou, D.; Shi, Y.; Zhang, D.; Zuo, J.; Zeng, C.; Mamtawla, G.; Huang, L.; Gao, X.; Zhang, L.; Wang, X. Liver-secreted FGF21 induces sarcopenia by inhibiting satellite cell myogenesis via klotho beta in decompensated cirrhosis. Redox Biol. 2024, 76, 103333. [Google Scholar] [CrossRef]
  111. Lee, W.H.; Bates, E.A.; Kipp, Z.A.; Pauss, S.N.; Martinez, G.J.; Blair, C.A.; Hinds, T.D., Jr. Insulin receptor responsiveness governs TGFβ-induced hepatic stellate cell activation: Insulin resistance instigates liver fibrosis. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2025, 39, e70427. [Google Scholar] [CrossRef]
  112. Zhu, X.; Zeng, C.; Yu, B. White adipose tissue in metabolic associated fatty liver disease. Clin. Res. Hepatol. Gastroenterol. 2024, 48, 102336. [Google Scholar] [CrossRef]
  113. Fernández-Mincone, T.; Contreras-Briceño, F.; Espinosa-Ramírez, M.; García-Valdés, P.; López-Fuenzalida, A.; Riquelme, A.; Arab, J.P.; Cabrera, D.; Arrese, M.; Barrera, F. Nonalcoholic fatty liver disease and sarcopenia: Pathophysiological connections and therapeutic implications. Expert Rev. Gastroenterol. Hepatol. 2020, 14, 1141–1157. [Google Scholar] [CrossRef]
  114. Luo, W.; Zhou, Y.; Tang, Q.; Wang, Y.; Liu, Y.; Ai, L. Downhill running and caloric restriction attenuate insulin resistance associated skeletal muscle atrophy via the promotion of M2-like macrophages through TRIB3-AKT pathway. Free Radic. Biol. Med. 2024, 210, 271–285. [Google Scholar] [CrossRef]
  115. Yang, Y.; Wang, T.T.; Xie, H.A.; Hu, P.P.; Li, P. Experimental cell models of insulin resistance: Overview and appraisal. Front. Endocrinol. 2024, 15, 1469565. [Google Scholar] [CrossRef]
  116. Zhao, R.; Ji, X.; Wang, F. Association of triglyceride glucose-related obesity indices with sarcopenia among U.S. adults: A cross-sectional study from the National Health and Nutrition Examination Survey. Sci. Rep. 2025, 15, 574. [Google Scholar] [CrossRef]
  117. Lee, J.H.; Jun, H.S. Role of Myokines in Regulating Skeletal Muscle Mass and Function. Front. Physiol. 2019, 10, 42. [Google Scholar] [CrossRef]
  118. Santos, J.; Maio, M.C.; Lemes, M.A.; Laurindo, L.F.; Haber, J.; Bechara, M.D.; Prado, P.S.D., Jr.; Rauen, E.C.; Costa, F.; Pereira, B.C.A.; et al. Non-Alcoholic Steatohepatitis (NASH) and Organokines: What Is Now and What Will Be in the Future. Int. J. Mol. Sci. 2022, 23, 498. [Google Scholar] [CrossRef]
  119. Minniti, G.; Pescinini-Salzedas, L.M.; Minniti, G.; Laurindo, L.F.; Barbalho, S.M.; Vargas Sinatora, R.; Sloan, L.A.; Haber, R.S.A.; Araújo, A.C.; Quesada, K.; et al. Organokines, Sarcopenia, and Metabolic Repercussions: The Vicious Cycle and the Interplay with Exercise. Int. J. Mol. Sci. 2022, 23, 13452. [Google Scholar] [CrossRef]
  120. Barbalho, S.M.; Flato, U.A.P.; Tofano, R.J.; Goulart, R.A.; Guiguer, E.L.; Detregiachi, C.R.P.; Buchaim, D.V.; Araújo, A.C.; Buchaim, R.L.; Reina, F.T.R.; et al. Physical Exercise and Myokines: Relationships with Sarcopenia and Cardiovascular Complications. Int. J. Mol. Sci. 2020, 21, 3607. [Google Scholar] [CrossRef]
  121. Ferenc, K.; Jarmakiewicz-Czaja, S.; Filip, R. What Does Sarcopenia Have to Do with Nonalcoholic Fatty Liver Disease? Life 2023, 14, 37. [Google Scholar] [CrossRef]
  122. Fouda, S.; Jeeyavudeen, M.S.; Pappachan, J.M.; Jayanthi, V. Pathobiology of Metabolic-Associated Fatty Liver Disease. Endocrinol. Metab. Clin. N. Am. 2023, 52, 405–416. [Google Scholar] [CrossRef]
  123. Seo, J.Y.; Cho, E.J.; Kim, M.J.; Kwak, M.S.; Yang, J.I.; Chung, S.J.; Yim, J.Y.; Yoon, J.W.; Chung, G.E. The relationship between metabolic dysfunction-associated fatty liver disease and low muscle mass in an asymptomatic Korean population. J. Cachexia Sarcopenia Muscle 2022, 13, 2953–2960. [Google Scholar] [CrossRef]
  124. Xia, N.; Wang, J.; Lu, Q.; Fan, X.; Jiang, Z.; Yu, Q. Relationship between metabolic dysfunction-associated steatotic liver disease and sarcopenia: A systematic review and meta-analyses. Clin. Nutr. ESPEN 2025, 68, 679–690. [Google Scholar] [CrossRef]
  125. Malik, A.; Javaid, S.; Malik, M.I.; Qureshi, S. Relationship between sarcopenia and metabolic dysfunction-associated steatotic liver disease (MASLD): A systematic review and meta-analysis. Ann. Hepatol. 2024, 29, 101544. [Google Scholar] [CrossRef]
  126. Polyzos, S.A.; Vachliotis, I.D.; Mantzoros, C.S. Sarcopenia, sarcopenic obesity and nonalcoholic fatty liver disease. Metab. Clin. Exp. 2023, 147, 155676. [Google Scholar] [CrossRef]
  127. Duarte, M.P.; Almeida, L.S.; Neri, S.G.R.; Oliveira, J.S.; Wilkinson, T.J.; Ribeiro, H.S.; Lima, R.M. Prevalence of sarcopenia in patients with chronic kidney disease: A global systematic review and meta-analysis. J. Cachexia Sarcopenia Muscle 2024, 15, 501–512. [Google Scholar] [CrossRef]
  128. Chen, M.; Cao, Y.; Ji, G.; Zhang, L. Lean nonalcoholic fatty liver disease and sarcopenia. Front. Endocrinol. 2023, 14, 1217249. [Google Scholar] [CrossRef]
  129. Ramírez-Mejía, M.M.; Qi, X.; Abenavoli, L.; Barranco-Fragoso, B.; Barbalho, S.M.; Méndez-Sánchez, N. NAFLD-MASLD-MAFLD continuum: A swinging pendulum? Ann. Hepatol. 2024, 29, 101526. [Google Scholar] [CrossRef]
  130. Nakamura, A.; Yoshimura, T.; Ichikawa, T. Liver Disease-Related Sarcopenia: A Predictor of Poor Prognosis by Accelerating Hepatic Decompensation in Advanced Chronic Liver Disease. Cureus 2023, 15, e49078. [Google Scholar] [CrossRef]
  131. Liang, Y.; Ye, X.; Pan, M.; Chen, Y.; Yuan, Y.; Luo, L. Impact of steatotic liver disease subtypes, sarcopenia, and fibrosis on all-cause and cause-specific mortality: A 15.7-year cohort study. BMC Gastroenterol. 2025, 25, 75. [Google Scholar] [CrossRef] [PubMed]
  132. Zhang, F.; Liu, L.; Li, W. Correlation of sarcopenia with progression of liver fibrosis in patients with metabolic dysfunction-associated steatotic liver disease: A study from two cohorts in China and the United States. Nutr. J. 2025, 24, 6. [Google Scholar] [CrossRef]
  133. Tanigawa, R.; Nakajima, A.; Eguchi, Y.; Takahashi, H.; Loomba, R.; Suganami, H.; Tanahashi, M.; Saito, A.; Iida, Y.; Yamashita, S. Effects of Pemafibrate on LDL-C and Related Lipid Markers in Patients with MASLD: A Sub-Analysis of the PEMA-FL Study. J. Atheroscler. Thromb. 2025, 32, 823–839. [Google Scholar] [CrossRef]
  134. Ananthakrishnan, A.I.; Mahin, A.; Prasad, T.S.K.; Abhinand, C.S. Transcriptome Profiling and Viral-Human Interactome Insights Into HBV-Driven Oncogenic Alterations in Hepatocellular Carcinoma. Microbiol. Immunol. 2025, 69, 326–338. [Google Scholar] [CrossRef]
  135. Fodor Duric, L.; Belčić, V.; Oberiter Korbar, A.; Ćurković, S.; Vujicic, B.; Gulin, T.; Muslim, J.; Gulin, M.; Grgurević, M.; Catic Cuti, E. The Role of SHBG as a Marker in Male Patients with Metabolic-Associated Fatty Liver Disease: Insights into Metabolic and Hormonal Status. J. Clin. Med. 2024, 13, 7717. [Google Scholar] [CrossRef]
  136. Han, E.; Han, K.D.; Lee, Y.H.; Kim, K.S.; Hong, S.; Park, J.H.; Park, C.Y. Association of temporal MASLD with type 2 diabetes, cardiovascular disease and mortality. Cardiovasc. Diabetol. 2025, 24, 289. [Google Scholar] [CrossRef]
  137. Wu, W.; Cheng, J.; Hou, Z.; Yan, Q.; Wang, X.; He, J.; Zhu, Y.; Li, J. Association between the dietary index for gut microbiota and all-cause/cardiovascular mortality in patients with metabolic dysfunction-associated steatotic liver disease. Diabetol. Metab. Syndr. 2025, 17, 263. [Google Scholar] [CrossRef]
  138. Baars, T.; Gieseler, R.K.; Patsalis, P.C.; Canbay, A. Towards harnessing the value of organokine crosstalk to predict the risk for cardiovascular disease in non-alcoholic fatty liver disease. Metab. Clin. Exp. 2022, 130, 155179. [Google Scholar] [CrossRef] [PubMed]
  139. Mahmoud, M.C.; Abdelmalek, M.F. The Liver in Diabetes and Metabolic Syndrome. Clin. Liver Dis. 2025, 29, 407–429. [Google Scholar] [CrossRef]
  140. Jiang, Z.; Wu, S.; Zhou, S.; Zheng, H.; Bai, Y.; Zhang, Y.; Yao, M. Photobiomodulation mediates endoplasmic reticulum-mitochondria contact and ameliorates lipotoxicity in MASLD via Mfn2 upregulation. J. Photochem. Photobiol. B 2025, 270, 113209. [Google Scholar] [CrossRef] [PubMed]
  141. Laurindo, L.F.; Dogani Rodrigues, V.; de Lima, E.P.; Leme Boaro, B.; Mendes Peloi, J.M.; Ferraroni Sanches, R.C.; Penteado Detregiachi, C.R.; José Tofano, R.; Angelica Miglino, M.; Sloan, K.P.; et al. Targeting Atherosclerosis via NEDD4L Signaling-A Review of the Current Literature. Biology 2025, 14, 220. [Google Scholar] [CrossRef]
  142. Bril, F.; Elbert, A. Metabolic dysfunction-associated steatotic liver disease and urinary system cancers: Mere coincidence or reason for concern? Metab. Clin. Exp. 2025, 162, 156066. [Google Scholar] [CrossRef]
  143. Kang, M.K.; Song, J.E.; Loomba, R.; Park, S.Y.; Tak, W.Y.; Kweon, Y.O.; Lee, Y.R.; Park, J.G. Comparative associations of MASLD and MAFLD with the presence and severity of coronary artery calcification. Sci. Rep. 2024, 14, 22917. [Google Scholar] [CrossRef] [PubMed]
  144. Bagheri Lankarani, K.; Jamalinia, M.; Zare, F.; Heydari, S.T.; Ardekani, A.; Lonardo, A. Liver-Kidney-Metabolic Health, Sex, and Menopause Impact Total Scores and Monovessel vs. Multivessel Coronary Artery Calcification. Adv. Ther. 2025, 42, 1729–1744. [Google Scholar] [CrossRef] [PubMed]
  145. Sandireddy, R.; Sakthivel, S.; Gupta, P.; Behari, J.; Tripathi, M.; Singh, B.K. Systemic impacts of metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH) on heart, muscle, and kidney related diseases. Front. Cell Dev. Biol. 2024, 12, 1433857. [Google Scholar] [CrossRef] [PubMed]
  146. Targher, G.; Bertolini, L.; Poli, F.; Rodella, S.; Scala, L.; Tessari, R.; Zenari, L.; Falezza, G. Nonalcoholic fatty liver disease and risk of future cardiovascular events among type 2 diabetic patients. Diabetes 2005, 54, 3541–3546. [Google Scholar] [CrossRef]
  147. Tana, C.; Ballestri, S.; Ricci, F.; Di Vincenzo, A.; Ticinesi, A.; Gallina, S.; Giamberardino, M.A.; Cipollone, F.; Sutton, R.; Vettor, R. Cardiovascular risk in non-alcoholic fatty liver disease: Mechanisms and therapeutic implications. Int. J. Environ. Res. Public Health 2019, 16, 3104. [Google Scholar] [CrossRef]
  148. Simon, T.G.; Roelstraete, B.; Khalili, H.; Hagström, H.; Ludvigsson, J.F. Mortality in biopsy-confirmed nonalcoholic fatty liver disease: Results from a nationwide cohort. Gut 2021, 70, 1375–1382. [Google Scholar] [CrossRef]
  149. Capone, F.; Vacca, A.; Bidault, G.; Sarver, D.; Kaminska, D.; Strocchi, S.; Vidal-Puig, A.; Greco, C.M.; Lusis, A.J.; Schiattarella, G.G. Decoding the Liver-Heart Axis in Cardiometabolic Diseases. Circ. Res. 2025, 136, 1335–1362. [Google Scholar] [CrossRef]
  150. Xie, W.; Gan, J.; Zhou, X.; Tian, H.; Pan, X.; Liu, W.; Li, X.; Du, J.; Xu, A.; Zheng, M. Myocardial infarction accelerates the progression of MASH by triggering immunoinflammatory response and induction of periostin. Cell Metab. 2024, 36, 1269–1286.e1269. [Google Scholar] [CrossRef]
  151. Sun, J.-Y.; Du, L.-J.; Shi, X.-R.; Zhang, Y.-Y.; Liu, Y.; Wang, Y.-L.; Chen, B.-Y.; Liu, T.; Zhu, H.; Liu, Y. An IL-6/STAT3/MR/FGF21 axis mediates heart-liver cross-talk after myocardial infarction. Sci. Adv. 2023, 9, eade4110. [Google Scholar] [CrossRef]
  152. Ahmed, A.E.; Halawany, K.W.; Alyahyawi, F.S.; Khormi, A.H.; AlQibti, H.M.; Saifain, O.M.; Alqarni, T.M.; Alqurashi, J.S.; Alabdali, R.M.; Alowaimer, S.T. Effect of Blood Pressure Control on Cardiovascular Events in Patients with Chronic Kidney Disease: A Systematic Review. Cureus 2025, 17, e86230. [Google Scholar] [CrossRef]
  153. Capalbo, O.; Giuliani, S.; Ferrero-Fernández, A.; Casciato, P.; Musso, C.G. Kidney–liver pathophysiological crosstalk: Its characteristics and importance. Int. Urol. Nephrol. 2019, 51, 2203–2207. [Google Scholar] [CrossRef] [PubMed]
  154. Bae, H.; Nguyen, C.M.; Ruiz-Orera, J.; Mills, N.L.; Snyder, M.P.; Jang, C.; Shah, S.H.; Hübner, N.; Seldin, M. Emerging Technologies and Future Directions in Interorgan Crosstalk Cardiometabolic Research. Circ. Res. 2025, 136, 1494–1506. [Google Scholar] [CrossRef] [PubMed]
  155. Ahmed, Y.R.; El-Hagrassi, A.M.; Nasr, N.N.; Abdallah, W.E.; Hamed, M.A. Chemical Composition and Therapeutic Potential of Syngonium podophyllum L. Leaves against Hypercholesterolemia in Rats: Liver, Kidney, and Heart Crosstalk. Curr. Bioact. Compd. 2024, 20, 48–63. [Google Scholar] [CrossRef]
  156. Rad, N.K.; Heydari, Z.; Tamimi, A.H.; Zahmatkesh, E.; Shpichka, A.; Barekat, M.; Timashev, P.; Hossein-Khannazer, N.; Hassan, M.; Vosough, M. Review on kidney-liver crosstalk: Pathophysiology of their disorders. Cell J. 2024, 26, 98–111. [Google Scholar]
  157. Li, Z.; Zhang, X.; Zhu, W.; Xu, Q.; Yu, C.; Zhao, Q.; Siebel, S.; Zhang, C.; Costa Lima, B.G.; Li, X. GDF15 is a critical renostat in the defense against hypoglycemia. Biorxiv 2023. [Google Scholar] [CrossRef]
  158. Waitupu, A.; Pratiwi, L.; Sutanto, H.; Santoso, D.; Hertanto, D.M. Molecular pathophysiology of chronic kidney disease–mineral and bone disorder: Focus on the fibroblast growth factor 23–Klotho axis and bone turnover dynamics. Exp. Physiol. 2025. [Google Scholar] [CrossRef]
  159. Atić, A.; Gulin, M.; Hudolin, T.; Mokos, I.; Kaštelan, Ž.; Ćorić, M.; Bašić-Jukić, N. Association of the Expression of Bone Morphogenetic Protein Subtypes in Recipient Epigastric Arteries and Long-Term Outcomes After Kidney Transplantation. Turk. J. Nephrol. 2025, 34, 206–212. [Google Scholar] [CrossRef]
  160. Chou, Y.-H.; Pan, S.-Y.; Shih, H.-M.; Lin, S.-L. Update of pericytes function and their roles in kidney diseases. J. Formos. Med. Assoc. 2024, 123, 307–317. [Google Scholar] [CrossRef]
  161. Castillo, R.F.; Pérez, R.G.; González, A.L. Beneficial effects of physical exercise on the osteo-renal Klotho-FGF-23 axis in Chronic Kidney Disease: A systematic review with meta-analysis. Int. J. Med. Sci. 2024, 21, 332. [Google Scholar] [CrossRef]
  162. Taskapan, H.; Mahdavi, S.; Bellasi, A.; Martin, S.; Kuvadia, S.; Patel, A.; Taskapan, B.; Tam, P.; Sikaneta, T. Ethnic and seasonal variations in FGF-23 and markers of chronic kidney disease–mineral and bone disorder. Clin. Kidney J. 2024, 17, sfae188. [Google Scholar] [CrossRef]
  163. Ren, B.; Liang, J.; Zhang, J.; Yang, L.; Wei, X.; Guo, M.; Wu, W.; Ding, X. Clinical value of serum Klotho protein in patients with acute traumatic brain injury complicated by acute kidney injury. Sci. Rep. 2025, 15, 20468. [Google Scholar] [CrossRef] [PubMed]
  164. Peng, X.; Li, S.; Song, Y.; Luo, Y.; Liu, S.; Zhao, T. Association between the serum Klotho levels and diabetic kidney disease: Results from the NHANES 2007-2016 and Mendelian randomization study. J. Diabetes Metab. Disord. 2025, 24, 156. [Google Scholar] [CrossRef] [PubMed]
  165. Wang, Y.; Zhang, Y.; Gao, M.; Chen, Z.; Lu, J.; Li, Y.; Di, Y.; Zhao, Y.; Liu, B.; Tang, R. Lipocalin-2 promotes CKD vascular calcification by aggravating VSMCs ferroptosis through NCOA4/FTH1-mediated ferritinophagy. Cell Death Dis. 2024, 15, 865. [Google Scholar] [PubMed]
  166. Bingül, İ.; Kalayci, R.; Tekkeşin, M.S.; Olgac, V.; Bekpinar, S.; Uysal, M. Chenodeoxycholic acid alleviated the cyclosporine-induced nephrotoxicity by decreasing oxidative stress and suppressing renin-angiotensin system through AT2R and ACE2 mRNA upregulation in rats. J. Mol. Histol. 2025, 56, 23. [Google Scholar] [CrossRef]
  167. Weng, S.-E.; Hsu, W.-T.; Hsiao, F.-Y.; Lee, C.-M. Non-steroidal anti-inflammatory drugs, renin-angiotensin system blockade or diuretics and risk of acute kidney injury: A case-crossover study. Arch. Gerontol. Geriatr. 2024, 123, 105394. [Google Scholar] [CrossRef]
  168. Guerrache, A.; Micheau, O. TNF-related apoptosis-inducing ligand: Non-apoptotic signalling. Cells 2024, 13, 521. [Google Scholar] [CrossRef]
  169. Wen, Y.; Crowley, S.D. hypertension. Renal effects of cytokines in hypertension. Adv. Exp. Med. Biol. 2018, 27, 70–76. [Google Scholar]
  170. Momper, J.D.; Nigam, S.K. Developmental regulation of kidney and liver solute carrier and ATP-binding cassette drug transporters and drug metabolizing enzymes: The role of remote organ communication. Expert Opin. Drug Metab. Toxicol. 2018, 14, 561–570. [Google Scholar] [CrossRef]
  171. Nadeem, A.; Ahmad, S.F.; Al-Harbi, N.O.; Ibrahim, K.E.; Sarawi, W.; Attia, S.M.; Alasmari, A.F.; Alqarni, S.A.; Alfradan, A.S.; Bakheet, S.A.; et al. Role of ITK signaling in acute kidney injury in mice: Amelioration of acute kidney injury associated clinical parameters and attenuation of inflammatory transcription factor signaling in CD4+ T cells by ITK inhibition. Int. Immunopharmacol. 2021, 99, 108028. [Google Scholar] [CrossRef]
  172. Mohamadi Yarijani, Z.; Najafi, H. Effects of Peptides and Bioactive Peptides on Acute Kidney Injury: A Review Study. Iran. Biomed. J. 2025, 29, 90–103. Available online: https://pmc.ncbi.nlm.nih.gov/articles/PMC12394725/ (accessed on 20 November 2025).
  173. Zhang, Y.Q.; Peng, W.X.; Li, Y.F.; Lu, P.; Liu, H.; Liu, W.G. GATA1 Transcriptionally Upregulates LMCD1, Promoting Ferroptosis in Sepsis-Associated Acute Kidney Injury Through the Hippo/YAP Pathway. Kaohsiung J. Med. Sci. 2025, 41, e70071. [Google Scholar] [CrossRef] [PubMed]
  174. Katopodis, P.; Pappas, E.M.; Katopodis, K.P. Acid-base abnormalities and liver dysfunction. Ann. Hepatol. 2022, 27, 100675. [Google Scholar] [CrossRef]
  175. Dejong, C.; Deutz, N.; Soeters, P. Ammonia and glutamine metabolism during liver insufficiency: The role of kidney and brain in interorgan nitrogen exchange. Scand. J. Gastroenterol. 1996, 31, 61–77. [Google Scholar] [CrossRef] [PubMed]
  176. Ballmer, P.; McNurlan, M.; Hulter, H.; Anderson, S.E.; Garlick, P.; Krapf, R. Chronic metabolic acidosis decreases albumin synthesis and induces negative nitrogen balance in humans. J. Clin. Investig. 1995, 95, 39–45. [Google Scholar] [CrossRef]
  177. HoleČek, M.; Šafránek, R.; Ryšavá, R.; KadlČíková, J.; Šprongl, L. Acute effects of acidosis on protein and amino acid metabolism in perfused rat liver. Int. J. Exp. Pathol. 2003, 84, 185–190. [Google Scholar] [CrossRef] [PubMed]
  178. Hoste, E.; De Corte, W. Clinical consequences of acute kidney injury. Contrib. Nephrol. 2011, 174, 56–64. [Google Scholar]
  179. Patidar, K.R.; Naved, M.A.; Grama, A.; Adibuzzaman, M.; Ali, A.A.; Slaven, J.E.; Desai, A.P.; Ghabril, M.S.; Nephew, L.; Chalasani, N. Acute kidney disease is common and associated with poor outcomes in patients with cirrhosis and acute kidney injury. J. Hepatol. 2022, 77, 108–115. [Google Scholar] [CrossRef]
  180. Tonon, M.; Rosi, S.; Gambino, C.G.; Piano, S.; Calvino, V.; Romano, A.; Martini, A.; Pontisso, P.; Angeli, P. Natural history of acute kidney disease in patients with cirrhosis. J. Hepatol. 2021, 74, 578–583. [Google Scholar] [CrossRef]
  181. Kazory, A.; Ronco, C. Hepatorenal syndrome or hepatocardiorenal syndrome: Revisiting basic concepts in view of emerging data. Cardiorenal Med. 2019, 9, 1–7. [Google Scholar] [CrossRef]
  182. Schulz, M.; Diehl, V.; Trebicka, J.; Wygrecka, M.; Schaefer, L. Biglycan: A regulator of hepatorenal inflammation and autophagy. Matrix. Biol. 2021, 100–101, 150–161. [Google Scholar] [CrossRef]
  183. Fede, G.; Privitera, G.; Tomaselli, T.; Spadaro, L.; Purrello, F. Cardiovascular dysfunction in patients with liver cirrhosis. Ann. Gastroenterol. Q. Publ. Hell. Soc. Gastroenterol. 2015, 28, 31. [Google Scholar]
  184. Chahal, D.; Liu, H.; Shamatutu, C.; Sidhu, H.; Lee, S.S.; Marquez, V. comprehensive analysis of cirrhotic cardiomyopathy. Aliment. Pharmacol. Ther. 2021, 53, 985–998. [Google Scholar] [CrossRef] [PubMed]
  185. Chung, H.S.; Choi, K.M. Organokines in disease. Adv. Clin. Chem. 2020, 94, 261–321. [Google Scholar] [CrossRef] [PubMed]
  186. Gianopoulos, I.; Mantzoros, C.S.; Daskalopoulou, S.S. Adiponectin and adiponectin receptors in atherosclerosis. Endocr. Rev. 2025, 46, 1–25. [Google Scholar] [CrossRef]
  187. Bashandy, S.A.; Elbaset, M.A.; Ibrahim, F.A.; Abdelrahman, S.S.; Moussa, S.A.A.; El-Seidy, A.M. Management of cardiovascular disease by cerium oxide nanoparticles via alleviating oxidative stress and adipokine abnormalities. Sci. Rep. 2025, 15, 5709. [Google Scholar] [CrossRef]
  188. Zadgaonkar, U. The Interplay Between Adipokines and Body Composition in Obesity and Metabolic Diseases. Cureus 2025, 17. [Google Scholar] [CrossRef]
  189. Asbaghi, O.; Kelishadi, M.R.; Larky, D.A.; Bagheri, R.; Amirani, N.; Goudarzi, K.; Kargar, F.; Ghanavati, M.; Zamani, M. The effects of green tea extract supplementation on body composition, obesity-related hormones and oxidative stress markers: A grade-assessed systematic review and dose–response meta-analysis of randomised controlled trials. Br. J. Nutr. 2024, 131, 1125–1157. [Google Scholar] [CrossRef]
  190. Zhao, K.; Zhang, H.; Ding, W.; Yu, X.; Hou, Y.; Liu, X.; Li, X.; Wang, X. Adipokines regulate the development and progression of MASLD through organellar oxidative stress. Hepatol. Commun. 2025, 9, e0639. [Google Scholar] [CrossRef]
  191. Soltani, S.; Meshkini, F.; Torabinasab, K.; Razmpoosh, E.; Toupchian, O.; Zeraattalab-Motlagh, S.; Hemmati, A.; Sadat Sangsefidi, Z.; Abdollahi, S. Low fat-diet and circulating adipokines concentrations: A systematic review and meta-analysis of randomized controlled trials. Diabetol. Metab. Syndr. 2025, 17, 152. [Google Scholar] [CrossRef]
  192. Tilg, H.; Ianiro, G.; Gasbarrini, A.; Adolph, T.E. Adipokines: Masterminds of metabolic inflammation. Nat. Rev. Immunol. 2024, 1–16. [Google Scholar] [CrossRef]
  193. Turner, L.; Charrouf, R.; Martínez-Vizcaíno, V.; Hutchison, A.; Heilbronn, L.K.; Fernández-Rodríguez, R. The effects of time-restricted eating versus habitual diet on inflammatory cytokines and adipokines in the general adult population: A systematic review with meta-analysis. Am. J. Clin. Nutr. 2024, 119, 206–220. [Google Scholar] [CrossRef]
  194. Gao, D.; Bing, C.; Griffiths, H.R. Disrupted adipokine secretion and inflammatory responses in human adipocyte hypertrophy. Adipocyte 2025, 14, 2485927. [Google Scholar] [CrossRef] [PubMed]
  195. Duwaerts, C.C.; Maher, J.J. Macronutrients and the adipose-liver axis in obesity and fatty liver. Cell. Mol. Gastroenterol. Hepatol. 2019, 7, 749–761. [Google Scholar] [CrossRef]
  196. Martínez-Uña, M.; López-Mancheño, Y.; Diéguez, C.; Fernández-Rojo, M.A.; Novelle, M.G. Unraveling the role of leptin in liver function and its relationship with liver diseases. Int. J. Mol. Sci. 2020, 21, 9368. [Google Scholar] [CrossRef]
  197. De la Cruz-Color, L.; Dominguez-Rosales, J.A.; Maldonado-González, M.; Ruíz-Madrigal, B.; Sánchez Muñoz, M.P.; Zaragoza-Guerra, V.A.; Espinoza-Padilla, V.H.; Ruelas-Cinco, E.d.C.; Ramírez-Meza, S.M.; Torres Baranda, J.R. Evidence that peripheral leptin resistance in omental adipose tissue and liver correlates with MASLD in humans. Int. J. Mol. Sci. 2024, 25, 6420. [Google Scholar] [CrossRef]
  198. Song, P.; Kwon, Y.; Joo, J.Y.; Kim, D.G.; Yoon, J.H. Secretomics to Discover Regulators in Diseases. Int. J. Mol. Sci. 2019, 20, 3893. [Google Scholar] [CrossRef]
  199. Kim, B.; Ronaldo, R.; Kweon, B.-N.; Yoon, S.; Park, Y.; Baek, J.-H.; Lee, J.M.; Hyun, C.-K. Mesenchymal Stem Cell-Derived Exosomes Attenuate Hepatic Steatosis and Insulin Resistance in Diet-Induced Obese Mice by Activating the FGF21-Adiponectin Axis. Int. J. Mol. Sci. 2024, 25, 10447. [Google Scholar] [CrossRef] [PubMed]
  200. Fiorenza, M.; Checa, A.; Sandsdal, R.M.; Jensen, S.B.; Juhl, C.R.; Noer, M.H.; Bogh, N.P.; Lundgren, J.R.; Janus, C.; Stallknecht, B.M. Weight-loss maintenance is accompanied by interconnected alterations in circulating FGF21-adiponectin-leptin and bioactive sphingolipids. Cell Rep. Med. 2024, 5, 101629. [Google Scholar] [CrossRef] [PubMed]
  201. Rashad, N.; Gameil, D.; Sakr, M.M.H.; Ahmed, S.; Mohammad, L.G.; Elsayed, A.M.; Shaker, G.E. Altered retinol binding protein-4 (RBP-4) mRNA in obesity is associated with the susceptibility and progression of Nonalcoholic fatty liver disease (NAFLD). Zagazig Univ. Med. J. 2024, 30, 5140–5151. [Google Scholar] [CrossRef]
  202. Fan, J.; Hu, J. Retinol binding protein 4 and type 2 diabetes: From insulin resistance to pancreatic β-cell function. Endocrine 2024, 85, 1020–1034. [Google Scholar] [CrossRef]
  203. Zheng, J.; Zhang, W.; Xu, R.; Liu, L. The role of adiponectin and its receptor signaling in ocular inflammation-associated diseases. Biochem. Biophys. Res. Commun. 2024, 717, 150041. [Google Scholar] [CrossRef]
  204. Yang, H.-M.; Kim, J.; Kim, B.-K.; Seo, H.J.; Kim, J.-Y.; Lee, J.-E.; Lee, J.; You, J.; Jin, S.; Kwon, Y.-W. Resistin regulates inflammation and insulin resistance in humans via the endocannabinoid system. Research 2024, 7, 0326. [Google Scholar] [CrossRef] [PubMed]
  205. Vasileva, E.; Stankova, T.; Batalov, K.; Staynova, R.; Nonchev, B.; Bivolarska, A.; Karalilova, R. Association of serum and synovial adipokines (chemerin and resistin) with inflammatory markers and ultrasonographic evaluation scores in patients with knee joint osteoarthritis-a pilot study. Rheumatol. Int. 2024, 44, 1997–2005. [Google Scholar] [CrossRef] [PubMed]
  206. Francesca, M. Pathways Involved in Inflammaging and Frailty: The Paradigm of Visfatin in Inflammation and Metabolic Diseases and the Factors Influencing the Response to COVID-19 Vaccines. 2025. Available online: https://tesidottorato.depositolegale.it/handle/20.500.14242/188902 (accessed on 20 November 2025).
  207. Varra, F.-N.; Varras, M.; Varra, V.-K.; Theodosis-Nobelos, P. Molecular and pathophysiological relationship between obesity and chronic inflammation in the manifestation of metabolic dysfunctions and their inflammation-mediating treatment options. Mol. Med. Rep. 2024, 29, 95. [Google Scholar] [CrossRef]
  208. Taweesap, P.; Potue, P.; Khamseekaew, J.; Iampanichakul, M.; Jan, O.B.; Pakdeechote, P.; Maneesai, P. Luteolin Relieves Metabolic Dysfunction-Associated Fatty Liver Disease Caused by a High-Fat Diet in Rats Through Modulating the AdipoR1/AMPK/PPARγ Signaling Pathway. Int. J. Mol. Sci. 2025, 26, 3804. [Google Scholar] [CrossRef] [PubMed]
  209. Alsaedi, H.Z.R.; Dashti, N.; Fadaei, R.; Moradi, N.; Naeini, F.B.; Afrisham, R. WISP3/CCN6 Adipocytokine Marker in Patients with Non-alcoholic Fatty Liver Disease (NAFLD) and its Association with Some Risk Factors. Curr. Top. Med. Chem. 2025. [Google Scholar] [CrossRef]
  210. Mosialou, I.; Shikhel, S.; Liu, J.M.; Maurizi, A.; Luo, N.; He, Z.; Huang, Y.; Zong, H.; Friedman, R.A.; Barasch, J.; et al. MC4R-dependent suppression of appetite by bone-derived lipocalin 2. Nature 2017, 543, 385–390. [Google Scholar] [CrossRef]
  211. Ding, L.; Ren, J.-Y.; Huang, Y.-F.; Zhang, J.-Z.; Bai, Z.-R.; Leng, Y.; Tian, J.-W.; Wei, J.; Jin, M.-L.; Wang, G. Resistin upregulates fatty acid oxidation in synoviocytes of metabolic syndrome-associated knee osteoarthritis via CAP1/PKA/CREB to promote inflammation and catabolism. Arthritis Res. Ther. 2025, 27, 1–16. [Google Scholar] [CrossRef]
  212. Nafakhi, I.A.; Abdalsada, N.H. The association between retinol binding protein 4 (RBP4) and peripheral neuropathy in type-II diabetic patients. Anaesth. Pain Intensive Care 2024, 28, 1091–1096. [Google Scholar] [CrossRef]
  213. Cao, X.; Zhong, G.; Jin, T.; Hu, W.; Wang, J.; Shi, B.; Wei, R. Diagnostic value of retinol-binding protein 4 in diabetic nephropathy: A systematic review and meta-analysis. Front. Endocrinol. 2024, 15, 1356131. [Google Scholar] [CrossRef]
  214. Engin, A. Adiponectin resistance in obesity: Adiponectin leptin/insulin interaction. Obes. Lipotoxicity 2024, 431–462. [Google Scholar]
  215. Elgohary, M.N.; Bassiony, M.A.; Abdulsaboor, A.; Alsharqawy, A.-h.M.; Gad, S.A. Serum Resistin Level as A Biomarker in the Non-Alcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis Patients. Zagazig Univ. Med. J. 2025. [Google Scholar] [CrossRef]
  216. Zeng, L.; Li, Y.; Hong, C.; Wang, J.; Zhu, H.; Li, Q.; Cui, H.; Ma, P.; Li, R.; He, J. Association between fatty liver index and controlled attenuation parameters as markers of metabolic dysfunction-associated fatty liver disease and bone mineral density: Observational and two-sample Mendelian randomization studies. Osteoporos. Int. 2024, 35, 679–689. [Google Scholar] [CrossRef] [PubMed]
  217. Vachliotis, I.D.; Anastasilakis, A.D.; Rafailidis, V.; Polyzos, S.A. Osteokines in nonalcoholic fatty liver disease. Curr. Obes. Rep. 2024, 13, 703–723. [Google Scholar] [CrossRef]
  218. Shimonty, A.; Bonewald, L.F.; Huot, J.R. Metabolic health and disease: A role of osteokines? Calcif. Tissue Int. 2023, 113, 21–38. [Google Scholar] [CrossRef]
  219. Chondrogianni, M.E.; Kyrou, I.; Androutsakos, T.; Flessa, C.-M.; Menenakos, E.; Chatha, K.K.; Aranan, Y.; Papavassiliou, A.G.; Kassi, E.; Randeva, H.S. Anti-osteoporotic treatments in the era of non-alcoholic fatty liver disease: Friend or foe. Front. Endocrinol. 2024, 15, 1344376. [Google Scholar] [CrossRef]
  220. Monti, F.; Perazza, F.; Leoni, L.; Stefanini, B.; Ferri, S.; Tovoli, F.; Zavatta, G.; Piscaglia, F.; Petroni, M.L.; Ravaioli, F. RANK–RANKL–OPG axis in MASLD: Current evidence linking bone and liver diseases and future perspectives. Int. J. Mol. Sci. 2024, 25, 9193. [Google Scholar] [CrossRef]
  221. Zhang, M.; Tu, W.; Zhang, Q.; Wu, X.; Zou, X.; Jiang, S. Osteocalcin reduces fat accumulation and inflammatory reaction by inhibiting ROS-JNK signal pathway in chicken embryonic hepatocytes. Poult. Sci. 2022, 101, 102026. [Google Scholar] [CrossRef]
  222. Zhang, M.; Dong, K.; Du, Q.; Xu, J.; Bai, X.; Chen, L.; Yang, J. Chemically synthesized osteocalcin alleviates NAFLD via the AMPK-FOXO1/BCL6-CD36 pathway. J. Transl. Med. 2024, 22, 782. [Google Scholar] [CrossRef]
  223. Wu, X.; Zou, X.; Zhang, M.; Hu, H.; Wei, X.; Jin, M.; Cheng, H.-W.; Jiang, S. Osteocalcin prevents insulin resistance, hepatic inflammation, and activates autophagy associated with high-fat diet–induced fatty liver hemorrhagic syndrome in aged laying hens. Poult. Sci. 2021, 100, 73–83. [Google Scholar]
  224. Mohammad Rahimi, G.R.; Niyazi, A.; Alaee, S. The effect of exercise training on osteocalcin, adipocytokines, and insulin resistance: A systematic review and meta-analysis of randomized controlled trials. Osteoporos. Int. 2021, 32, 213–224. [Google Scholar] [CrossRef]
  225. Wang, Y.J.; Jin, C.H.; Ke, J.F.; Wang, J.W.; Ma, Y.L.; Lu, J.X.; Li, M.F.; Li, L.X. Decreased Serum Osteocalcin is an Independent Risk Factor for Metabolic Dysfunction-Associated Fatty Liver Disease in Type 2 Diabetes. Diabetes Metab. Syndr. Obes. Targets Ther. 2022, 15, 3717–3728. [Google Scholar] [CrossRef]
  226. Chen, H.; Chen, S.; Liu, D.; Liang, Y.; Li, H.; Bao, Y.; Zhu, Z.; Dong, K.; Li, W.; Feng, L.; et al. Associations between multiple metabolic biomarkers with steatotic liver disease subcategories: A 5-year Chinese cohort study. Cell Rep. Med. 2025, 6, 101884. [Google Scholar] [CrossRef]
  227. Coombes, J.D.; Manka, P.P.; Swiderska-Syn, M.; Vannan, D.T.; Riva, A.; Claridge, L.C.; Moylan, C.; Suzuki, A.; Briones-Orta, M.A.; Younis, R.; et al. Osteopontin Promotes Cholangiocyte Secretion of Chemokines to Support Macrophage Recruitment and Fibrosis in MASH. Liver Int. Off. J. Int. Assoc. Study Liver 2025, 45, e16131. [Google Scholar] [CrossRef]
  228. De Muynck, K.; Heyerick, L.; De Ponti, F.F.; Vanderborght, B.; Meese, T.; Van Campenhout, S.; Baudonck, L.; Gijbels, E.; Rodrigues, P.M.; Banales, J.M.; et al. Osteopontin characterizes bile duct-associated macrophages and correlates with liver fibrosis severity in primary sclerosing cholangitis. Hepatology 2024, 79, 269–288. [Google Scholar] [CrossRef]
  229. Wang, S.; Yin, J.; Liu, Z.; Liu, X.; Tian, G.; Xin, X.; Qin, Y.; Feng, X. Metabolic disorders, inter-organ crosstalk, and inflammation in the progression of metabolic dysfunction-associated steatotic liver disease. Life Sci. 2024, 359, 123211. [Google Scholar] [CrossRef]
  230. Yang, W.; Jin, Q.; Xiao, D.; Li, X.; Huang, D. Interaction mechanism and intervention strategy between metabolic dysfunction-associated steatotic liver disease and intestinal microbiota. Front. Microbiol. 2025, 16, 1597995. [Google Scholar] [CrossRef]
  231. Lopes, M.R.; Direito, R.; Guiguer, E.L.; Catharin, V.C.S.; Zutin, T.L.M.; Rubira, C.J.; Catharin, V.; Sloan, K.P.; Sloan, L.A.; Junior, J.L.Y.; et al. Bridging the Gut Microbiota and the Brain, Kidney, and Cardiovascular Health: The Role of Probiotics. Probiotics Antimicrob Proteins 2025. [Google Scholar] [CrossRef]
  232. Borozan, S.; Fernandez, C.J.; Samee, A.; Pappachan, J.M. Gut-Adipose Tissue Axis and Metabolic Health. Curr. Issues Mol. Biol. 2025, 47, 424. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The relationship between the liver and the heart (Created in https://BioRender.com, last accessed on 20 November 2025). The heart interacts with the liver during MASLD progression through various pathways, most of which are associated with cardiokine production. Cardiokines can be harmful for MASLD progression, leading to increased liver fibrosis associated with insulin resistance, hepatic inflammation, and hepatic lipid accumulation. On the other hand, MASLD exacerbates cardiac fibrosis, leading to the infiltration of inflammatory cells, which in turn is associated with increased oxidative stress and the development of cirrhotic cardiomyopathy.
Figure 1. The relationship between the liver and the heart (Created in https://BioRender.com, last accessed on 20 November 2025). The heart interacts with the liver during MASLD progression through various pathways, most of which are associated with cardiokine production. Cardiokines can be harmful for MASLD progression, leading to increased liver fibrosis associated with insulin resistance, hepatic inflammation, and hepatic lipid accumulation. On the other hand, MASLD exacerbates cardiac fibrosis, leading to the infiltration of inflammatory cells, which in turn is associated with increased oxidative stress and the development of cirrhotic cardiomyopathy.
Ijms 26 11547 g001
Figure 2. The relationship between the liver and the kidneys (Created in https://BioRender.com, last accessed on 20 November 2025). The liver and the kidneys interact with each other during health and disease. During MASLD, the kidneys’ health is worsened due to elevated renal tubular stress and an increase in the splanchnic vessels’ dilation, causing renal hypoxia and hypoperfusion. On the other hand, during MASLD, the kidneys produce renokines, such as NGAL and renin, which are associated with increased renal vasoconstriction and portal hypertension, as well as worsened steatohepatitis due to increased liver inflammation, oxidative stress, and lipid accumulation.
Figure 2. The relationship between the liver and the kidneys (Created in https://BioRender.com, last accessed on 20 November 2025). The liver and the kidneys interact with each other during health and disease. During MASLD, the kidneys’ health is worsened due to elevated renal tubular stress and an increase in the splanchnic vessels’ dilation, causing renal hypoxia and hypoperfusion. On the other hand, during MASLD, the kidneys produce renokines, such as NGAL and renin, which are associated with increased renal vasoconstriction and portal hypertension, as well as worsened steatohepatitis due to increased liver inflammation, oxidative stress, and lipid accumulation.
Ijms 26 11547 g002
Figure 3. The relationship between the liver and the adipose tissue (Created in https://BioRender.com, last accessed on 20 November 2025). The adipose tissue plays a crucial role in maintaining the body’s homeostasis. When the liver is diseased, e.g., with MASLD, the adipose tissue primarily interacts with the organ by producing leptin, resistin, and visfatin, adipokines that worsen the prognosis of the liver disease. Leptin, resistin, and visfatin induce liver fibrosis and oxidative stress, accompanied by worsened steatohepatitis. On the other hand, MASLD worsens adipose tissue-related insulin resistance, inflammation, and oxidative stress.
Figure 3. The relationship between the liver and the adipose tissue (Created in https://BioRender.com, last accessed on 20 November 2025). The adipose tissue plays a crucial role in maintaining the body’s homeostasis. When the liver is diseased, e.g., with MASLD, the adipose tissue primarily interacts with the organ by producing leptin, resistin, and visfatin, adipokines that worsen the prognosis of the liver disease. Leptin, resistin, and visfatin induce liver fibrosis and oxidative stress, accompanied by worsened steatohepatitis. On the other hand, MASLD worsens adipose tissue-related insulin resistance, inflammation, and oxidative stress.
Ijms 26 11547 g003
Figure 4. The relationship between the liver and the bones (Created in https://BioRender.com, last accessed on 20 November 2025). Bones also play a critical role in maintaining the body’s homeostasis. They are related to the production of several molecules, called osteokines, which are involved in various actions during the occurrence and progression of MASLD. Some aggressive osteokines, including FGF-23, osteopontin, and sclerostin, play a critical role in modulating liver fibrosis, inflammation, and oxidative stress during MASLD, thereby exacerbating portal hypertension as an outcome. On the other hand, MASLD induces insulin resistance, inflammation, oxidative stress, vascular calcification, and endothelial dysfunction through its interaction with osteokine production.
Figure 4. The relationship between the liver and the bones (Created in https://BioRender.com, last accessed on 20 November 2025). Bones also play a critical role in maintaining the body’s homeostasis. They are related to the production of several molecules, called osteokines, which are involved in various actions during the occurrence and progression of MASLD. Some aggressive osteokines, including FGF-23, osteopontin, and sclerostin, play a critical role in modulating liver fibrosis, inflammation, and oxidative stress during MASLD, thereby exacerbating portal hypertension as an outcome. On the other hand, MASLD induces insulin resistance, inflammation, oxidative stress, vascular calcification, and endothelial dysfunction through its interaction with osteokine production.
Ijms 26 11547 g004
Table 1. Tissue-Derived Organokines Role in Metabolic Conditions Related to MASLD.
Table 1. Tissue-Derived Organokines Role in Metabolic Conditions Related to MASLD.
Tissue-Derived CytokineCondition/Stimulus for ReleaseRelations with MASLDReferences
Hepatokines
ANGPTL3Liver lipid overloadImpairs lipid clearance; associated with dyslipidemia and liver fat[48]
Activin E↑ in obesity and MASLDRegulatory molecule that prevents fatty acid influx into the liver[49]
Fetuin-ALiver stress and insulin resistancePromotes insulin resistance and hepatic lipid accumulation[50]
FGF-21 Fasting and metabolic stressEnhances fatty acid oxidation; protective role in NAFLD[51]
Selenoprotein POxidative stress and liver dysfunctionInduces insulin resistance and hepatic inflammation[52]
Myokines
BDNF↑ hepatic lipid oxidationRegulates satiety; deficiency is associated with hyperphagia and ↑ hepatic fat deposition[53,54,55]
FGF-21 (muscle-derived)Metabolic stress and fastingImproves lipid metabolism; protective in MASLD[56]
IrisinPhysical exerciseImproves insulin sensitivity; reduces hepatic steatosis and inflammation[57,58]
IL-6 (from muscle)Acute exercise and chronic inflammationDual role: acute exercise-induced IL-6 is protective; chronically elevated levels may worsen hepatic inflammation[59,60]
IL-15Elevated serum levels in obesity; ↑ in NASH, it recruits NK lymphocytes to the liver (excess aggravates inflammation)This process increases hepatic lipid accumulation and modulates macrophage infiltration in the liver [61,62]
MyostatinMuscle inactivity; metabolic disordersThis condition hinders muscle growth and is linked to insulin resistance and the accumulation of liver fat[63,64]
Cardiokines
ANPEffective hypervolemia and atrial distensionRegulates liver glycogen metabolism and glucose homeostasis[65]
BNPCardiac damage, acute myocardial infarction, and subclinical cardiac dysfunction in cirrhosisBiomarker of volume overload and ↑ in cirrhotic cardiomyopathy[66,67]
GDF-15Tissue injury and oxidative stressLinked to metabolic regulation and hepatic stress adaptation[68,69]
IL-33Cardiac stress and inflammationMay reduce liver inflammation and fibrosis in the early stages[70]
MyostatinAdipose tissue and muscle produce myostatin and worsen peripheral resistance; accumulation of uremic toxins↑ Muscle atrophy; stimulates hepatic stellate cells (fibrosis)[54,71]
Natriuretic peptidesCardiac stretch and heart failureImprove lipid metabolism; may protect against NAFLD progression[72,73]
Renokines
ErythropoietinHypoxia and anemiaModulates insulin sensitivity; may reduce liver steatosis[74,75]
KlothoKidney function regulationAnti-inflammatory and antioxidant; protective effect in MAFLD[76,77]
NGAL Renal tubular stressA marker of kidney stress, correlated with the severity of liver injury[78,79]
Renin↓ Renal perfusion; ↓ [Na+] in the distal tubuleActivation of the RAAS; worsening steatohepatitis[73]
Adipokines
AdiponectinCaloric restriction and healthy adipose tissueAnti-inflammatory and insulin-sensitizing; protective against liver steatosis and fibrosis[80,81]
LeptinIncreased fat massHyperleptinemia promotes hepatic inflammation and fibrosis; it is elevated in MAFLD[82]
ResistinInflammation and obesity↑ Insulin resistance is associated with hepatic lipid accumulation[83,84]
VisfatinVisceral obesity and inflammationActivates pro-inflammatory pathways; ↑ production of TNF-α, IL-6, IL-1β; worsens steatohepatitis (NASH); ↑ liver fibrosis; ↑ oxidative stress[85,86]
Osteokines
FGF-23Elevated levels in NAFLD↑ Insulin resistance[87,88,89]
NGALKnown as a biomarker for acute kidney injuryOrganogenesis and modulation of inflammation; elevated in metabolic diseases, including liver diseases[90]
OsteocalcinBone remodeling, mechanical loading↓ accumulation of lipids in the liver[43,91]
OsteopontinInflammation and tissue injuryPromotes hepatic inflammation and fibrosis; elevated in MASLD and NASH[92]
SclerostinMechanical unloading of boneImpairs insulin sensitivity; may contribute to metabolic dysfunction in liver disease[93,94]
↓: decrease; ↑: increase; ANGPTL3: Angiopoietin-Like 3; ANP: Atrial Natriuretic Peptide; BNP: B-Type Natriuretic Peptide; BDNF: Brain-Derived Neurotrophic Factor; FGF-21: Fibroblast Growth Factor 21; FGF-23: Fibroblast Growth Factor 23; GDF-15: Growth Differentiation Factor 15; IL: Interleukin; MAFLD: Metabolic-associated fatty liver disease; MASLD: Metabolic dysfunction-associated steatotic liver disease; NAFLD: Non-Alcoholic Fatty Liver Disease; NGAL: Neutrophil gelatinase-associated lipocalin; NASH: Nonalcoholic steatohepatitis; RAAS: Renin-Angiotensin-Aldosterone System; TNF-α: Tumor Necrosis Factor-α.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Maria Barbalho, S.; Laurindo, L.F.; Valenti, V.E.; Méndez-Sánchez, N.; Ramírez-Mejía, M.M.; Goulart, R.d.A. Organokine-Mediated Crosstalk: A Systems Biology Perspective on the Pathogenesis of MASLD—A Narrative Review. Int. J. Mol. Sci. 2025, 26, 11547. https://doi.org/10.3390/ijms262311547

AMA Style

Maria Barbalho S, Laurindo LF, Valenti VE, Méndez-Sánchez N, Ramírez-Mejía MM, Goulart RdA. Organokine-Mediated Crosstalk: A Systems Biology Perspective on the Pathogenesis of MASLD—A Narrative Review. International Journal of Molecular Sciences. 2025; 26(23):11547. https://doi.org/10.3390/ijms262311547

Chicago/Turabian Style

Maria Barbalho, Sandra, Lucas Fornari Laurindo, Vitor Engracia Valenti, Nahum Méndez-Sánchez, Mariana M. Ramírez-Mejía, and Ricardo de Alvares Goulart. 2025. "Organokine-Mediated Crosstalk: A Systems Biology Perspective on the Pathogenesis of MASLD—A Narrative Review" International Journal of Molecular Sciences 26, no. 23: 11547. https://doi.org/10.3390/ijms262311547

APA Style

Maria Barbalho, S., Laurindo, L. F., Valenti, V. E., Méndez-Sánchez, N., Ramírez-Mejía, M. M., & Goulart, R. d. A. (2025). Organokine-Mediated Crosstalk: A Systems Biology Perspective on the Pathogenesis of MASLD—A Narrative Review. International Journal of Molecular Sciences, 26(23), 11547. https://doi.org/10.3390/ijms262311547

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