Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Molecular Medicine
Join Editorial Board Propose a Special Issue
Print ISSN: 1107-3756 Online ISSN: 1791-244X
Journal Cover
December-2025 Volume 56 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
December-2025 Volume 56 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Metabolic dysfunction‑associated steatotic liver disease: Pathogenesis, model and treatment (Review)

  • Authors:
    • Qinge Ma
    • Kejia Liu
    • Chenyu Chang
    • Lei Wang
    • Zhangyang Shen
    • Jiaxin Li
    • Mozili Adu
    • Qingyuan Lin
    • Huilian Huang
    • Xutao Wu
    • Rongrui Wei
  • View Affiliations / Copyright

    Affiliations: Key Laboratory of Modern Preparation of Traditional Chinese Medicine of Ministry of Education, Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Laboratory Service Center, The Second Affiliated Hospital, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, P.R. China
    Copyright: © Ma et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 227
    |
    Published online on: October 17, 2025
       https://doi.org/10.3892/ijmm.2025.5668
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Metabolic dysfunction‑associated steatotic liver disease (MASLD) is caused by multiple factors that lead to the buildup of steatosis and fat deposition in hepatocytes. These changes are the primary hallmarks of the disease and result in significant impairment of liver function. Consequently, the quality of life of patients and their ability to work are adversely affected. The pathogenesis of MASLD involves both Western and Chinese medicines, with these mechanisms markedly influencing the onset and progression of MASLD; they are not independent but rather interrelated. Conducting histopathological diagnosis of MASLD in the liver is challenging in humans. Consequently, both in vivo and in vitro models are essential. Researchers must select appropriate methods and model types to establish MASLD models that most suitably mimic the human body. Currently, both pharmacological and non‑pharmacological treatments have some efficacy in improving the condition of MASLD and the combination of the two is more helpful in providing more effective treatment for patients, but further research and clinical trials are needed to verify in the future. Therefore, the present review comprehensively summarized the pathogenesis, model and treatment of MASLD. It will provide an important basis for subsequent research on MASLD.

Introduction

Non-alcoholic fatty liver disease (NAFLD) was proposed by Ludwig in 1986. It refers to a fatty liver disease characterized by excessive fat deposition in hepatocytes (with a fat content of ≥5%), excluding alcohol and other factors related to chronic liver diseases (1-4). In 2020, the Asian Pacific Association for the Study of the Liver proposed the term 'metabolic dysfunction-associated fatty liver disease (MAFLD)', emphasizing the role of metabolic disorders in the progression of fatty liver disease (5). In 2023, an international panel of experts reached a consensus and the European Association for the Study of the Liver, from an etiological perspective, renamed it as 'metabolic dysfunction-associated steatotic liver disease (MASLD)' (6,7). This change highlights the critical role of metabolic abnormalities in the occurrence and development of the disease (8). While avoiding 'stigmatization', this concept also provides explanations for the coexistence of MASLD and alcoholic-associated liver disease (9,10).

The etiology of MASLD is complex, with its core being the interplay between metabolic dysfunction (such as insulin resistance and dyslipidemia) and multifactorial elements (including genetics, environment and lifestyle), rather than being attributed to a single factor. Researchers have established various animal models of MASLD using diverse methodologies to investigate its pathophysiological characteristics. Currently, common interventional drugs mainly include: Farnesoid X receptor agonists, peroxisome proliferator-activated receptor (PPAR)α/γ/δ agonists, GLP-1 agonists and fibroblast growth factor 19/21 analogs (11). For individuals with a body mass index >35, bariatric surgery is primarily recommended. In the treatment of MASLD, although chemical drugs have advanced rapidly, their efficacy remains limited with notable adverse effects. By contrast, traditional Chinese medicine (TCM) not only enables comprehensive regulation targeting the multifactorial pathogenesis of MASLD, but also strives to improve the quality of life of patients, thereby opening up new possibilities for the management of MASLD (12).

In recent years, influenced by unhealthy lifestyles and dietary habits, the prevalence of MASLD has been increasing annually. The global incidence rate has reached 25-35%, with a prevalence of 29.2% in China (13). It is projected that the number of MASLD patients in China will reach 315 million by 2030, imposing a heavy economic burden on society (14). Even mild fatty liver disease increases the risk of death by 71% and this risk is positively associated with the severity of the disease (14). Currently, while the incidence of viral liver diseases is on the decline, the incidence of MASLD is rising, making early intervention for MASLD an urgent priority. The present study analyzed the pathogenesis, experimental models and therapeutic approaches of MASLD, aiming to provide novel scientific insights and strategies for future research, clinical treatment and drug development.

Methods

To compile the research progress on the pathogenesis, model and treatment of MASLD as comprehensively as possible, the present study searched for 'pathogenesis', 'model', 'treatment' and 'traditional Chinese medicine' in existing scientific databases. The references related to MASLD were obtained from both online and offline databases, spanning the period from 1980-2025 with a total of 385 references. Online databases included PubMed (https://pubmed.ncbi.nlm.nih.gov/), Web of Science (https://www.webofscience.com/), Elsevier (https://www.elsever.com/), Sci-Hub (https://sci-hub.st/), Wiley (https://www.wiley.com/), SpringerLink (https://link.springer.com/), Google Scholar (https://scholar.google.com/), EMBASE (https://www.embase.com/), Cochrane Library and China National Knowledge Infrastructure (CNKI) (https://www.cnki.net/). Other references were obtained from ancient Chinese books, pharmacopoeias and other articles. The present authors used BioGDP (https://biogdp.com/) as well as Adobe Illustrator (Adobe Systems, Inc.) for graphing.

Pathogenesis of MASLD

Research on the pathogenesis of MASLD in chemical pharmaceuticals

The pathogenesis of MASLD remains unclear due to its complexity and diversity. As the main metabolic organ, the liver's metabolism is closely connected with multiple organs and systems in the human body. There are intricate links between these organs and systems. Some consider that the hypothesis of 'multiple parallel strikes' could be an improved explanation of the causes of MASLD development and these strikes included oxidative stress, endoplasmic reticulum (ER) stress, lipid metabolism disorders, abnormal adipokines and cytokines production and mitochondrial dysfunction (15). One study found that mitochondrial dysfunction might be a central factor in the development of MASLD and other factors that cause disease progression also involve mitochondrial dysfunction (16). The pathogenesis of MASLD is now being investigated by combining different pathogenic pathways for an improved understanding of metabolic diseases.

The 'multiple strikes' hypothesis

The 'first strike' mainly refers to insulin resistance (IR) caused by obesity and type 2 diabetes mellitus (T2DM) and hepatic steatosis caused by excessive accumulation of triglyceride (TG) and cholesterol in the liver parenchymal cells in the form of lipid droplets. The 'Two-hit' hypothesis includes inflammatory cytokines, lipid peroxidation, mitochondrial dysfunction and oxidative stress (17), of which oxidative stress is the primary driver (18). This hypothesis posits that with steatosis, the liver undergoes ER stress, oxidative stress and mitochondrial dysfunction, leading to an increase in oxidative metabolites, hepatocellular damage and secretion of a large number of inflammatory factors, which ultimately leads to further deterioration into liver disease, liver fibrosis, cirrhosis, inflammatory necrosis and other reactions (19,20). In the early stages of MASLD, classical activation of macrophage M1 in the liver can promote further inflammation and IR, as well as steatosis (21). Among them, inflammatory cytokines such as interleukin (IL)-6, IL-8, tumor necrosis factor-α (TNF-α), IL-1β and cyclooxygenase-2 contribute to the development of chronic inflammatory diseases (22). IL-6 has been shown to have both anti-inflammatory and pro-inflammatory effects, but its role in MASLD is controversial. IL-10, produced primarily by monocytes and B cells, is an anti-inflammatory factor with immunomodulatory properties (22). Other inflammatory cells of the innate immune system, such as natural killer T cell and natural killer cells, also play the important roles in the pathogenesis of MASLD and MASH (21). However, the factors that induce MASLD in clinical practice are diverse and as research has deepened, the two-hit hypothesis has already shown certain limitations and cannot fully explain the pathogenesis of MASLD. Recently, the 'three-strike' and 'multiple-strike' hypotheses have been proposed, which include gut microbiota toxins, IR, cytokines and inflammation, oxidative stress due to mitochondrial dysfunction, lipid peroxidation, ER stress, intrinsic immune regulation, circadian rhythm disruption and immune system development. Imbalances, circadian patterns and genetics are all parallel and mutually reinforcing factors that contribute to the onset and development of MASLD (23,24) (Fig. 1).

The mechanism 'Multiple Strike'
hypothesis of MASLD. MASLD, metabolic dysfunction-associated
steatotic liver disease; NAFLD, non-alcoholic fatty liver disease;
NASH, nonalcoholic steatohepatitis.

Figure 1

The mechanism 'Multiple Strike' hypothesis of MASLD. MASLD, metabolic dysfunction-associated steatotic liver disease; NAFLD, non-alcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis.

LSEC with abnormal window aperture

The role of liver sinusoidal endothelial cell (LSEC) in the development and progression of MASLD is being gradually recognized. LSEC is the most predominant liver nonparenchymal cell (NPC), accounting for 15-20% of the total number of hepatocytes but only 3% of the total volume of the liver (25). The biggest difference between LSECs and other vascular endothelial cells is the presence of window pores and the lack of a basement membrane. LSECs are considered the most permeable endothelial cells in mammals (25). Window pores are small pores with diameters ranging from 50-200 nm, accounting for 2-20% of the endothelial surface area (26). MASLD manifests as NAFL in the early stage, which is not accompanied, or only accompanied by mild, inflammation. LSEC acts as the first line of defense in the initiation of MASLD (27), exerting anti-inflammatory effects, preventing the over-activation of the hepatic immune system and maintaining the microenvironmental homeostasis of hepatic sinusoids. It has been shown that the unique window pore structure of LSECs in a physiological state serves an important role in maintaining the stability of the liver, which manifests pro-vasodilatory, strong endocytosis, anti-inflammatory and anti-fibrotic effects (28). This window pore regulates the act in the endothelial cell skeleton of LSECs through the form of calcium-calmodulin-actin complex to adjust and control the size of the window pore on the endothelial cells of LSEC; if the size, number, and permeability of window pores in LSECs could not be effectively improved, they would disappear in the course of time. From the initial manifestation of fat accumulation to inflammatory symptoms, as well as hepatic fibrosis and in severe cases, cirrhosis, LSEC regulates blood flow through the secretion of two substances, nitric oxide (NO) and endothelin-1 (ET-1) (27). NO promotes vasodilatation and ET-1 induces vasoconstriction. Reduced NO bioavailability induces LSEC capillarization characterized by loss of the LSEC window pore and basement membrane formation and leads to an imbalance in its secretion of vasoactive substances, which contributes to MASLD progression. With the progression of MASLD, LSEC undergoes capillarization and hepatic sinusoidal endothelial dysfunction, which gradually shifts from an anti-inflammatory to a pro-inflammatory phenotype and promotes the progression of inflammation, fibrosis and angiogenesis. Hepatic sinusoidal endothelial dysfunction precedes the development of hepatic inflammation or fibrosis. It is a major feature and early event in MASLD pathogenesis (29).

Insulin resistance

IR refers to the phenomenon of reduced sensitivity of tissues and organs to insulin, which is the initiation and central link in the progression of MASLD (30). It has been found that the degree of IR in MASLD patients is positively associated with the severity of the disease (31). IR acts on target organs mainly the liver, skeletal muscle and adipose tissue; the body's sensitivity to insulin is reduced, so that the biological effect of insulin on target organs is reduced and glucose uptake and utilization are reduced, thus failing to maintain blood glucose stability. Compensatory overproduction of pancreatic islets and eventual development of hyperinsulinemia, result in elevated hepatic synthesis and accumulation of total cholesterol (TC) and elevated low-density lipoprotein (LDL) levels, leading to inactivation of the enzyme LDL lipase, which is critical for cholesterol clearance. Large amounts of TG cannot be cleared and IR-induced hyperinsulinemia inhibits apolipoprotein B100 synthesis and reduces very low-density lipoprotein (VLDL) related lipid output from hepatocytes (32). In addition, IR leads to decreasing lipid metabolism, increasing lipolysis and hepatic uptake of large amounts of free fatty acid (FFA). By contrast, fatty acid β-oxidation is inhibited by hyperinsulinemia and large amounts of FFA are deposited in the liver, exacerbating hepatocellular steatosis (33). Excessive deposition of lipids further exacerbates IR and they are mutually influencing and reinforcing processes (34). MASLD also leads to lower levels of adiponectin (ADPN), a protein released from adipocytes. ADPN aggravates steatosis in hepatocytes (35) and plays a role in regulating glucose and lipid metabolism, as well as exhibiting anti-inflammatory and anti-insulin resistance effects (36). In addition, Shah and Fonseca (37) found that iron overload could reduce insulin sensitivity and IR, which are closely related to the occurrence of MASLD. The main reason is that iron can activate the nuclear factor-kappa B (NF-κB) signaling pathway. Activation of this pathway causes hepatocytes to produce an inflammatory response, which accelerates the formation of MASLD. Although iron is an essential trace element in the human body, a study found that iron overload is closely related to the occurrence of MASLD (37). Iron overload can generate reactive oxygen species (ROS) and cause oxidative stress. Iron overload can also affect lipid metabolism and insulin signaling, which can accelerate the progression of MASLD (38). Using hepatic puncture biopsy, Nelson et al (39) discovered that 34.5% of 849 patients with MASLD had intrahepatic iron deposition, which suggested that iron deposition was a risk factor for the progression of MASLD patients to advanced liver diseases.

Adipokines

Adipokines are small-molecule proteins secreted by tissues to regulate adipocytes, mainly including leptin (LP) and ADPN. A study (40) found that IR is closely related to the secretion of LP, resistin and ADPN. ADPN is an adipokine that affects hepatic fat and glucose metabolism, which interacts with adiponectin receptor1 and adiponectin receptor 2. Adiponectin receptor 2 is mainly expressed in liver tissues. Clinical studies have shown that the serum level of ADPN in patients with MASLD is low and negatively is associated with the severity of fatty liver (40). In addition, serum lipocalin levels are reduced in MASLD patients and are associated with the degree of hepatic necroinflammation, leading to the suggestion that hypo-lipocalinemia will lead to the progression of MASLD, which may be related to the fact that ADPN acts on the sterol regulatory element-binding protein-1C to inhibit lipolysis of fats (41). Studies have shown a close relationship between LP and the development of MASLD and that LP can lead to the activation of hepatic LP and can lead to the activation of hepatic stellate cells in MASLD and promote the development of MASLD to liver fibrosis (41). Moreover, LP is also associated with IR; when the feedback regulation mechanism between pancreatic islets-adipocytes is damaged, the sensitivity of insulin to LP decreases. It makes the fatty acid content of hepatocytes rise, which promotes the increase in the synthesis of TGs in the hepatocytes, leading to the formation of MASLD (42). Wang et al (43) found that the amount of ADPN expression can be used to predict the occurrence of MASLD to a certain extent. Resistin is a prepeptide adipocytokine containing a 108-amino-acid segment, named after its anti-insulin effect. In another study, RES was found to be highly expressed in adipose tissue and serum of both high-fat diet-induced obese rats and transgenic obese rats (44). Activation of resistin can induce the release of inflammatory factors and promote the development of liver fibrosis (45).

Lipotoxicity

Lipotoxicity refers to lipid metabolism disorders leading to an increase in FFA, excessive FFA makes pancreatic islet β-cells dysfunctional, inducing them to produce a large amount of NO, causing a series of cytotoxic injuries. Markedly increased plasma FFA level in MASLD patients is the main reason for increased IR in the body. In addition, increasing plasma FFA content leads to fatty acid oxidation overload in hepatocytes, induces mitochondrial damage and generates large amounts of ROS, causing oxidative stress, ER stress and inflammatory response, which advances the process of disease progression. The mechanisms of hepatic fatty acid metabolism on MASLD are shown in Fig. 2.

The mechanism of hepatic fatty acid
metabolism on MASLD. FAO, fatty acid beta-oxidation; FATP, fatty
acid transport protein; MASLD, metabolic dysfunction-associated
steatotic liver disease; VLDL, very low-density lipoprotein.

Figure 2

The mechanism of hepatic fatty acid metabolism on MASLD. FAO, fatty acid beta-oxidation; FATP, fatty acid transport protein; MASLD, metabolic dysfunction-associated steatotic liver disease; VLDL, very low-density lipoprotein.

Lipotoxicity promotes hepatocyte apoptosis

The normal number of hepatocytes is controlled by hepatocyte apoptosis, which maintains the liver at a normal size and plays an important role in the development of the liver and the maintenance of internal homeostasis, it is the defender of hepatocytes against infections, tumors and autoimmune reactions; under pathological conditions apoptosis of hepatocytes is the central link in the basis of injury and other liver diseases. Lipotoxicity induces apoptosis in hepatocytes, called lipoapoptosis. It has been demonstrated that FFA-treated hepatocytes show an increase in the expression of pro-apoptotic proteins and apoptosis regulators upregulated by tumor suppressor genes, a process that is accompanied by a decrease in the expression of the anti-apoptotic protein B-cell lymphoma-2. The aforementioned process initiates the mitochondria-induced apoptosis pathway, which activates caspase-3,6,7 leading to apoptosis (46). Hepatocyte apoptosis is closely related to the pathogenesis of MASLD (47).

Lipotoxicity-induced liver mitochondrial damage

Mitochondria are an important site for hepatic FFA oxidation. When FFA and related metabolites in the liver increase in excess, it causes mitochondrial microstructure swelling and mitochondrial dysfunction, resulting in impaired β-oxidation, altering the permeability of the ER membrane, inducing the production of a large number of ROS. ATP generation is reduced, causing oxidative stress and mitochondrial damage (48). In the meantime, ROS generated by oxidative stress in the mitochondrial membrane react with unsaturated fatty acids of mitochondrial membrane phospholipids, nucleic acids and other macromolecules to cause lipid peroxidation and the products of lipid peroxidation produce endogenous ROS and O2−, which can damage mitochondrial DNA structure and further diminish the anti-oxidant effect of mitochondria (49). In addition, ROS generated by oxidative stress in hepatocytes can trigger an inflammatory response. This response induces neutrophil infiltration and activates Kupffer cells (KCs) to secrete ROS and TNF-α. These events create a vicious cycle of oxidative stress, leading to VLDL deposition in the liver and the development of MASLD. Further hepatocyte necrosis may occur due to other cytokines, such as transforming growth factor-β (TGF-β), IL-6 and C-reactive protein (49,50).

Lipotoxicity-induced inflammation promotes MASLD

Persistent inflammation is the main driver of MASLD progression to MASH and fibrosis and activation of toll-like receptors (TLRs) is thought to be a key factor in triggering hepatic steatotic inflammation. The liver, as a central immune organ, possesses the largest number of resident macrophages, known as KCs. KCs account for 35% of the liver's NPC and 80-90% of all tissue macrophages in the body. KCs are considered to be the first immune cells to come into contact with intestinal or hepatic autoimmune reactive substances and are rich in the expression of TLRs (51). Accumulation of lipids in hepatocytes will result in lipotoxicity, which will lead to hepatocyte damage or death. Damaged or dead hepatocytes release damage-associated pattern molecules, including mitochondrial DNA (mtDNA) and high mobility group protein-1 (HMGB1) (52). Among them, mtDNA can directly activate recombinant TLR9 in KCs, triggering an inflammatory cascade (53). Recombinant toll-like receptor 4 (TLR4) (Fig. 3) is another TLR family member upregulated in MASLD and lipopolysaccharide (LPS), ROS, HMGB1 and various damage-associated molecular patterns can bind to TLR4 in KCs to promote TNF-α, IL-1β, IL-6 and interferon-γ (IFN-γ) production of various pro-inflammatory cytokines, thereby contributing to hepatic inflammation and the progression of MASLD (54). In addition to triggering the inflammatory response through lipotoxicity that causes hepatocyte injury or death, excess FFA in the liver can also exacerbate the inflammatory response through direct or indirect activation of TLR4. Circulating FFA, especially saturated fatty acid (SFA), acts as non-microbial TLR4 agonists and triggers inflammatory responses. SFA is thought to have a similar recognition pathway to LPS, a natural ligand of TLR4, in order to activate the TLR4 signaling pathway, thereby triggering downstream inflammatory pathways (55). Furthermore, to activate TLR4, SFA is also thought to activate recombinant TLR2 to promote the development of inflammatory responses (56). It has been shown that SFA can activate TLR by interacting with TLR co-receptors, such as cluster of differentiation36 (CD36) or LDL receptors, to promote inflammation (57). In addition, FFA can interact with various other cytokines, such as hepatocyte nuclear factor 4-α (HNF-4α), leading to overall changes in signaling pathways that regulate metabolism and stress (58). Lipotoxicity can also further induce ER stress, impair autophagy and promote aseptic inflammatory responses, thereby exacerbating hepatocyte injury and death (59,60). Cholesterol synthesis is markedly increased in patients with MASLD, suggesting that cholesterol may also be one of the important driving forces in its development (61). Cholesterol not only promotes de novo lipid synthesis but also induces lipid peroxidation (62). Moreover, it has been found that increased dietary cholesterol intake leads to hepatic inflammation and oxidative stress in mice, suggesting that cholesterol probably plays a contributory role in the disease progression of MASLD (63).

The mechanism of lipotoxicity-induced
inflammation promotes MASLD. LPS, lipopolysaccharide; MASLD,
metabolic dysfunction-associated steatotic liver disease; MD2,
myeloid differentiation factor 2; TLR4, Toll-like receptor 4.

Figure 3

The mechanism of lipotoxicity-induced inflammation promotes MASLD. LPS, lipopolysaccharide; MASLD, metabolic dysfunction-associated steatotic liver disease; MD2, myeloid differentiation factor 2; TLR4, Toll-like receptor 4.

ER stress contributes to the development of MASLD

Lipid accumulation in hepatocytes can affect mitochondrial function, ER membrane fluidity and calcium ion homeostasis. It has been found that thioesterase superfamily member 2 (THEM2) promotes the uptake of saturated fatty acids by ER phospholipid membranes, reduces ER membrane fluidity and affects the activity of ER Ca2+associated ATPase. The dysregulation of Ca2+ signaling can cause protein misfolding and secretion (64). Non-esterified fatty acids can expand the membrane structure of the endoplasmic reticulum-mitochondria association. Changes in the membrane structure of the ER allow Ca2+ to flow into the cytoplasm or mitochondria, which in turn alters the open channels of the mitochondria and cytochrome c is released into the cytoplasm. Ca2+ entering the cytoplasm can activate the Ca2+ dependent protein kinase, which together with cytochrome c induces the onset of apoptosis (65). ROS generated by mitochondria can enter the ER and trigger the unfolded protein response (UPR), which can activate three types of ER transmembrane protein receptors: Protein kinase RNA-like endoplasmic reticulum kinase (PERK), activating transcription factor-6α (ATF-6α) and inosital-requiring enzyme-1α (IRE1α), which mediate three signaling pathways. The three transmembrane proteins mediate three signaling pathways, which can maintain the stability of ER protein synthesis to a certain extent in the early stage of steatosis, but under the continuous stimulation of lipids and ROS, the three transmembrane proteins mediate the signaling pathways that can activate cellular inflammation and apoptosis, accelerating the progression of hepatic steatosis (66).

In the process of hepatocellular steatosis, one of the hallmarks of MASLD is the appearance of the UPR in hepatocytes, which can inhibit mitochondrial lipid oxidation through the UPR-induced ATF-6α pathway and promote fatty acid synthesis via the PERK, ATF-6α and IRE1α signaling pathways, which together promote intracellular lipid accumulation (67,68). Hepatocyte lipid accumulation and ER stress form a positive feedback loop that continuously exacerbates hepatocyte steatosis. The occurrence of chronic inflammation in hepatocytes is also one of the main features of metabolic disorders. The inflammatory response can activate the UPR through three major signaling pathways (PERK, ATF-6α and IRE1α). The UPR can also interact with NF-κB and N-terminal kinase/activator protein 1-regulated pro-inflammatory pathways to accelerate disease progression (66,69).

Imbalance of the intestinal flora
Effects of intestinal permeability

The intestinal flora is a symbiotic group of microorganisms that act synergistically with the host. Under normal circumstances, the intestinal flora of the human body is in a state of equilibrium. Imbalances in the intestinal flora can be caused by environmental factors, immune levels, bile secretion, changes in gastric acidity and alkalinity and impaired intestinal peristalsis. In recent years, it has been found that dysbiosis is one of the causative factors of MASLD (70). The intestinal flora mainly influences the liver through the 'gut-liver axis'. Normally, there is a balance between the barrier function of the intestine and the detoxification capacity of the liver and trace amounts of intestinal bacteria cross the mucosa and enter the venous bloodstream and are cleared by the liver (71). When intestinal flora dysbiosis occurs, metabolic disorders will occur, the permeability of the intestinal tract will increase and a large amount of intestinal bacterial metabolites, bacterial components and other harmful substances will enter the liver through the portal vein via the intestinal-hepatic axis and through the enterohepatic circulation reach the liver and bind to the TLR4 of hepatocytes and exacerbate the hepatic inflammatory response, oxidative stress and lipid accumulation, which further stimulate the inflammatory response. Intestinal microorganisms change their phenotype and virulence by sensing the various adverse signals generated during intestinal stress, shifting from a commensal to a pathogenic mode and causing damage to the organism (72). Ley et al (73) found that changes in the composition of the gut microbial community associated with obesity as well as IR were observed after the adoption of two different dietary patterns. IR, an important feature of MASLD, can be improved after antibiotic treatment (74). A study conducted by Le et al (75) demonstrated that when gut microbes from successfully modeled MASLD mice were transplanted into normal control mice, the normal control mice developed MASLD and this study strongly confirmed that the development of MASLD is associated with the development of MASLD and its effects on the gut microbial community. This study also confirms the correlation between the development of MASLD and gut microbes. A Chinese cohort study reported that high-alcohol-producing Klebsiella Pneumoniae (HiAlc-Kpn), which produces large amounts of alcohol, was detected in 61% of MASLD patients. In order to investigate the relationship between HiAlc bacteria and fatty liver disease, they fed specific pathogen free (SPF) mice with HiAlc-Kpn. It is noteworthy that HiAlc-Kpn feeding induced chronic hepatic steatosis. They investigated the relationship between intestinal flora imbalance and severe MASLD lesions (MASH and fibrosis) and showed that an increase in the number of Ruminococcus was positively associated with an aggravation of the degree of fibrosis (76). In addition, LPS is an important component of the cell wall of Gram-negative bacilli. It is recognized by pattern receptors and mediates the LPS-CD14-TLR4 signaling pathway, which activates intrahepatic KCs. These KCs release large amounts of cytokines, such as IL-6, CD68 and TNF-α. LPS also causes inflammation and metabolic disorders in vivo, including increased fat burning, elevated circulating free FFA and TG and deposition of FFA in the liver. This process may contribute to inflammation and trigger IR, which can ultimately lead to MASLD. The deposition of FFA in the liver may also promote inflammation, leading to IR, which in turn triggers the development of MASLD (77). Dysbiosis may also contribute to MASH through other mechanisms, such as ethanol production and interference with choline metabolism (22). Overall, intestinal flora and its harmful metabolites (including EtOH, SFA, polyamines and H2S) may be drivers of liver injury (76).

Effects of Bile acid (BA) metabolism

BA is synthesized in the liver and the main raw material for synthesis is cholesterol. BA regulates glucose and lipid metabolism. BA, along with its downstream receptors farnesoid X receptor (FXR) and Takeda G protein-coupled receptor 5 (TGR5), plays an important role in lipid metabolism in the liver (78). BA also regulates the cholesterol metabolic pathway, allowing cholesterol to be eliminated as a water-soluble product and impaired regulation can lead to an inflammatory response that can exacerbate the development of MASLD (79). In a MASLD mouse model, Jiang et al (80) found that the administration of antibiotics to mice on a high-fat diet reduced triacylglycerol accumulation in the liver. Activation of TGR5 by BA in brown adipose tissue and muscle increases energy expenditure and reduces diet-induced obesity (81). Activated TGR5 can downregulate the expression level of inflammatory factors, promote energy expenditure in adipose and muscle tissues and regulate the body's immunity, which has a positive effect on improving MASLD (82). Watanabe et al (83) found in an animal experiment that cholesterol and TG levels were markedly elevated in mice lacking FXR in vivo. FXR has a positive effect on lipid regulation and also reduces the generation of inflammatory responses; FXR plays a key role in the regulation of BA and regulates lipid metabolism. In addition, BA can affect the growth of bacteria and is a bacteriostatic substance. Conversely, intestinal flora can also regulate BA, intestinal flora and BA metabolism affect each other. However, the effects of different flora are inconsistent. For example, Aspergillus and Anaplastic bacilli in the intestine will reduce the synthesis of BA and aggravate inflammatory reactions, while Actinobacteria will increase the synthesis of BA, reduce the inflammatory response and reduce the damage of hepatocytes (84). The normal metabolism of BA plays a crucial role in maintaining the balance of intestinal flora. Gut microorganisms can also accelerate the metabolism of primary BA and produce secondary BA, increasing BA types (85). LPS produced by intestinal flora can stimulate NF-κB to recruit inflammatory factors and increase the level of inflammation in the body. These studies suggest that influencing the gut flora by modulating the signaling pathways between BA and their controlled receptors is a promising new therapeutic approach for the treatment of MASLD (86).

Cellular autophagy

Phenotypic changes in LSEC are one of the key events in the progression of MASLD in humans, suggesting that the progression of MASLD inflammation, hepatic fibrosis and impaired hepatic lipid metabolism may affect the expression of Fc γ receptor IIb (FcγR IIb) and macrophage function in LSEC (87). Autophagy is an intracellular process that maintains homeostasis in vivo by forming double-membrane autophagosomes that encapsulate to-be-degraded material and bind to lysosomes to self-digest excessive or defective organelles. Autophagy can be categorized into three types: Macroautophagy, microautophagy and molecular chaperone-mediated autophagy, of which the most common is macroautophagy. Autophagy is related to MASLD and macroautophagy is the main type of autophagy that regulates MASLD. Liver autophagy is defective and autophagy level is reduced in MASLD patients and autophagy shows different functions at different stages of MASLD (87). In the early stage of MASLD, autophagy inhibits apoptosis (87), while in the late stage, autophagy is pro-apoptotic, which also indicates that autophagy is involved in the whole stage of MASLD development (87). Recombinant autophagy-related protein 7 (Atg7), a core member of the anti-thymocyte globulin (ATG) gene family, is responsible for driving the classical degradation and the major stages of autophagy, which is a key component of the autophagy-associated gene family. Abnormal expression of Atg7, a core member of the ATG gene family, can drive the main stage of classical degradation of autophagy, resulting in defective autophagy in cells. In Atg7 knockout mice, hepatic autophagy activity was markedly reduced and intracellular lipid accumulation was increased, leading to the formation of MASLD (88). Phosphatidylinositol-3 kinase (PI3K) is a key regulator of the activation of the mammalian target of rapamycin (mTOR), which is considered an important regulator upstream of the ATG gene. The PI3K inhibitor 3-methyladenine blocked the formation of autophagosomes, increased the lipid content of hepatocytes and promoted the formation of MASLD (89).

Inflammatory response induced by immune system dysfunction

Inflammatory response induced by immune system dysfunction has been found to be one of the hallmarks of MASH and a progressive form of MASLD (90). Under normal conditions, the liver has immune defense and immunoregulatory functions and when the intensity of the immune system's response to autoantigens exceeds the limits of immune regulation and affects its physiological functions, it will cause inflammatory damage to its tissues or organs, leading to the occurrence of autoimmune diseases. The liver has a large number of immune cells involved in the immune response and it has been found that macrophages are the largest proportion of immune cells (80-90%) in the human liver and they are closely related to the development and severity of MASLD (91). In patients with MASLD, macrophage infiltration occurs around the portal vein and is observed at an early stage before evidence of inflammation appears. In addition, macrophages are capable of producing a variety of inflammatory factors, such as NF-κB, TNF-α, IL-2, IL-6 and IL-8 and activating nuclear transcription factors like NF-κB, which also play an important role in the progression of MASLD (92). NF-κB, as a nuclear transcriptional regulator of inflammatory genes, which is activated, enters the nucleus and promotes the transcription and expression of inflammatory factors, releasing a large number of inflammatory factors. After entering the liver, NF-κB leads to oxidative stress in hepatocytes, activates the expression of pro-apoptotic proteins in mitochondria, promotes hepatocyte apoptosis and exacerbates the progression of MASLD (93). TNF-α is one of the important regulators mediating hepatocyte injury and is also a bridge between inflammation and metabolism that plays a key role in the development of MASLD. TNF-α regulates the inflammatory signaling pathway and exacerbates the inflammatory response in MASLD through pathways such as NF-κB and p38 mitogen-activated kinase (94). IL-2 can promote the proliferation of activated B cells and is also a growth factor for all T cell subpopulations and can positively feedback regulate the production of cytokines such as TNF-α, leading to a further increase in IL-2 in MASLD livers (95). IL-6 can inhibit the activity of lipoprotein esterase, which can reduce the ability to catabolize lipids and promote the formation of hepatic lipids. IL-8 can cause inflammatory cells to accumulate in the liver and cause inflammation and lipid accumulation in hepatocytes can also stimulate the inflammation of MASLD hepatocytes. The accumulation of lipids in hepatocytes can also stimulate KCs to release TNF-α, which further promotes the production of IL-8 and participates in the inflammatory response of hepatocytes, leading to the aggravation of hepatocyte injury (96).

Law of genetics

Genetic biomarkers for MASLD include DNA sequence variants, such as single nucleotide polymorphism (SNP) and the most studied SNPs in MASLD are rs738409 and rs58542926, which are located in patatin-like phospholipase domain containing protein 3 (PNPLA3) and transmembrane 6 superfamily member 2 (TM6SF2), respectively. The risk of developing MASLD has been found to be influenced by single nucleotide gene polymorphisms. For example, SNPs in the PNPLA3 gene and mutations in the PNPLA3 gene are associated with MASLD. PAPLA3 is one of the members of the patatin-like phospholipase family that have been identified as being closely associated with the development of MASLD (97,98), PNPLA3 was found to be expressed mainly in the liver and adipocytes, with non-specific TG lipase and acylglycerol transacylase activities involved in TG hydrolysis in hepatocytes. Mutations in PNPLA3 antagonize normal proteasomal degradation and increase the risk of MASLD and its severity. TM6SF2 is a multiple transmembrane protein that is involved in lipid transfer, VLDL secretion and TC synthesis. For the rs58542926 variant of the TM6SF2 gene (E167K), a mutation in the TM6SF2 allele results in decreased VLDL secretion and fatty acid retention in the liver, leading to hepatic steatosis and progression of MASLD (99). Hydroxysteroid (17-β) dehydrogenase 13 (HSD17B13) is a gene encoding the hepatic lipid droplet protein 17β-hydroxysteroid dehydrogenase type 13 and it has been found that HSD17B13 expression is markedly upregulated in patients and mice with MASLD, suggesting that HSD17B13 usually produces a product that promotes hepatocyte injury (100). Other SNP genes associated with MASLD include neuromucin and glucokinase regulatory protein. In addition, the level of immunity and metabolic rate of an individual are genetically related.

MicroRNA factors

MicroRNAs are a class of RNA regulators, ~22 nucleotides in length and a growing number of studies on diet-induced and genetic (ob/ob) models of obese rodents and patients with severe MASLD suggest a potential role for miRNAs in MASLD pathogenesis and IR (101). A number of miRNAs have been identified, including miR-122 and miR-21 and their expression levels in both peripheral blood and liver have been associated with the development of MASLD (102). One study found that serum miR-122 was upregulated 7.2-fold in patients with MASH compared with healthy subjects and 3.1-fold compared with patients with simple steatosis. This suggests that serum miR-122 is an extra-hepatic feature of MASH. Moreover, in a mouse model without elevated alanine aminotransferase (ALT), the elevated serum miR-122 levels were positively associated with the severity of MASLD (103). It has been observed that the expression level of miR-21 in the liver of diet-induced MASH mice progressively decreased with disease progression, compared with the control group (103). It has been found that miR-27a attenuates neoplastic liver lipogenesis and obesity-induced MASLD by inhibiting the fatty acid synthase (FAS) gene and stearoyl coenzyme A desaturase-1 in the liver. Histological analyses also showed reduced lipid accumulation in the livers of mice with hepatic miR-27a overexpression, suggesting reduced hepatic steatosis; furthermore, trichrome staining of miR-27a overexpressed livers showed markedly reduced fibrosis and lower MASLD activity scores, reflecting improved development of MASLD (104). miR-155 is one of the miRNAs that can regulate KCs, which are involved in inflammatory processes that control innate and adaptive immunity in alcoholic and MASLD diseases (103). Szabo and Csak (105) proposed that miR-155 was a major regulator of inflammation and mice lacking miR-155 on methionine choline deficiency (MCD) with attenuated steatosis but no change in serum ALT or inflammation indicative of liver damage after diet-induced steatohepatitis. In another MASH model, however, miR-155 deficiency resulted in enhanced hepatic steatosis (105). A study has found miR-16 to be elevated in MASLD patients with simple steatosis and others have found that serum miR-16 is elevated in MASH patients and is associated with the stage of fibrosis (106). It has been observed that the expression of miR-197, miR-146b, miR-181d and miR-99a is markedly decreased in MASLD patients. Additionally, the hepatic levels of miR-301a-3p and miR-34a-5p increase monotonically from simple steatosis to MASH to cirrhosis. Conversely, miR-375 levels decrease monotonically during this progression (107). Some MASLD-related miRNAs, such as miR-149, had elevated expression levels in both FA-treated human hepatocellular carcinomas cells and MASLD animal models (102).

H2S factors

H2S is a mammalian endogenous signaling molecule that plays an important role in the pathophysiology of the liver. H2S has been reported to prevent the elevation of lipid peroxides malondialdehyde induced by MCD feeding (108). Wu et al (109) found that a high-fat diet reduced hepatic anti-oxidant defenses by downregulating glutathione peroxidase (GPx) and superoxide dismutase (SOD) activities. H2S effectively restored the activities of these enzymes and these results suggested that H2S could provide anti-oxidant effects against high-fat diet-induced hepatotoxicity. It was likewise demonstrated that H2S could inhibit hepatic fat accumulation through downregulation of FAS and upregulation of CPT-1, thus allowing the liver to recover from steatosis. In addition, H2S attenuated MASLD by upregulating the activities of anti-oxidant enzymes (GPx and SOD) and concluded that H2S could alleviate MASLD. It was concluded that H2S administration could alleviate the inhibition of MASLD accumulation by downregulating the expression of FAS and upregulating CPT-1 in obese mice fed a high-fat diet, thus ameliorating MASLD. In addition, H2S inhibited oxidative stress by increasing the activity of anti-oxidant enzymes such as GPx and SOD. These results suggested that H2S played an important role in regulating lipid and anti-oxidant metabolism (109).

Circadian pattern

Some studies have found a large number of circadian rhythmically expressed genes in the liver, which are involved in maintaining the metabolic balance of the body. MASLD is closely related to daily lifestyle. Irregular lifestyle may cause liver overload, including lack of sleep and little exercise. In addition, long-term intake of high-fat and high-protein foods, omitting breakfast, adding meals before bedtime and other bad dietary habits are also risk factors for MASLD (110). Sleep deprivation leads to an increased risk of morbidity and mortality (111). Epidemiologic studies have shown that sleep deprivation leads to altered glucose homeostasis, IR, weight gain, obesity, metabolic syndrome and DM, all of which are associated with MASLD. In experimental studies, it has been found that sleep disorders may induce MASLD through pro-inflammatory markers such as TNF-α, IL-1β and IL-6. In addition, sleep deprivation increases growth hormone-releasing peptide levels and decreases LEP levels, which increases appetite and further contributes to obesity and chronic insomnia activates the hypothalamo-pituitary-adrenal axis, which increases stress hormones, exacerbates IR and promotes the development of MASLD (112,113). Another study found that increased mRNA expression in hepatic biological clock genes, a decrease in levels of key metabolism-regulating enzymes, hepatic inflammation and steatosis can occur, which is associated with glucose and fatty acid metabolism (111). In the case of liver-specific knockout of BMAL1, which results in the loss of key metabolic gene oscillations in the liver and subsequently exacerbates oxidative damage to hepatocytes and induces IR. The glucocorticoid rhythm plays a key role in coordinating glucose, lipid and protein metabolism and it is an entrainment signal for the systemic circadian rhythm through the hypothalamic suprachiasmatic nucleus, the peripheral clock in the liver and adipose tissue. The kidney is regulated by the autonomic nervous system and rhythmic entrainment signals. It has been noted that the oscillation of the cellular redox state, independently of the biological clock transcriptional feedback loop and in the metabolic process, may control the circadian rhythm (114). Circadian rhythm disruption will lead to cellular dysfunction, which in turn affects the metabolic function of the liver (115). It induces metabolic dysfunction and the occurrence of obesity, fatty liver, metabolic syndrome and other conditions.

Psychosocial factors

Social stress is closely related to emotional and physical health and one of the factors that can trigger the development of MASLD is excessive stress (116). Some studies suggest that emotional problems such as anxiety and depression may have an effect on the progression of chronic liver diseases, including MASLD (117). A study by Youssef et al (118) confirmed the association of depression and anxiety with the severity of histologic features of MASLD by examining 567 patients, demonstrating that depression was markedly dose-dependent with more severe hepatocellular ballooning and that patients with subclinical depression were 2.1 times more likely to develop more severe hepatocellular ballooning than patients without depressive symptoms.

Diet and lifestyle-related factors

Studies have shown that hyperinsulinemia, lipid and lipoprotein metabolism changes caused by high-fat diet (HFD), high-carbohydrate diet, or high-fat and high-carbohydrate diets may promote the occurrence and development of MASLD (119). Yu et al (120) explored the relationship between dietary choline and MASLD, choline deficiency stimulated hepatic fat accumulation and increased dietary choline intake in normal-weight Chinese women associated with a reduced risk of MASLD. Soft drinks and meat intake were markedly associated with the increased risks of MASLD (121,122). High salt intake may affect MASLD by increasing plasma levels of triglyceride (TG) and it intake also stimulates endogenous fructose production. Lanaspa et al (123) found that a high salt diet activated the aldose reductase pathway in the liver, leading to endogenous fructose production, which induced LEP resistance with the development of the metabolic syndrome and MASLD. In a study based on a northern Chinese population, the correlation between salt intake and MASLD in DM patients was analyzed by Spearman analysis, which showed a positive correlation between salt intake and the incidence of MASLD, suggesting that the likelihood of MASLD in DM patients increases with increasing salt intake (124). A variety of mechanisms, including the anti-oxidant, anti-inflammatory and anti-fibrotic effects of coffee, may be related to the protective effects of coffee against MASLD (125). Studies have shown that caffeine may stimulate the hepatic autophagy-lysosomal pathway and induce fatty acid oxidation (126). However, how caffeine activates autophagic flux is still unclear (125). Active smoking has also been associated with a high risk of MASLD and a study has demonstrated the association of smoking history with advanced liver fibrosis in patients with MASLD (127). A cross-sectional study of 2,691 Chinese men found that ex-smokers and heavy smokers (≥40 cigarettes/day) had a higher prevalence of MASLD than never-smokers and some studies have reported that smoking is an independent risk factor for MASLD, which may exacerbate MASLD (128,129).

Experimental models of MASLD

Animal models

Commonly used laboratory animals include rats, mice and rabbits. Among them, rats and mice are the most widely used. Rats are represented by Wistar and Sprague Dawley (SD). There are more diverse mouse strains, with C57BL/6J being the most common. The present study summarized rat, mouse, rabbit, monkey, chicken, hamster and guinea pig models. The models summarized include diet-induced models, drug-induced models, special models and spontaneous models. Diet-induced models include HFD, MCD, choline-deficient amino acid-defined (CDAA) and atherogenic diet. It has the advantages of simple operation, low cost, repeatability and low mortality, but the disadvantage is that it is time-consuming. Drug-induced models usually use drugs such as streptozotocin (STZ), carbon tetrachloride (CCl4), LPS and tetracycline. Their advantages are short modeling time and economy, while its disadvantages are that the drugs have high toxicity. Special models mainly use gene knockout methods to make model animals prone to fatty liver. Compared with other models, the modeling time of spontaneous formation of fatty liver in special models is short, but it has requirements for the variety of animals and high cost. Spontaneous models mainly include monkeys. Monkeys are similar to humans in genes and can develop MASLD in old age without the need for a high-fat diet or drug induction. The modeling method of the nutritional model is to feed laboratory animals with high-sugar, high-fat and high-calorie diets or to create an MCD. The drug-induced model aims to build the model by administering drugs such as CCl4, tetracycline, polychlorinated biphenyl 118, so that the drugs exert their effects or even cause poisoning. The special strain model is to select certain diseases that can spontaneously form fatty liver, hyperlipidemia and other diseases closely related to MASLD or can spontaneously present symptoms relates to MASLD. The aforementioned modeling methods can be used independently and the pathological characteristics or disease phenotypes of the model can be reasonably selected according to different experimental requirements, they can also be used in combination to reduce the modeling time and improve the success rate of the model. The pathogenesis of MASLD is complex and a single animal model cannot fully mimic human MASLD. The ideal animal experimental model is a composite model formed by combining gene mutation or specific target gene modification with diet and drug/toxin induction. The phenotype of such a MASLD animal model is closer to that of human MASLD and the experimental results are more applicable to humans.

MASLD model in rats

Rats are of moderate size and have strong fertility and blood collection abilities. Currently, commonly used rat models for constructing MASLD diseases in laboratories include Wistar rats, SD rats and Zucker rats (Table I).

Table I

Metabolic dysfunction-associated steatotic liver disease models in different strains of rats.

Table I

Metabolic dysfunction-associated steatotic liver disease models in different strains of rats.

Authors,yearSpeciesTimeInducerSampleTestingMechanism(Refs.)
Salman et al, 2022Wistar rats14 weeksHFDBlood from heart, liver tissueSerum biochemical indicators, serum ELISA, real-time RNAHepatic steatosis with abnormal liver function and lipid profile(130)
Zhou et al, 2019Wistar rats13 weeksPCB118Liver tissueHistopathology, serum biochemical indicatorsALT↑, TG↑, TC↑, GLU↑, LDL-C↑, HDL-C↑, IL-1β↑, TNF-α↑, TGF-β1↑, MMP2↑, α-SMA mRNA↑, hepatic steatosis, inflammatory infiltration, structural disorders of liver lobules and fibrosis(131)
Stephen Robert et al, 2021Wistar rats8 weeksVegetable oil-CCl4Liver tissueHistopathology, serum biochemical indicators, oxidative stress indicators, gene expression analysisBody weight↑, ALT↑, ALP↑, AST↑, GGT-sensitive enzymes↑, liver injury, pathology showing hepatotoxicity of hepatocytes(132)
Zhang et al, 2018SD rats12 weeksHFDBlood from vena cava, liver tissueSerum biochemistry indicators, histopathology, ultrastructural observationLipids↑, blood glucose↑, high-density lipoprotein cholesterol↑, LDL-C↑, ALT↑, glutamine aminotransferase↑, insulin↑(133)
Lin et al, 2019SD rats6 weeksHFDBlood from heartSerum biochemical indicators, liver histopathologyBody weight↑, liver weight↑, liver index↑, GLU↑, HOMA-IR↑(134)
Jin et al, 2018SD rats9 weeks CCl4Blood from abdominal aortaSerum biochemical indicators, histopathologyYellowish liver surface, size↑, weight↑(135)
Chen et al, 2017SD rats10 weeksCDAALiver tissueSerum biochemical indicators, serum fibrosis indicators; histopathologyDegeneration, necrosis, regeneration, fatty liver changes, hepatic fibrosis(136)
Yan et al, 2015SD rats8 weeksHFD+STZBlood from abdominal aorta, liver tissueHistopathology, serum biochemical indicatorsBlood glucose↑, blood lipids↑, liver function↓, lipidosis(137)
Li and Li 2017SD rats12 weeksHigh fat solutionLiver tissueSerum biochemical indicators, histopathologyAST↑, ALT↑, AKP↑, TC↑, TG↑, LDL↑, fat infiltration, inflammatory response, liver weight↑(138)
Yong et al, 2020SD rats2 days0.9% tetracyclineBlood from abdominal aorta, liver tissueHistopathologySteatosis, hepatocellular damage(139)
Che and Han 2018SD rats32 daysMSGBlood from heart, liver tissueHistopathologyLiver weight↑, ALT↑, AST↑, ALP↑, hepatic steatosis(140)
Hakkak et al, 2022Zucker rats9 weeksAIN-93G dietLiver tissueSerum biochemical indicators, histopathology, liver weightALT↑, AST↑, body weight↑, fatty degeneration(141)
Matsumoto et al, 2021Zucker rats10 weeks53% corn amylumBlood from heartSerum biochemical indicators, histopathologyLipid droplets↑, ALP↑, ALT↑, AST↑(142)

[i] ALT, alanine aminotransferase; AST, aspartate aminotransferase; GLU, glucose; HFD, high-fat diet; IL-1β, HOMA-IR, homeostatic model assessment for iInsulin resistance; interleukin-1β; LDL, low-density lipoprotein; STZ, streptozotocin; TC, total cholesterol; TG, triglyceride; TGF-β, transforming growth factor-β; TNF-α, tumor necrosis factor-α.

MASLD model in mice

It is found that 93% of genomic regions of mice are arranged in an order similar to that of human beings and they are characterized by inexpensive feeding, rapid reproduction, easy modeling and small inter-individual differences that facilitate the observation of parallel experiments. MASLD mouse models can simulate different pathogenic factors and the development of MASLD at each stage of the disease, guide the search for the pathogenesis of MASLD and its potential therapeutic targets and also be used for the screening and evaluation of MASLD drugs, which are closely related to the research of MASLD. Mouse models have been induced by high-fat and high-sugar diets, subcutaneous injections of CCl4 and gastrointestinal nutritional solutions, with high-fat diets being the main modality. Mice commonly used in current experimental studies include C57BL/6J mice, Kunming mice and gene-deficient mice (Table II).

Table II

Metabolic dysfunction-associated steatotic liver disease models in different strains of mice.

Table II

Metabolic dysfunction-associated steatotic liver disease models in different strains of mice.

Authors, yearSpeciesTimeInducerSampleTestingMechanism(Refs.)
Xia et al, 2022C57BL/6N mice12 weeksHFDLiver tissueSerum biochemical indicators, histopathologyBody weight↑, TG↑, TC↑, FFA↑, hepatic steatosis(143)
Shi et al, 2022C57BL/6 mice24 weeksHFDBlood from heart, liver tissueSerum biochemical indicators, histopathologyLipid droplet deposits↑, vacuolate lesions(144)
Yu et al, 2022C57BL/6 mice8 weeksHFDLiver tissueSerum biochemical indicators, histopathologyLiver steatosis, ALT↑, AST↑, TC↑, TG↑(145)
De Minicis et al, 2017C57BL/6 mice4 weeks CDAA+CCl4Liver tissueSerum biochemical indicators, histopathologyLiver steatosis, liver fibrosis, tumours, IGF-2↑, SPP-1↑(146)
Li et al, 2013C57BL/6 mice8 weeks HFD+CCl4Blood from heart, liver tissueSerum biochemical indicators, histopathologyHepatic steatosis, fibrosis, abundant vacuolar hepatocytes(147)
Feng et al, 2021C57BL/6 mice23 daysDXM + HFDBlood from heart, liver tissueSerum biochemical indicators, histopathologyALT↑, AST↑, TG↑, TC↑, LDL↑, HDL↓(148)
Asgharpour et al, 2016C57BL/6J mice8 weeksHFDLiver tissueHistopathologyBody weight↑, IR↑, TG↑, LDL-C↑(149)
Ma et al, 2021C57BL/6J mice16 weeksNutrition SolutionLiver tissue, spleen, abdominal fatSerum biochemical indicators, histopathology, immune factorTG↑, TC↑, liver deposits↑, lipid vacuoles↑(150)
Jahn et al, 2019KM mice16 weeksHFD/HFSDBlood from heart, liver tissueSerum biochemical indicators, histopathologyLiver steatosis, liver fibrosis(151)
Wang et al, 2011KM mice8 weeksHFD+5% CCl4Blood from heart, liver tissueLiver morphology, histopathologyFatty liver disease, liver fibrosis(152)
Liu et al, 2020KM mice8 weeks CCl4Blood from heart, liver tissueSerum biochemical indicators, histopathologyTG↑, TC↑; inflammatory cell infiltration(153)
Malloy et al, 2013ob/ob mice14 weeksMRBlood from heart, liver tissueSerum biochemical indicators, histopathologyBody weight↑, TG↑, IR/HOMA ratio↑(154)
Son et al, 2021db/db mice3 weeksMCDBlood from heart, liver tissueSerum biochemical indicators, histopathologyLiver steatosis, inflammatory cell infiltration and fibrosis, TG↑, TNF-α↑, IL-6↑(155)
Yu et al, 2022ApoE−/− mice7 weeksHFDLiver tissueSerum biochemical indicators, histopathologyLiver steatosis, liver weight↑, liver index↑, TG↑, TC↑(156)
Song et al, 2020ApoE−/− mice4 weeksHFD+LPSBlood from abdominal aorta, liver tissueSerum biochemical indicators, histopathologyHepatic tissue inflammatory cell infiltration, hepatic tissue steatosis, inflammatory factors↑(157)
Yang et al, 2020LivKO mice14 weeksWestern diet feedingLiver tissueSerum biochemical indicators, histopathologyHepatomegaly, liver steatosis, fat droplets↑, inflammatory cell infiltration, TG↑, TC↑, body(158)
Piguet et al, 2015PTENL−/− mice32 weeksSDLiver tissueHistopathologyweight↑ BS↑, insulin↑, TG↑, TC↑(159)
Semba et al, 2013FLS mice12 weeksSDLiver tissueSerum biochemical indicators, histopathology, western blottingLipocalin-2, CXCL1 and CXCL9 expression, hepatic inflammatory cell infiltration, hepatocyte injury, liver steatosis(160)
Li et al, 2012PNPLA3 transgenic mice12 weeksRegular feedBlood from heart, liver tissueSerum biochemical indicators, histopathologyTC↑, TG↑(161)
Yin et al, 2006ICR mice72 hSD+Tetr acyclineBlood from heart, liver tissueSerum biochemical indicators, histopathologyLiver steatosis, TC↑, TG↑, β-oxidation of fatty acids↓(162)
Li et al, 2016.ICR mice8 weeksHFD dietBlood from heart, liver tissueSerum biochemical indicators, histopathologyTG↑, TC↑, HDL↑(163)
Yu et al, 2016FVB/N mice3 weeksAKT/SB plasmid+ salineBlood from heart, liver tissueSerum biochemical indicators, histopathologyFat vacuoles, cytoplasmic fat deposition, hepatocyte swelling(164)
López-Lemus et al, 2018BALB/c mice6 monthsHFDBlood from heart, liver tissueSerum biochemical indicators, histopathologyHepatic steatosis, degree of germinal epithelial loss changes(165)
Musolino et al, 2020DIAMOND mice27 weeksWDBlood from heart, liver tissueSerum biochemical indicators, histopathology, western blottingALT↑, TG↑, LDL-C↑, dyslipidaemia(166)
Chow et al, 2011ArKO mice6 weeksSoy-free dietBlood from heart, liver tissueSerum biochemical indicators, histopathologyHepatic steatosis, TG↑(167)
Komatsu et al, 2019AIM−/− mice12 weeksHFDLiver tissueSerum biochemical indicators, histopathologyFatty degeneration, inflammation, hepatic fibrosis(168)
Nguyen et al, 2021SREBP-1c KO mouse12 weeksHFDLiver tissueSerum biochemical indicators, histopathology, GTT, western blotting, RNA, H2SLiver steatosis(169)
Deguise et al, 2021Smn2b/− mice7 daysSDBlood from heart, liver tissueSerum biochemical indicators, histopathologyProtein output, complement, coagulation, iron homeostasis, IGF-1 metabolism disorders,(170)
Cano et al, 2011MAT1A-KO mice7 daysProtein rodent dietBlood from heart, liver tissueSerum biochemical indicators, histopathologyVLDL↑, TG↑, TC↑(171)
Heintz et al, 2019Cyp2b-null mice10 weeksHFDLiver tissue, lipidLipidomics, lipid quantification, lipid annotationKetosis↑, LP↑, TC↑, TG↑(172)
Edmunds et al, 2020LKO mice12 weeksHFDLiver tissue, lipidSerum biochemical indicators, histopathology; western blottingHepatic fat deposits↑, hepatic steatosis, TG↑, TC↑, blood glucose↑, decreased insulin sensitivity↑(173)
Yang et al, 2025 AlbSmad4−/− mice16 weeksHFDBlood from heart, liver tissueSerum biochemical indicators, histopathologyALT↓, TG↓, NEFA↓, ASK1-P38-JNK↓, CXCL1↓(174)

[i] ALT, alanine aminotransferase; AST, aspartate aminotransferase; CDAA, choline deficent aminoacid-defined; DXM, dexamethasone; FFA, free fatty acid; HFD, high-fat diet; IL-6, interleukin-6; LDL-C, low-density lipoprotein cholesterol; LPS, lipopolysaccharide; MCD, methionine choline deficiency; MR, methionine restriction; LP, leptin; SD, Sprague Dawley; TC, total cholesterol; TG, triglyceride; TNF-α, tumor necrosis factor-α; VLDL, very low-density lipoprotein; WD, western diet.

MASLD model in other animals

In addition to rodents, other animals are commonly used to establish MASLD models, including rabbits, monkeys and chickens (Table III). These models are helpful for studying the disease progression and providing crucial evidence for exploring the pathogenesis of MASLD and evaluating the efficacy of drugs (Table IV).

Table III

Metabolic dysfunction-associated steatotic liver disease models models in other rodents.

Table III

Metabolic dysfunction-associated steatotic liver disease models models in other rodents.

Authors, yearSpeciesTimeInducerSampleTestingMechanism(Refs.)
Yang et al, 2013Microtus fortis12 weeksHFDLiver tissueSerum biochemical indicators, histopathologyHepatocellular steatosis, diffuse steatosis(175)
Yu et al, 2021Meriones unguieulataus4 weeksHFDLiver tissueSerum biochemical indicators, histopathologyPunctate and focal necrosis of hepatocytes(176)
Bhathena et al, 2011Bio F1B hamster5 weeksHFDLiver tissueSerum biochemical indicators, histopathologyFat deposition↑, fatty liver, mild diabetes(177)
Cui et al, 2017Hamsters2 weeksHFDLiver tissueSerum biochemical indicators, histopathologyTG↑, hepatic adipogenesis from head↑, FFA↑, FFA oxidation↓, p-AMPKα↓, Sirt1↓(178)
Zhao et al, 2022Golden hamster2 weeksHFFCDLiver tissueSerum biochemical indicators, histopathology, western blottingLiver steatosis, lipid accumulation, abnormal serum, liver lipid markers(179)
Skat-Rørdam et al, 2021Guinea pig16 weeksHFDLiver tissueSerum biochemical indicators, histopathologyLiver steatosis, inflammation, significant abnormalities in serum, liver biochemical indicators(180)
Jin et al, 2022Guinea pig6 weeksHCDBlood from heart, liver tissueSerum biochemical indicators, histopathology, western blottingPathological changes in the liver, lipid accumulation and pathological changes in the aortic wall(181)
Ipsen et al, 2019Guinea pig25 weeksHFHSDBlood from heart, liver tissueRT-qPCRLiver fibrosis and cirrhosis(182)
Pedersen et al, 2023Guinea pig32 weeksHFDBlood from heart, liver tissueSerum biochemical indicators, histopathologyAUC↑, body weight↑, AST↑, TC↑, TG↑, fatty degeneration↑, inflammation↑(183)

[i] AST, aspartate aminotransferase; AUC, area under curve; FFA, free fatty acid; HCD, high-carbohydrate diet; HFD, high-fat diet; RT-qPCR, reverse transcription-polymerase chain reaction; SD, Sprague Dawley; TC, total cholesterol; TG, triglyceride.

Table IV

Metabolic dysfunction-associated steatotic liver disease models in other different animals.

Table IV

Metabolic dysfunction-associated steatotic liver disease models in other different animals.

Authors, yearSpeciesTimeInducerSampleTestingMechanism(Refs.)
Wang et al, 2018Japanese white rabbit12 weeksHFCDBlood from heart, liver tissueSerum biochemical indicators, histopathologyTC↑, TG↑, FFA↑(184)
Sturzeneker et al, 2019New Zealand rabbit8 weeksSpecific dietBlood from heart, liver tissueSerum biochemical indicators, histopathology, ICHdeposition↑, hepatocyte damage, hepatic fibrosis↑(185)
Nguyen et al, 2019New Zealand rabbit11 weeksHFDLiver tissueLiver fatLiver steatosis(186)
Zheng et al, 2018Macaca mulatta8 weeksSpecific dietLiver tissueUltrasound imaging of liverFat droplets↑, inflammatory cell infiltration, hepatocyte ballooning, hepatic fibrosis(187)
Cydylo et al, 2017Macaque7 yearsHFDLiver tissueH&E staining, MTC stainingDisorders of glucose-lipid metabolism, inflammatory response, hepatic fat deposition, hepatic fibrosis(188)
Kramer et al, 2015Marmoset6 yearsHFDBlood from heart, liver tissueSerum biochemical indicators, histopathology, ICH, liver lipidHepatomegaly with cytoplasmic vacuolation, IR, multifocal inflammation with ballooning hepatocyte degeneration(189)
Hamid et al, 2019Hy-Line Brown chicken32 weeksSpecific dietBlood from heart, liver tissueCecal microbiotaDysbiosis of cecal microbiota, AST↑, ALP↑, UA↑(190)
Zhu et al, 2022Hy-Line Brown chicken52 weeksCorn-soybean dietsLiver tissueHistopathology; western blottingChanges in the expression levels of miR-216, miR-217, TG, TC(191)
Tian et al, 2019Lu's blue-shelled egg-laying hens30 weeksSpecific dietLiver tissueSerum biochemical indicators, histopathology, western blottingTG↑, TC↑, miR-34a-5p positively associated with ACSL1 protein expression(192)
Song et al, 2022Rhode island white hen80 weeksSpecific dietBlood from heart, liver tissueSerum biochemical indicators, histopathologyDyslipidaemia, severe degree of hepatic steatosis, significant atherosclerotic plaque formation(193)

[i] AST, aspartate aminotransferase; HFD, high-fat diet; FFA, free fatty acid; IR, insulin resistance; TC, total cholesterol; TG, triglyceride; UA, uric acid.

Cell models

At present, animal models are the most commonly used in the research of MASLD. However, animal models have some unfavorable factors, such as large individual differences, long model-building cycles, difficult control of experimental conditions and differences between animals and humans. By contrast, cell models are superior in overcoming the aforementioned factors. According to experimental requirements, cell viability can be maintained all the time, which is close to the process of human diseases and the cellular mechanisms of diseases can be studied in a targeted manner. Therefore, establishing a cell model of MASLD in vitro has important theoretical significance and broad application value for studying its pathogenesis and disease development and further preventing and treating MASLD.

Existing model cells can be classified into human primary hepatocytes, animal primary hepatocytes, human hepatocytes, hepatocyte-like cells (HLCs), induced pluripotent stem cells (iPSCs) and liver slices. The culture forms can be divided into monoculture, co-culture and three-dimensional culture. Advantages and disadvantages of different cell lines and different culture methods were shown in Fig. 4.

The advantages and disadvantages of
different cell lines and different culture methods.

Figure 4

The advantages and disadvantages of different cell lines and different culture methods.

Single cell culture

Monoculture refers to the adherent culture of one type of cell, which is the most basic culture method. Compared with co-culture, monoculture has the advantages of simple operation, unlimited subculture and short model-building time, which is conducive to high-throughput drug screening experiments. Therefore, it is widely used in the research of fatty liver. Human hepatocyte cell lines, primary human hepatocytes (PHH), animal primary hepatocytes and HLC can be used for induction culture. The excessive deposition of extracellular matrix (ECM) leads to the occurrence of fibrosis (194), which is a major cause of liver function impairment. The limitation of monoculture is that it cannot explore the cell-ECM interaction and cannot well mimic the phenotype of liver fibrosis. The three-dimensional liver fibrosis cell model mentioned below can solve this problem and can also well simulate the human ECM structure in vitro (195).

PHH

PHH is generally extracted from surgically resected tissues and separated from the tissues by two-step collagenase perfusion or magnetic cell separation techniques (196), which can well mimic the physiological situation of the human liver. However, due to its rapid dedifferentiation in vitro and the need to obtain ethical permission for specimen acquisition, these reasons limit the use of primary hepatocytes. The improved medium supplemented with various chemicals solves the problem of limited traditional primary hepatocyte culture time. The PHH cultured with this medium can be comparable to PHH spheroids for at least 4 weeks and has the potential to simulate steatosis disease models (197).

Due to individual differences among different donors and the influence of various factors in the cell separation process, the experimental results may be unstable and have poor reproducibility. In addition, the culture conditions of human primary hepatocytes are relatively harsh, they can only be cultured in the short term and cannot be subcultured indefinitely, which is not conducive to the progress of the experiment. The scarcity of human liver samples and ethical issues also hinder the wide application of human primary hepatocytes.

Animal primary hepatocytes

Animal primary hepatocytes are often derived from the livers of rodents and the hepatocytes are isolated and extracted by perfusion or non-perfusion methods. The non-perfusion method mechanically separates and digests with collagenase or trypsin on small pieces of liver tissue. The non-perfusion method is simple to operate but often has the problem of incomplete digestion, while the perfusion method can greatly improve the viability of the isolated hepatocytes, with a survival rate as high as 80% (198). The two-step collagenase perfusion method is the standard procedure for isolating primary hepatocytes. The improved standard procedure can show higher cell viability (85-95%) through multi-parameter perfusion control (199). Animal primary cells solve the ethical and quantitative restriction problems of human primary hepatocytes. However, they are affected by different animal sources and batches, have poor reproducibility, are relatively sensitive and have harsh cultural conditions.

HLC

HLC can be derived from embryonic stem cells, induced pluripotent stem cells, mesenchymal stem cells, endodermal cells and hepatic progenitor cells. Hepatocyte nuclear and epidermal growth factors are often used to promote cell-directed differentiation. Oncostatin M (OSM) helps to promote the maturation of fetal hepatocytes and OSM is often added during the maturation stage to promote complete cell differentiation. Although the subsequent culture time of HLC is longer than that of traditional PHH, its induction time is long (2-4 weeks) and the cost is high (200). There is also no unified standard for the types and ratios of cytokines and nutrients during the differentiation process and these factors limit the application prospects of HLC.

Human hepatocyte cell lines

Hepatocyte cell lines have become high-quality in vitro models due to their advantages, such as strong repeatability, stable subculture and controllable conditions. Commonly used human hepatocyte cell lines include hepatocellular carcinoma cell lines such as HepG2, human hepatoma cells and protein phospholipase C, which are taken from human liver cancer tissues and normal hepatocyte cell lines such as LO2. Compared with primary hepatocytes, immortal cell lines have the following advantages: Stable growth, unlimited lifespan, stable phenotype and simpler culture conditions than primary hepatocytes and they are easy to standardize among different laboratories. FFA are lipotoxic substances and the most commonly used stimulants for inducing MASLD in vitro models (201). In addition, oleic acid (OA) and palmitic acid (PA) are also commonly used in the experiments. When studying MASLD, two hepatocyte cell lines can be selected for mutual verification to increase the credibility of the experimental results. Zhao et al (202) induced LO2 cells with 3% ethanol for 48 h and the levels of aspartate aminotransferase (AST), ALT, TG content and ROS markedly increased, proving that the cells acquired the characteristics of fatty liver and oxidative stress. Yu et al (203) induced HepG2 cells with 0.5 mmol/l OA for 24 h and TG and ROS markedly increased and the ability of cells to take up glucose markedly decreased, indicating that the cells showed symptoms of steatosis, oxidative stress and IR.

Induced pluripotent stem cell-derived hepatocyte-like cells

Induced pluripotent stem cells can differentiate into various somatic cells without causing immune rejection and ethical issues. The HLCs differentiated from human iPSCs can be comparable to primary hepatocytes in terms of morphological and functional characteristics and have the advantages of wide sources, large cultivation quantities and stable phenotypes, making up for the deficiencies of primary hepatocytes (204). However, the induced pluripotent hepatocyte technology still has problems, such as low efficiency of inducing cell transformation, possible gene mutations during the induction process and tumorigenic risks, which limit its wide application.

Liver slices

Precision liver slices are an in vitro culture technology between the organ and cell levels. Compared with cell models, liver slices are closer to the complex structure and composition of the human liver. Liver slices contain hepatocytes, KCs and hepatic stellate cells, creating a multi-cellular environment. The interaction among various hepatocytes can offer an improved simulation of the liver tissue environment. However, its disadvantage is that the survival time is short and it cannot be cultured for a long time.

Co-culture

The co-culture model can make up for the defects of the monoculture model. Introducing a second type of cell into the model increases the interaction between cells and can offer an improved simulation of the physiological functions of the liver. The advantages of the co-culture model are that it compensates for the shortcomings of the monoculture model, has simple culture conditions and enables high-throughput experiments. However, there are not a great number of existing co-culture model types and there is great potential for future development. Chen and Ma (205) established a co-culture model of HepG2 cells and THP-1 macrophages. Firstly, the macrophages adhered to a sterile glass slide and then the slide was placed into a 6-well plate inoculated with HepG2 cells. A mixed fatty acid with a concentration of 1 mmol/l (the ratio of PA:OA was 1:2) was added and an MASLD model was established after 24-h induction. Giraudi et al (206) co-cultured HuH7 and human hepatic stellate cells. After 24-h induction with fatty acids, the intracellular lipid accumulation increased and the expression of α-smooth muscle actin increased, indicating that the hepatic stellate cells were activated and the cells showed the characteristics of steatosis and fibrosis. In another study, human primary hepatocytes, hepatic stellate cells and KCs were co-cultured. The cells were stimulated with fatty acids, glucose, insulin and inflammatory cytokines to simulate MASH. In this model, the de novo lipogenesis in the cells was enhanced and the cells showed symptoms of oxidative stress, inflammation, fibrosis and activation of hepatic stellate cells (207). The co-culture of human primary hepatocytes and endothelial cells can support hepatocytes to maintain their phenotypic morphology, improve their specific functions and form capillary-like structures, which is more conducive to simulate the in vivo environment and studying the pathological mechanisms of fatty liver disease (208).

Three-dimensional culture

In the living liver tissue, there is substance transport and signal transduction between hepatocytes and the ECM and the ECM also plays a role in supporting the three-dimensional structure (209). When human primary hepatocytes are cultured in the traditional two-dimensional system for a long time, morphological changes occur due to epithelial-mesenchymal transition, resulting in the loss of hepatocyte polarity and related liver functions (210). The construction of a three-dimensional model is more complicated. For the three-dimensional model of PHH, KCs and hepatic stellate cells co-cultured with the help of a three-dimensional microphysiological system, at least two weeks of induction with FFA is required (211). The co-culture mode with multiple hepatocytes can more accurately simulate the liver microenvironment, but it requires more stringent culture conditions. In the case of few or no precedents, researchers are required to preliminarily explore the culture medium suitable for the survival and proliferation of multiple cells, at least considering the effects of pH value, osmotic pressure and gas environment. Three-dimensional culture is divided into three-dimensional monoculture and three-dimensional co-culture.

Three-dimensional monoculture

Kostrzewski et al (212) established a three-dimensional human primary hepatocyte model. The model group contained 0.6 mmol/l of fatty acids and both the control group and the model group were supplemented with appropriate concentrations of insulin and glucose. After 7 days of culture, the lipid accumulation in the model group increased markedly and further increased after 14 days. The fatty acids consumed by the model group were more than four times that of the control group. After model establishment, there was no significant change in the leakage of AST and ALT in the cells, indicating that the cell activity was normal and not affected by the modeling solution. The intracellular glutathione level, lactate dehydrogenase release, urea production and mitochondrial activity were also not affected by the modeling solution, proving that the cells in this three-dimensional model had complete functions. The expressions of genes CYP2E1, IGFBI, PDK4 and cholesterol 7 alpha-hydroxylase were all markedly increased, indicating that the model had the characteristics of steatosis. This model had a significant effect in detecting anti-fatty liver drugs such as metformin. The three-dimensional model can offer an improved simulation of the in vivo environment, has a longer culture time and is conducive to repeated induction experiments. The spherical three-dimensional model can be used for high-throughput screening experiments with high efficiency. The disadvantage is that it is very time-consuming and expensive to establish a three-dimensional model. Since it is still in the early stage of use, some functions remain to be verified.

Three-dimensional co-culture model

The three-dimensional co-culture model can be used to study more complex fatty liver phenotypes, such as inflammation and liver fibrosis. In the three-dimensional co-culture model, human hepatocytes are cultured together with non-parenchymal cells, which can simulate the interactions between various types of cells in the liver in three-dimensional space. The advantage of the three-dimensional co-culture model is that it can maintain important metabolic functions for a long time and induce fatty liver pathological phenotypes under relevant conditions.

Hepatic spheroids

Hepatic spheroids are spheres formed by the self-aggregation of hepatocytes cultured in suspension without a substrate-promoting cell attachment. The size of the spheroids can be controlled by changing the number of starting cells and this controllability provides convenience for standardized measurement. The cell sources for hepatic spheroid culture can be primary hepatocytes, hepatocyte cell lines and induced pluripotent stem cells. Single-cell and multi-cell cultures can both form hepatic spheroids. Compared with the planar culture of cells, the three-dimensional structure presents superior characteristics of the liver. HepG2 cell spheroids are more sensitive to hepatotoxic substances. Compared with HepG2 cells cultured in a monolayer, the half-maximal effective concentration values of a number of drugs are markedly reduced (213). Hepatic spheroids cultured from PHH can survive up to 7 weeks (214) and can achieve 100% specificity and 69% sensitivity in distinguishing hepatotoxic substances (215). Compared with hepatic spheroids derived from single-cell culture, multi-cell co-culture spheroids may have more development potential. Hepatic spheroids constructed by co-culturing PHH and NPC perform outstandingly in glycogen storage (216) and albumin synthesis, indicating that co-culture hepatic spheroids can offer an improved imitation of the structure and function of the human liver (217).

Organoids

Organoids have a self-renewing stem cell population and can exhibit some organ characteristics, which contain single or multiple cell types. It has been reported that organoids from adult hepatocytes can be cultured for up to 2.5 months (218). PHH and pluripotent stem cells can both simulate the adult liver. For the induction of pluripotent stem cells, a three-dimensional model can be constructed after monolayer culture induction to maturity or directly cultured in three dimensions at the undifferentiated stage (219). The induction of MASLD requires the addition of FFA to the culture medium. Liver organoids in this hepatotoxic environment for a long time show lipid droplet formation and TG accumulation and the upregulation of the expressions of genes related to lipid metabolism, inflammatory cytokines and fibrosis markers, showing the typical biochemical characteristics of MASLD progression (220). In addition, liver organoids can also be derived from liver cancer cells to simulate the cancer microenvironment, playing an important role in the field of anti-cancer research.

Liver-on-a-chip

Liver-on-a-chip technology is used to simulate the minimum functional unit of liver tissue, mainly constructed with the assistance of computer aided design software. When designing a microfluidic chip, factors such as flow rate, design size and aspect ratio need to be considered. In a three-dimensional culture system, microfluidic devices can reproduce the characteristics of the multi-cell microenvironment by controlling various parameters (221) and conducting cell culture and sample secretions. Although liver-on-a-chip technology is costly and the chip manufacturing process is complex, it can mimic the complex in vivo liver microenvironment, precisely control chemical gradients and manipulate parameters such as time, so it has broad development prospects in the research field of cell-matrix and cell-cell interactions.

3D-bioprinting

3D-bioprinting technology polymerizes various biological materials through inkjet, extrusion, laser polymerization, or digital light processing. In the field of liver model construction, a vascularized hepatic lobule model containing hepatocytes and endothelial cells has been developed through extrusion technology. Compared with a simple mixture of hepatocytes and endothelial cells, this model has increased albumin and urea secretion and a superior imitation of liver functions (222). With the development of 3D-bioprinting technology, the optimization of 'ink' has also been put on the agenda. Type I collagen is the main component of the ECM. When collagen I is mixed with thiolated hyaluronic acid in different ratios, it was found that the ratio of 3:1 can offer an improved maintenance of biological activity (223). The bioprinting method for MASLD needs to be further explored. One possible idea is to culture monolayers of steatotic hepatocytes as 'ink' to print three-dimensional structures or to induce lipotoxic substances after printing a liver model (224,225).

Chronic stress method

The chronic stress method uses one or several stress factors to change the emotional state of experimental animals (226). Although this method has a good effect, it takes a long time and only one emotional stimulus is applied to the rats for a long time, which is likely to cause the rats to adapt to the stimulus, resulting in insignificant emotional changes. To improve the errors caused by this method, unpredictable stress is now mostly used in the experiment, which can minimize errors and prevent rats from adapting to the stimulus. Sun et al (227) used fixed-time foot electroshock combined with noise stimulation to increase the diversity of chronic stress methods.

Chronic restraint method

The chronic restraint method is a way to restrict the activities of experimental animals by placing them in restraint devices, thus triggering emotional changes in the experimental animals. Some scholars have established the MASLD model by using long-term chronic restraint stress. They mainly applied restraint stress to rats by using plastic restrainers. To prevent the rats from breaking free, they also carried out punctures multiple times to make the restrainers fit closely with the rats. The rats were restrained for 6 h every day and this lasted for 9 weeks (228-231).

Model of blood stasis syndrome

Since MASLD is a chronic disease, according to the theory that chronic diseases often lead to deficiency and blood stasis, patients with this disease usually present with various syndromes of deficiency and blood stasis, among which the syndrome of blood stasis is the most common. Patients with this syndrome show symptoms such as masses in the right hypochondrium, loss of appetite, abdominal distension, weakness, loose stools, dull complexion and a pale and dark tongue. Liu et al (232) prepared the MASLD model of blood stasis syndrome by feeding rats with a high-fat and HFFD and intraperitoneally injecting 30 mg/kg STZ. The results showed that the rats' body weight firstly increased then decreased. Their fur was yellowish, dull, wilting and lacking luster. Their mental state was poor, they were inactive and easily startled and their claws were dull and had a low temperature. Moreover, the levels of ALT, AST, TC, TG and LDL-C in rats were increased and there was inflammatory infiltration in the cytoplasm of hepatocytes. Researchers have prepared the MASLD model of blood stasis type by using high-fat feed combined with leg-binding stimulation and reagent intervention methods. After 8 weeks, the rats showed manifestations such as dull hair and irritability, an increased liver index, diffuse fatty hepatocytes, varying degrees of macrovesicular and microvesicular steatosis and occasionally inflammatory cell infiltration. Meanwhile, they also had pathological manifestations consistent with MASLD (233).

Treatment for MASLD

Treatments for MASLD of synthetic drugs

In the process of treating MASLD, since the changes in liver tissue of most patients are at the stage of simple steatosis, the priority of treatment should be to solve the problem of overweight and improve the patient's IR. The secondary aim is to avoid 'additional blows' leading to MASH and acute liver failure and to reduce hepatic fat deposition in patients (234). Currently, the main clinical drugs for MASLD are lipid-lowering drugs, insulin sensitizers, anti-fibrotic and thyroid hormone drugs. Moreover, inhibition or reduction of microRNA activity by targeted drugs can alleviate the condition, which provides a new idea for the proposal of MASH typing and the development of individualized treatment modalities (235,236). The precise treatment of different subtypes of MASH patients with targeted drugs is also the development trend of future drug research (Fig. 5). The drugs for the targets in the treatment of MASLD disease are shown in Table V.

The means of drug therapy and
non-drug therapy of MASLD. HMG-CoA, hydroxy methylglutaryl coenzyme
A; LDL, low-density lipoprotein; MASLD, metabolic
dysfunction-associated steatotic liver disease; NAFLD,
non-alcoholic fatty liver disease; ROS, reactive oxygen
species.

Figure 5

The means of drug therapy and non-drug therapy of MASLD. HMG-CoA, hydroxy methylglutaryl coenzyme A; LDL, low-density lipoprotein; MASLD, metabolic dysfunction-associated steatotic liver disease; NAFLD, non-alcoholic fatty liver disease; ROS, reactive oxygen species.

Table V

Treatments for metabolic dysfunction-associated steatotic liver disease of synthetic drugs.

Table V

Treatments for metabolic dysfunction-associated steatotic liver disease of synthetic drugs.

Authors, yearNameTargetClinical ProgressClassification(Refs.)
Abenavoli et al, 2018OCAFXRPhase IIIInsulin sensitizer(249)
Mudaliar et al, 2013GS-9674FXRPhase IInsulin sensitizer(244)
Cui et al, 2020LJN452 FXRFXRPhase IIInsulin sensitizer(242)
Abenavoli et al, 2018LMB763FXRPhase IIInsulin sensitizer(249)
Lewis et al, 2015PioglitazonePPARγPhase IIIInsulin sensitizer(253)
Pawlak et al, 2015ElafibranorPPARα/δPhase IIIInsulin sensitizer(270)
Chen et al, 2015SaroglitazarPPARα/γPhase IIIInsulin sensitizer(252)
Harrison et al, 2020IVA337PPARα/δPhase IIInsulin sensitizer(254)
Knudsen and Lau, 2019LiraglutideGLP-1Phase IIInsulin sensitizer(246)
Lewis et al, 2015SomatostatinGLP-1Phase IIInsulin sensitizer(253)
Sumida et al, 2018INT-767FXR/TGR5Preclinical studyInsulin sensitizer(251)
Sanyal et al, 2010Vitamin EFXRPhase IIIOxidative stress and inflammation(257)
Sanyal et al, 2010PentoxifyllineTNF-αPhase IIOxidative stress and inflammation(257)
Kruger et al, 2018CenicrivirocCCR2/CCR5Phase IIOxidative stress and inflammation(267)
Zhu et al, 2021Amlexanox IKKε/TBK1Phase IIOxidative stress and inflammation(261)
Hu et al, 2023PXS-4728AVAP-1Phase IOxidative stress and inflammation(262)
Xiang et al, 2016NalmefeneTLR4Phase IIOxidative stress and inflammation(263)
Fernandes et al, 2014SolithromycinIntestinal lipasePhase IIIntestinal targeted preparation(279)
Chan et al, 2017OrlistatIntestinal lipasePhase IIIntestinal targeted preparation(260)
Chalasani et al, 2020GR-MD-02Galectin-3Phase IIAnti-fibrosis drug(273)
Bi et al, 2022SimtuzumabLOXL2Phase IIAnti-fibrosis drug(274)
Chalasani et al, 2020PirfenidoneTGF-βPreclinical studyAnti-fibrosis drug(273)
Dai et al, 2021OltiprazLXRαPhase IIAnti-fibrosis drug(288)

[i] CCR2/5, C-C motif chemokine receptor 2/5; FXR, farnesoid X receptor; GLP-1, glucagon-like peptide-1; OCA, obeticholic acid; PPAR, peroxisome proliferator-activated receptor; TGF-β, transforming growth factor-β; TGR 5, takeda G protein-coupled receptor 5; TLR 4, Toll like receptor 4; TNF-α, tumor necrosis factor-α.

Lipid-lowering drugs

Lipid metabolism disorder is the main clinical characteristic of MASLD patients and it is an important factor in the development of MASLD to hepatitis and cirrhosis, so lipid-lowering has become an important method of treatment for MASLD patients. At present, the best lipid-lowering effect of drugs is from statins, their mechanisms are mainly through the inhibition of hydroxy methylglutaryl coenzyme A (HMG-CoA), thereby blocking the synthesis of hepatic cholesterol and causing hepatic compensatory LDL receptor synthesis, so that the plasma LDL and LDL clearance increases. At the same time, statins can also exert anti-oxidant and anti-inflammatory effects (237). However, there are more problems in the use of statins, such as producing hepatotoxicity and muscle toxicity (238). In recent years, it has been found that inhibition of proprotein convertase-subtilisin-kexin type 9 (PCSK9) can markedly reduce LDL levels. Another experimental study found that the combination of statins with PCSK9 inhibitors was more effective than statins alone (239). In addition, ezetimibe and PCSK9 inhibitors (IIb, C) can be used in combination in statin-intolerant patients (240).

Insulin sensitizers

IR is a key link in the pathogenesis of MASLD, insulin sensitizers can effectively target the 'one strike' and increase insulin sensitivity to improve IR. Thus, enhancing the sensitivity of effector organs to insulin has become an important way to treat MASLD. Metformin, glucagon-like peptide-1 (GLP-1), thiazolidinediones and other drugs are currently more popular.

Metformin

Metformin contains two guanidinium groups in its structure and is a common oral metformin hypoglycemic drug in clinical practice. It lowers blood glucose by decreasing glucose production in the liver, increasing glucose metabolism and thus improving IR and has a certain effect on weight loss and regulation of lipid metabolism (241). However, metformin is generally not used alone in the treatment of MASLD (242). Some studies have shown that metformin can markedly reduce the levels of ghrelin, insulin and C-peptide in MASLD patients, but the improvement of liver fibrosis and liver inflammation is not significant (243,244). Although metformin is not the first choice for the treatment of MASLD, it is effective in improving body weight, lipids and glucose metabolism in patients with MASLD or MASLD combined with T2DM (245).

Agonists

GLP-1 is a well-established target in the field of diabetes. Liraglutide is a GLP-1 agonist used for the treatment of type 2 diabetes (1.2 to 1.8 mg/day) and obesity (3 mg/day). In addition to acting on the pancreas, GLP-1 can improve peripheral insulin sensitivity, participate in the physiological regulation of blood glucose, increase hepatic glucose uptake and glycogen synthesis, delay gastric emptying and reduce appetite and reduce the occurrence of atherosclerosis (246). Several GLP-1 drugs have been studied in clinical trials for their ability to promote insulin secretion, induce β-cell proliferation, inhibit postprandial glycogen release and delay gastric emptying. It has been shown that GLP-1 receptor agonists can enhance insulin sensitivity in effector organs, promote fatty acid oxidation, reduce lipogenesis and improve glucose metabolism after binding to the receptor (247).

Polyene phosphatidylcholine has anti-oxidant, anti-inflammatory properties, reduces hepatocyte damage and apoptosis and can effectively target the pathological symptoms caused by MASLD and its combination with metformin drugs for the treatment of patients with MASLD as well as MASH is markedly more effective than monotherapy (248). Obeticholic acid (OCA), as a nuclear transcription factor FXR agonist, has good application in the treatment of MASLD by affecting the gene expression of various metabolic and cellular damage pathways, such as lipid metabolism, IR and oxidative stress (249).

Thiazolidinediones

Pioglitazones are first-generation insulin sensitizers, which can specifically bind and activate PPAR-γ, improve IR, participate in glucose and lipid metabolism and inhibit the expression of LP and the expression of TNF-α, so as to reduce the hepatic lipid deposition and inhibit liver inflammation and fibrosis formation (250). Sumida and Yoneda (251) found that pioglitazone not only improved IR but also improved hepatic glucose-lipid metabolism, steatosis and inflammatory necrosis in patients with MASLD by activating PPAR-γ. The American Academy of Liver Diseases recommended pioglitazone in 2017 guidelines for the diagnosis and treatment of MASLD (252). Pioglitazone is not widely available because of safety concerns, including congestive heart failure, bone fractures in women and the possibility that pioglitazone may lead to an increased risk of bladder cancer (253). The second-generation insulin sensitizer MSDC-0602K did not show the side effects associated with the first-generation insulin sensitizers. According to the report of the latest phase IIb 52-week double-blind study (254), MSDC-0602K markedly reduced fasting glucose, insulin, glycosylated hemoglobin and markers of liver injury without dose-limiting side effects. However, it did not show a significant effect on the liver histology of the biopsy technique used. The information gained from this trial may provide a pre-basis for future study.

Drugs that regulate oxidative stress and inflammation
Anti-oxidants

In the disease process of MASLD, increased oxidative stress and defective anti-oxidant defense mechanisms promote liver injury as well as disease progression to MASH (255). Therefore, drugs with anti-oxidant activity can be used in the treatment of MASLD. Vitamin E is present in the phospholipid bilayer of cell membranes and helps to prevent oxidative damage caused by free radicals (256). The randomized controlled trial of PIVENS selected non-diabetic and non-cirrhotic MASH patients, conducted a 2-year trial of vitamin E (800 IU/d), pioglitazone and placebo and showed that vitamin E had significant histological improvements and that 36% of patients treated with vitamin E had improved steatosis, inflammation and remission of MASH (257). This suggested that vitamin E is favorable for the treatment of MASLD and MASH (258). However, long-term use of vitamin E may increase the risk of prostate cancer and hemorrhagic shock, so vitamin E should not be used for long-term treatment of MASH and MASLD (259). Another study found that the active ingredient in silymarin could improve liver fibrosis and reverse MASLD by reducing oxidative stress and inflammation (260). Although silymarin did not meet the primary endpoint in MASH patients, it markedly improved liver fibrosis compared with placebo (260).

ZSP1601 is a pan-phosphodiesterase inhibitor, which is the first small molecule innovative drug to obtain clinical trial approval for the treatment of MASH in China. Phase Ia clinical trials were conducted in healthy populations and ZSP1601 had a peak time of 1.5-2.5 h and a half-life of 6.34-8.64 h. The drug was well tolerated when compared with the placebo group (261). Its safety and pharmacokinetic profile supports further efficacy assessment in MASH patients. In the Ib/IIa clinical trial, ZSP601 was shown to have a favorable safety and tolerability profile in MASH patients, with a pharmacokinetic profile consistent with that of healthy subjects. After receiving 28 days of treatment, different dose groups of ZSP601 showed significant improvements in metabolism, hepatic fat content, inflammation and fibrosis biomarker indices (262), supporting the evaluation of its long-term safety and efficacy in a larger sample size of MASH patients.

Apoptosis inhibitors

Apoptosis is closely related to the formation of hepatic fibrosis, hepatocyte apoptosis releases excessive DNA fragments, which stimulates hepatic stellate cell activation and promotes the formation of fibrosis. In the liver, TNF mediates a number of biological reactions, reduces oxidative stress and fights against fibrosis and it is a potential therapeutic drug for MASLD. Pentoxifylline has been suggested as a potential therapeutic agent for MASH because of its ability to reduce oxidative stress production and its anti-fibrotic effects. Researchers treated patients with biopsy-proven MASH with either placebo or pentoxifylline 400 mg/day for 1 year in a small RDBPCT clinical trial (263). The results showed an improvement in histologic features in 38.5% of the subjects in the pentoxifylline treatment group (13.8% in the control group, P=0.036).

Apoptosis signal-regulating kinase 1 (ASK1) is a kinase in the MAP3 kinase family that activates the p38/JNK pathway downstream of TNF-α, leading to apoptosis and fibrosis. Studies in mice have shown that ASK-1 promotes TNF-α-mediated IR and steatosis (263), whereas inhibition of ASK1 ameliorates diet-induced steatosis and fibrosis (264). GS-4997 is an oral ASK1 inhibitor and a randomized, double-blind, open phase II clinical trial in patients with MASH and fibrosis is currently underway to evaluate the efficacy of 24 weeks of treatment with GS-4997 or GS-4997 and simtuzumab. ASK-1 inhibitors have been shown to improve inflammation and fibrosis in animal models of MASH, but Selonsertib's phase III trial does not meet its primary endpoint (265).

Immunomodulators

C-C motif chemokine receptor 2/5 (CCR2/5) are inflammatory chemokine receptors that are highly expressed in MASH. Chemokines induce cell migration in the direction of increased chemokine concentration and exert their biological effects by interacting with G-protein-linked transmembrane receptors. CCR2 on the surface of macrophages promotes inflammation, neovascularization and activation of hepatic stellate cells, exacerbating hepatic fibrosis, whereas CCR5 is mainly expressed by hepatic stellate cells, causing hepatic fibrosis (266). Inhibition of the activity of such receptors can reduce the inflammatory response in the liver and prevent or slow down liver fibrosis. Cenicriviroc (CVC) is an orally potent dual inhibitor of CCR2/5, which has been shown to ameliorate the progression of hepatic fibrosis in MASH mice (267). A phase IIb clinical trial with a sample of MASH patients showed that the anti-fibrotic results of CVC in the second year were consistent with those of the first year and remained significant in the two-year treatment group, demonstrating good tolerability and efficacy (268). CVC was in a phase III clinical study (AURORA, NCT03028740) aimed at evaluating its efficacy and safety in adult MASH patients with hepatic fibrosis (269). However, the AURORA phase III trial was forced to be terminated prematurely due to the failure to meet the objectives of its first phase results.

A total of two kinases, iκb kinase ε (IKKε) and recombinant tank binding kinase 1 (TBK1), are closely associated with obesity and inflammation and results from animal experiments demonstrated that the expression of IKKε and TBK1 was upregulated in adipose tissue of diet-induced obese animals, but IR, obesity and adiposity were not observed in IKKε and TBK1-deficient animals. During drug screening experiments for amlexanox, a drug used in the treatment of asthma and aphthous ulcers, it was discovered that amlexanox inhibits both IKKε and TBK12 kinases and reduces body weight in diet-induced or genetically obese animals by increasing energy expenditure. Amlexanox improves insulin sensitivity and reduces hepatic steatosis and inflammatory gene expression. A Phase II RDBPCT clinical trial is underway to evaluate the effect of amlexanox on liver steatosis in patients with diabetes, obesity and fatty liver disease (270).

Anti-inflammatory agents

Elafibranor is an agonist of PPAR-α and PPAR-δ. Studies have shown that elafibranor has the effect of improving insulin sensitivity, balancing blood glucose, regulating lipid metabolism and reducing inflammatory response (270). Ratziu et al (271) conducted an international, multicenter, randomized, double-blind, placebo-controlled study to evaluate the safety and efficacy of elafibranor in patients with MASH. MASH patients without cirrhosis were randomized in a 1:1:1 ratio to receive elafibranor 80, 120 mg, or placebo for 52 weeks. Liver biopsies, as well as clinical and laboratory evaluations, were performed on patients every 2 months. The results of the available studies suggest that elafibranor 120 mg/day for 1 year in moderate-to-severe patients may alleviate MASH fibrosis.

Anti-fibrotic agents

The degree of hepatic fibrosis is used to determine the progression of MASLD and most targeted clinical trials end in the improvement of fibrosis. In addition to acting on the pathogenesis of MASLD, some anti-fibrotic drugs have great potential. It has been shown that activation of HSCs can exacerbate the degree of hepatic fibrosis (272). Galactose lectin-3 (Gal-3) protein is closely associated with inflammation and liver fibrosis. Inhibiting Gal-3 can markedly reduce the symptoms of liver fibrosis in animals and clinical trials have also shown that it has an improved therapeutic effect on subjects with MASH and bridging fibrosis (273). Pirfenidone is a TGF-β inhibitor, clinically used in the treatment of idiopathic pulmonary fibrosis and another study found that it markedly improved the pathological features of liver fibrosis in animals (273). Belapectin (GR-MD-02) is a Gal-3 inhibitor developed by Galectin Therapeutics. Gal-3 protein is involved in a variety of inflammatory and fibrotic processes. Animal studies have demonstrated that Belapectin reduces fibrosis. Clinical trials have also demonstrated that it is well tolerated in subjects with MASH with bridging fibrosis and has beneficial prevention and treatment of esophageal varices in patients with MASH liver cirrhosis (273).

Regulation of intestinal flora

Dysbiosis of intestinal flora is closely associated with liver disease and cancer (274). Thus, manipulation of the microbiome through diet, probiotics, prebiotics and other medications is a viable line of research for the treatment of liver diseases, including MASLD. It has been found that prebiotics can reduce the accumulation of TG in the liver by decreasing the production of fat and decreasing the expression of genes such as FAS (275). In addition, probiotics can repair leaky gut and delay the progression of NAFL to MASH by modulating nuclear receptor expression and improving IR in liver and adipose tissue (276). In a clinical study by Malaguarnera et al (277), Bifidobacterium bifidum was found to markedly improve lipids, IR index, liver tissue inflammation and liver fibrosis in patients with MASH. Wang et al (278) found that Bifidobacterium trifidum tablets markedly reduced the levels of lipids, inflammatory factors, LPS and lipocalin in a rat model of MASH. Triclosan is a widely used antibiotic that ameliorates the ecological dysbiosis of the gut microbiome associated with MASLD by inhibiting pathogenic gram-negative bacteria. Solithromycin is a new-generation macrolide antibiotic clinically used to treat bacterial infections. In a mouse model of MASH induced by both diet and STZ, solithromycin was shown to ameliorate hepatocellular ballooning and inflammation, reduce blood glucose levels and downregulate hepatic gluconeogenic enzyme expression (279). Based on preclinical results, a Phase II open clinical trial study is currently underway to evaluate whether 13 weeks of solithromycin treatment in non-cirrhotic MASH patients can improve liver histologic characteristics.

Thyroid hormone analogs

Thyroid hormone receptor (THR-β) is highly expressed in hepatocytes and plays a key role in regulating the impaired metabolic pathway of MASH (280). The degree of hepatic hypothyroidism is higher in patients with MASH (281). This suggests that hepatic hypothyroidism may contribute to the development of MASH to some extent. Moreover, drugs targeting this condition are currently approved for MASH treatment.

Resmetirom (MGL-3196)

Resmetirom is an oral, liver-targeted THR agonist, which is 28-fold more selective for THR-β than triiodothyronine for THR-α (282). In MASH, Resmetirom's selectivity for THR-β contributes to hepatic-mediated thyroid hormone metabolism while avoiding the adverse effects of excess thyroid hormones in the heart and bone mediated through THR-α. Resmetirom and other thyroid hormone analogs reduce hepatic TG, inflammatory factors, fibrosis markers and ALT levels and reduce hepatic steatosis lipid peroxidation (283). In a multicenter, randomized, double-blind, placebo-controlled Phase III clinical trial of MAESTRO-MASH, the proportions of patients who achieved the primary endpoint of MASH symptom resolution without worsening of hepatic fibrosis at 52 weeks were 25.9 and 29.9% in the Resmetirom 80 and 100 mg dosage groups, respectively, compared with 9.7% in the placebo group (P<0.001). In addition, there were significant reductions in several lipid and lipoprotein metrics, including the key secondary endpoint of LDL-C (284). This trial was the first phase III clinical trial to achieve the two primary endpoints proposed by the FDA in patients with MASH. Accordingly, based on these key clinical data, the FDA approved the drug on March 15, 2024, for the treatment of patients with non-cirrhotic MASH with moderate to advanced liver fibrosis. This is the first MASH treatment approved by the FDA and represents a landmark breakthrough in the field.

VK2809

VK2809 is another THR-β selective agonist with specificity for liver tissue and good therapeutic potential. It belongs to a family of prodrugs that cleave and release potent thyroxine analogs in vivo. A randomized, double-blind, multicenter and placebo-controlled phase II study was conducted to evaluate the efficacy, safety and tolerability of VK2809 in reducing LDL-C and liver fat content in patients with primary hypercholesterolemia and MASLD (285). Patients were dosed for 12 weeks and subsequently discontinued for 4 weeks. Results showed a statistically significant 53.8% median reduction in liver fat in patients using 5 mg of VK2809 daily, 88% of cases in patients using VK2809 showed ≥30% reduction in liver fat at 12 weeks. In all dose groups, VK2809 had a favorable safety and tolerability profile, with no adverse reactions reported.

Treatment of MASLD with medicinal plants

Medicinal plants have important advantages in preventing and improving MASLD, so treatment of MASLD by medicinal plants is a potential therapeutic means (286). Medicinal plants have a history of several hundred years in the prevention and treatment of liver diseases. Compared with chemical pharmaceuticals, medicinal plants have a holistic concept and the idea of diagnosis and treatment, which is the most significant and basic feature of medicinal plants and shows its advantages in the treatment of MASLD (287). The treatment of MASLD by medicinal plants focuses on the holistic theory of liver preservation, which manifests itself in a variety of mechanistic forms, including anti-oxidative stress, lipid metabolism regulation, anti-inflammation, anti-fibrosis and intestinal microbiota regulation (288). It can be seen that the treatment of MASLD by medicinal plants is more individualized and comprehensive, which is in line with the characteristics of MASLD (Fig. 5).

Mechanisms of components of unitary medicines in treating MASLD

Single medicinal plants have multi-component, multi-target and multi-dimensional effects in anti-MASLD. Exploring the monomers of medicinal plants for the treatment of MASLD studies have found that natural compounds such as polysaccharides, terpenoids, glycosides, alkaloids, flavonoids, phenols and other natural compounds have a wide range of biological activities. By regulating various signaling pathways, they can exert comprehensive benefits such as inhibiting inflammatory response, improving lipid metabolism, reducing IR, then effectively alleviating the symptoms of MASLD. A summary of some studies on the prevention and treatment of MASLD by medicinal plants is shown in Table VI.

Table VI

Treatments for metabolic dysfunction-associated steatotic liver disease from natural products.

Table VI

Treatments for metabolic dysfunction-associated steatotic liver disease from natural products.

Authors, yearNameMechanismClassification(Refs.)
Deng et al, 2021PolygoninHepatic steatosis↓, IR↓, glycolipid metabolism↓, oxidative stress↓, maintain the stability of liver lysosomes; fatty acids↓Glycoside(289)
Pan et al, 2016Ginsenoside ReIR↓, regulate lipid metabolism, mitochondrial dysfunction↓, oxidative stress↓, NF-κB ↓Glycoside(290)
Li et al, 2021Panax notoginseng saponinAST↓, GPT↓, LDL↓, TC↓, TG↓, intestinal permeability↓, LPS↓, TLR4↓Glycoside(291)
Liu et al, 2020Astragaloside IVTLR4↓, MyD88↓, NF-κB↓, AMPK↑, SREBP-1c↓, IR↓, fat deposition↓Glycoside(292)
Zhao et al, 2019Cassia obtusifolia anthraquinone glycosideTLR4↓, NF-κB↓, skin inflammation↓, blood lipids↓, lipid peroxidation reaction↓Glycoside(293)
Paudel et al, 2018CassiasideAffects Nrf2, HepG2 cells death↓, regulating NF-κB, liver inflammation response↑Glycoside(294)
Zhang et al, 2015GeniposideAST↓, GPT↓, LDL↓, TC↓, TG↓, HDL↑, TNF-α↓, IL-6↓, liver damage↓, clear oxygen free radicals↑Glycoside(295)
Lee et al, 2014SalidrosideACCase↓, Malonyl CoA↓, CPT-1↑, oxidation of fatty acids↑, hepatic steatosis↓Glycoside(296)
Xu et al, 2020 GentiopicrosidepAMPKα↑, oxidative stress↓, regulate Nrf2, TLR4↓, NF-κB↓, regulate inflammatory response and lipid metabolismGlycoside(297)
Molteni et al, 2016DidyminTLR/NF-κB↓, release of cytokines↓Glycoside(298)
Wang et al, 2016BaicalinInflammatory cytokines↓, FFA↓Glycoside(299)
Zhen et al, 2018Total glucosides of paeonyTLR4↓, C-Jun N-terminal kinase↓, TG↓, TC↓Glycoside(300)
Shen et al, 2020GypenosideIL-17↓, TNF-α↓, IL-10↑, regulate Treg/Th17 immune dysfunctionGlycoside(301)
Li et al, 2020HesperidinSREBP-1c↓, PPARα↑, TLR4↓, IL-1↓, TNF-α↓Glycoside(302)
Park et al, 2010OleuropeinHepatic steatosis↓, PPARγ↓Glycoside(303)
Chen et al, 2025BuddleosideRegulates AMPK-TFEB, lipid accumulation↓, inflammatory response↓, improve liver functionGlycoside(304)
Chitturi et al, 2018QuercetinRegulates TC metabolism, CYP7A1 activity↑, cytochrome overexpression↓Flavonoid(234)
Yin et al, 2019GenisteinTLR4/NF-κB↓, IR↓, TNF-α↓Flavonoid(305)
Liu et al, 2021LuteolinLPS↓, TLR4↓, inflammatory cytokines↓Flavonoid(306)
Kou et al, 2024Chuanpianin NobiletinLipid metabolism, lipid accumulation↓Flavonoid(307)
He et al, 2018NaringeninBlood lipids↓, obesity↓, IL-1β↓, TNF-α↓, IL-6↓, lipid toxicity↓Flavonoid(308)
Pan et al, 2017PuerarinAMPK↑, PPARα↑, FFA↓, IR↓Flavonoid(309)
Liang et al, 2016Ginkgo flavoneReduce levels of NF-κB and TNF-αFlavonoid(310)
Huang et al, 2019TanshinoneTLR4/NF-κB↓, liver fat accumulation↓, regulate oxidative stressTerpenoid(311)
Zheng et al, 2020CurcumolTLR4/NF-κB↓, liver function↑, lipid levels↑Terpenoid(312)
Han et al, 2018CelastrolTLR4↑, MyD88↑, NF-κB↑, steatosis↓, inflammatory response↓Terpenoid(313)
Ye et al, 2024Asiatic acidTLR4/NF-κB/NLRP3↓, liver inflammation↓Terpenoid(314)
Malekinejad et al, 2023LupeolTLR4↓, IR↓, oxidative stress↓Terpenoid(315)
Brusotti et al, 2017Betulinic acidMediates synthesis of phospholipids and TG, hepatic steatosis↓, LPL↓, cellular lipid accumulation↓Terpenoid(316)
Hou et al, 2015Ursolic acidLipolysis, TG↓, TC↓Terpenoid(317)
Zhang et al, 2012 AndrographolideExpression of adipogenic genes↓, cellular lipid accumulation↓, lipid metabolism↑Terpenoid(318)
Zhang et al, 2019DiosgeninAMPK↑, PPARα↑, FFA↓, IR↓Terpenoid(319)
Zhang et al, 2019Hawthorn acidStress response↓, NF-κB↓Terpenoid(319)
Zhou et al, 2019Alisol AABCA1/ABCG1↑, SERBP-1c↓ ACC↓, Fas↓, CPT1↑, ACOX1↑, AMPKα↑, TG↓, blood lipid↓, IR↓, liver steatosis↓Terpenoid(320)
Ho et al, 2019Alisol BCell lipid toxicity↓ FXR↑, lipid accumulation↓, hepatic lobular inflammation↓, peri cellular fibrosis↓Terpenoid(321)
Li et al, 2022ResveratrolTLR4/NF-κB↓, lipid deposition↓, oxidative stress↓, mitochondrial activity↑, TG↓, regulate intestinal microbiota compositionPhenol(322)
Feng et al, 2019CurcuminLPS↓, TLR4/NF-κB↓, TNF-α↓, IL-1β↓, improve intestinal oxidative stress, intestinal inflammatory response↓Phenol(323)
Zhang et al, 2024GastrodinSREBP1c↓, TLR4↓, lipid synthesis↓Phenol(324)
Xiao et al, 2017Litchi pulp phenolicsClear free radicals, regulate mitochondrial dysfunctionPhenol(325)
Aithal et al, 2008 ProanthocyanidinsLipid accumulation↓, hepatic steatosis↓, oxidative stress↓Phenol(326)
Castellino et al, 2019Chlorogenic acidJNK↓, IR↓, relieve liver metabolic indexPhenol(327)
Tan et al, 2017Green tea polyphenolsAMPK↑, ACC↑, SREBP-1c↑, BG↓, lipid↓Phenol(328)
Chen et al, 2018Salvianolic acid BRegulates SIRT3 and SOD2 pathways, improve liver lipid deposition, oxidative stress response↓Phenol(329)
Zhang et al, 2021Berberine hydrochlorideTLR4↓, IκBα↓, liver inflammation↓Alkaloid(330)
Kim et al, 2015Total alkaloids of corydalis yanhusuoTLR4/NF-κB↓, inflammation↓, hepatic lipid↓Alkaloid(331)
Sun et al, 2021MatrineLipogenesis↓, mitochondrial dysfunction↓, ER stress↓Alkaloid(332)
Cani and Delzenne 2009BerberineImproves mitochondrial swelling, promote mitochondrial fusion↑, liver lipid content↓, liver inflammation↓Alkaloid(333)
Zhang et al, 2015NuciferinePASK↓, improve abnormal accumulation of glycerophospholipid and linoleic acid, regulate FFA permeation, oxidative stressAlkaloid(334)
Rahban et al, 2022Atractylodes macrocephala polysaccharideTLR4↓, IL-1β↓, IL-6↓, TNF-α↓Polysaccharide(335)
Zhong et al, 2022Astragalus polysaccharideTLR4↓, NF-κB↓, NLRP3↓, liver inflammation↓, lipid accumulation↓Polysaccharide(336)
Wang et al, 2024Pericarpium citri reticulatae polysaccharideInterferes with tlr4/md2 signaling pathway, macrophage infiltration↓, LPS↓, inflammation↓Polysaccharide(337)
Yu et al, 2021Dendrobium nobile polysaccharideTLR4↓, HO-1↓, inflammatory↓Polysaccharide(338)
Liu et al, 2024Polygala fallax Hemsl PolysaccharideTLR-4↓, NF-κB↓, peroxidation↓, inflammatory↓Polysaccharide(339)
Zhang and Jin, 2019Dicliptera chinensis polysaccharidePPAR-γ↑, TLR-4↓, NF-κB↓, TNF-α↓, IL-6↓, IL-1β↓Polysaccharide(340)
Zhao et al, 2022Sodium alginateLPS↓, TLR4↓, inflammatory↓Polysaccharide(341)
Liu et al, 2017Bupleurum chinenseActivation of hepatic stellate cells↓, injury of hepatocytes↓Polysaccharide(342)
Xie et al, 2022ZingeroneRegulates TLR4, Nrf2, mRNA, intervene lipid metabolism, oxidative stressOther chemicals(343)
Lv et al, 2018; Wei et al, 2016RheinMDA↓, SOD↑, GSH-Px↑, oxidative stress↓, fat metabolism↓, fat infiltration of hepatocytes↓, inflammatory factors↓Anthraquinone(344,345)
Dong et al, 2016EmodinIRE1α↓, TNF-α↓, IL-6↓, IL-1β↓, regulate PPAR-γ, IR↓, FXR↑, blood lipid content↓, obesity↓, fat deposition↓Anthraquinone(346)
Ye et al, 2022FucoxanthinRegulates AMPK/NRF2/TLR4, lipid metabolism, oxidative stress, inflammationAlkene(347)
Borstlap et al, 2018SulforaphaneLPS↓, LPS/TLR4↓, fat metabolism↓, inflammatory↓Isothiocyanate(348)
Yu et al, 2022Schisandrin ALPS↓, TLR4↓, fat metabolism↓, inflammatory↓Lignan(349)
Gu et al, 2021Coix seed oilp-AMPK↓, sepp1↓, apoer2↓, fat accumulation↓Volatile oil(350)

[i] AMP, adenosine 5'-monophosphate; AMPK, AMP-activated protein kinase; AST, aspartate aminotransferase; FFA, free fatty acid; FXR, farnesoid X receptor; GSH, glutathione; HFD, high-fat diet; IL-6, interleukin-6; IR, insulin resistance; IRE1α, inosital-requiring enzyme-1α; JNK, Jun n-terminal kinase; LDL, low-density lipoprotein; LPS, lipopolysaccharide; NF-κB, nuclear factor-kappa B; PPAR-α, peroxisome proliferator-activated receptor-α; SOD, superoxide dismutase; SREBP-1c, sterol regulatory element binding protein-1c; TC, total cholesterol; TG, triglyceride; TLR 4, Toll like receptor 4; TNF-α, tumor necrosis factor-α.

Mechanism of Chinese traditional patent medicine in treating MASLD

Based on evidence-based treatment, medicinal plants combine different chemical components for specific etiology and pathogenesis and exert the advantages of multi-targeting and synergistic effects in the prevention and treatment of MASLD. The mechanisms are summarized in the following Table VII.

Table VII

Treatments for metabolic dysfunction-associated steatotic liver disease of Chinese traditional patent medicines.

Table VII

Treatments for metabolic dysfunction-associated steatotic liver disease of Chinese traditional patent medicines.

Authors, yearNameSourceMechanismClassification(Refs.)
Pan et al, 2021 Shen-Ling-Bai-Zhu-SanPrescriptions People's Welfare PharmacyLPS↓, TLR4/NLRP3↓, inflammatory factors↓Classic prescription(351)
Du et al, 2024 Fu-Zi-Li-Zhong-WanSanyin Ji Yi Bingzheng Fang LunTLR4↓, TRAM/TRAF3↑, LDL-C↓, FFA↓, HDL-C↑Classic prescription(352)
Liu et al, 2021Er-Chen-TangPrescriptions People's Welfare PharmacyLPS↓, TLR4↓, inflammatory factor↓Classic prescription(353)
Zhang et al, 2020Si-Wu-TangSecrets of treating wounds and bone-settingTLR4/JNK↓, inflammatory↓, hepatic steatosis↓Classic prescription(354)
Su et al, 2024Xiao-Yao-WanPrescriptions People's Welfare PharmacyRegulates TLR4-NF-κB, IL-6↓, TNF-α↓Classic prescription(355)
Feng et al, 2023Astragalus powderSheng Ji Zong LuAMPK/TLR4↑, hepatic steatosis↓Classic prescription(356)
Xia et al, 2016Zhi-Gan-TangHospital preparationImproves ALT, AST, LDL-C, TG Self-formulated(357)
Lu et al, 2019Qing-Hua-TangHospital preparationHepatic steatosis↓ Self-formulated(358)
Xie et al, 2021 Chai-Hu-Bao-Gan-SanHospital preparationALT↓, AST↓, TC↓, IL-1↓, IL-6↓, TNF-α↓Self-formulated prescription(359)
Xu et al, 2019 Yin-Chen-Hao-TangHospital preparationRegulates lipid metabolism and intestinal microecological balanceSelf-formulated prescription(360)
Zhan et al, 2021 Ban-Xia-Yang-Xin-TangHospital preparationTC↓, TG↓, LDL-C↓, FGP↓, FINS↓, HOMA-IR↓, ISI↑, HOMA-β↑, fat accumulation↓, improve IRSelf-formulated prescription(361)
Wan et al, 2020 Bu-Shen-Tiao-Gan-TangHospital preparationBlood lipid↓, improves IRSelf-formulated prescription(362)
Chen et al, 2020Wu-Cao-FenHospital preparationImproves liver function and blood lipid indicatorsSelf-formulated prescription(363)
Mian and Yang, 2018 Huo-Xue-Jiang-Zhi-TangHospital preparationImprove TNF-α, FFASelf-formulated prescription(364)
Zhang et al, 2024 Jian-Pi-Jiang-Zhi-TangHospital preparationHMGB1/TLR4/NF-κB↓, improve hepatic inflammatory responseEmpirical effective prescription(365)
Wang et al, 2023 Zhi-Mu-Shan-Zha-YinHospital preparationTLR4/MyD88/NF-κB↓, hepatocyte apoptosis↓, lipid metabolism disorder↓, oxidative stress↓Empirical effective prescription(366)
Wang et al, 2024 Tong-Ping-Zhi-Fang-TangHospital preparationTLR4↓, MyD88↓, NF-κB↓, liver injury↓Empirical effective prescription(367)
Ding et al, 2021 Qing-Gan-Jiang-Zhuo-TangHospital preparationTLR4↓, NF-κB↓, MyD88↓, liver fat accumulation↓Empirical effective prescription(368)
Chen et al, 2020 Hu-Gan-Qing-Zhi-PianHospital preparationTLR4/MyD88↓, inflammatory factor↓Empirical effective prescription(369)
Ye et al, 2021Hua-Zhi-Fu-Gan granuleHospital preparationRegulate the expression of TLR4 and NF-κBChinese patent drug(370)
Guo et al, 2022Deng-Zhan-Sheng-Mai capsuleHospital preparationLPS↓, TLR4/NF-κB↓, inflammatory response↓Chinese patent drug(371)
Liao et al, 2019Zhi-Bi-Tai capsuleHospital preparationMAPK-ERK-TLR4↓, inflammatory response↓Chinese patent drug(372)
Yang et al, 2022Ku-Huang granuleHospital preparationTLR4↓, MyD88↓, regulate IRChinese patent drug(373)

[i] ALT, alanine aminotransferase; AST, aspartate aminotransferase; FFA, free fatty acid; IL-6, interleukin-6; IR, insulin resistance; LDL, low-density lipoprotein; LPS, lipopolysaccharide; NF-κB, nuclear factor-kappa B; TC, total cholesterol; TG, triglyceride; TLR 4, Toll like receptor 4; TNF-α, tumor necrosis factor-α.

Non-pharmacological treatments

Non-pharmacological treatments for MASLD are mainly life-related initiatives. In general, weight control, improved dietary patterns and lifestyle modifications can prevent the development of metabolic syndrome and MASLD (374). Proper aerobic exercise with intermittent high-intensity exercise can effectively reduce excess body fat. Hannah and Harrison (375) found that MASLD patients' weight loss of 3-5% slowed down the process of fatty liver lesions, weight loss of 5-7% may result in reversal of fibrosis. Obese and diabetic patients are more likely to plan their diets reasonably, adjust their daily routines and reduce their calorie intake to prevent MASLD at source (Fig. 5).

Dietary management

Dietary management should follow the principles of nutritional balance, limiting energy intake and adjusting dietary structure. Firstly, to supplement dietary fiber and minerals: Eat more food rich in low calorie, high protein and high fiber, mix coarse and fine grains and choose more vegetables, fruits, fungi and seaweed to ensure sufficient fiber intake and maintain intestinal microbial homeostasis. Secondly, to supplement vitamins and appropriate amounts of micronutrients: eat more food rich in vitamins and micronutrients selenium (purple cabbage, beans, seafood and seaweed) can accelerate the decomposition of lipid peroxidation and prevent liver fibrosis (376). Reduce the intake of carbohydrates: Eat less high sugar, high fat, high cholesterol, spicy, fried and salty food. In order to reduce the degree of obesity and promote the oxidation and decomposition of fatty acids, reduce the content of FFA and the burden of fat accumulation on the liver (377). In addition, fasting therapy: Patients only drink a moderate amount of water and eat low-calorie food for a certain period of time, plus a moderate amount of exercise to prevent and control MASLD, usually not more than 10 days. After the fasting period, the use of food from soft and easy to digest food gradually over to ordinary meals. For patients with fatty liver caused by excessive weight loss, food containing unsaturated fatty acids can be given to increase satiety and nutrition according to the patient's own situation and their fat intake should not exceed 30% (378). In other aspects: Develop good dietary habits, eat three meals a day regularly and quantitatively, avoid overeating diet, regular breakfast, reduce the number of fast food and dinner, drink more tea and coffee.

Exercise training

Exercise training therapy is an effective means to promote the recovery of MASLD patients, which has advantages of being green and inexpensive. Aerobic exercise can improve the rate of fat oxidation, increase fat consumption and reduce the accumulation of liver fat (379). At present, with the social and economic development and the increase of pressure at work, people are physically and mentally exhausted. It is easy to occur a variety of chronic diseases due to less exercise training or insufficient continuity, so it is more important to guide the MASLD patients to engage actively in exercise training. Li et al (380) used exercise training to intervene in MASLD, 3 times a week, each time lasting 3-4 times, lasting 40-60 min and the continuous intervention for 24 weeks, which could effectively improve the lipid metabolism level of patients with mild, moderate and severe MASLD, reduce blood glucose and improve the function of the liver.

Emotion management

Anxiety, depression and other negative emotions will increase the risk of MASLD (381). Therefore, patients should always maintain a stable state of mind, guard against arrogance and impatience, combine work and rest and learn to apply exercise, music, muscle relaxation in daily life and other methods to alleviate their own negative emotions, relax the body and mind and ultimately to achieve the effect of liver detoxification and restoration of liver function.

Maintain healthy lifestyle

Circadian rhythm disorders can cause MASLD, so patients should develop good living habits, including go to bed early, get up early, not stay up late, regular personal work and rest. At the same time, regular checks of blood glucose, blood lipids, blood pressure and abdominal ultrasound, in order to timely detection of the occurrence of MASLD and its risk factors (381).

Conclusion and future perspectives

As a complex metabolic disease involving multiple liver injuries, the incidence of MASLD is not only increasing year by year, but also tends affect younger and younger individuals, so its prevention and treatment should not be postponed. The pathogenesis of MASLD is complex and progressive, the liver histopathological diagnosis of MASLD is difficult to achieve in humans and suitable experimental models must be established to study MASLD, thus posing great challenges to its prevention and drug development. The present study reviewed the complex pathogenesis of MASLD, including IR, lipotoxicity, immune system disorders inducing inflammatory responses, intestinal flora disorders and genetic factors, as well as the study of Chinese medicine on the pathogenesis of MASLD (Fig. 6). MASLD arises from the interconnection and influence of multiple organs and systems, which can not only act individually, but also interact and synergize with each other to promote the development of MASLD and the specific triggering mechanisms still need to be explored at a deeper level.

The pathogenesis, model and
treatments of MASLD. HCC, hepatocellular carcinoma; HF, hepatic
fibrosis; HS, hepatic steatosis; MASH, metabolic
dysfunction-associated steatohepatitis; MASLD, metabolic
dysfunction-associated steatotic liver disease; NAFL, non-alcoholic
fatty liver; NAFLD, non-alcoholic fatty liver disease.

Figure 6

The pathogenesis, model and treatments of MASLD. HCC, hepatocellular carcinoma; HF, hepatic fibrosis; HS, hepatic steatosis; MASH, metabolic dysfunction-associated steatohepatitis; MASLD, metabolic dysfunction-associated steatotic liver disease; NAFL, non-alcoholic fatty liver; NAFLD, non-alcoholic fatty liver disease.

In recent years, the study of the molecular mechanism of MASLD has attracted much attention. The notch signaling pathway is a highly conserved transmembrane signaling pathway. It activates signal transduction through receptor-ligand interactions, which affected gene expression. The Notch signaling pathway plays a key role in hepatic lipid metabolism disorders, inflammatory responses and liver fibrosis (382). adenosine 5'-monophosphate-activated protein kinase (AMPK), as a core hub of energy metabolism, plays a crucial role in MASLD by regulating fatty acid synthesis, oxidation, autophagy and inflammation. Researches showed that various monomers of medicinal plants by inhibiting key factors of lipid synthesis (SREBP-1c, FAS and ACC), promoting fatty acid oxidation (PPARα and CPT-1), improving insulin resistance, enhancing autophagy (ULK1 and LC3) and inhibiting inflammation (NF-κB) and oxidative stress (Nrf2) (383). A large amount of basic researches shows that the NF-κB pathway is a crucial pathway for mediating inflammatory and oxidative stress responses and also plays an important role in the occurrence and development of MASLD. When the internal environment of the body changed, the levels of inflammatory factors in the body increases markedly. The NF-κB pathway is activated by activating the expression of inflammation and immune related receptors on the surface of liver cell membranes. The NF-κB pathway regulates various factors upstream and downstream, further mediating the inflammatory response, oxidative stress response, liver fibrosis, cell apoptosis, autophagy and pyroptosis of liver tissue to promote the process of MASLD (384). Chitosan markedly improves symptoms of MASLD by inhibiting lipid production, regulating inflammatory responses, alleviating oxidative stress, improving insulin resistance and regulating gut microbiota through multiple mechanisms. The specific mechanisms included that it inhibits the expression of transcription factors, activates the AMPK signaling pathway to promote fatty acid oxidation and inhibits the activity of signaling pathways such as PI3K/AKT/mTOR (385).

Through the continuous efforts of researchers at home and abroad, the study of experimental models of MASLD disease (including animal models and cellular models) has made great progress. Ideal disease models should be similar to human pathogenesis, simple, inexpensive, fast modelling, low animal mortality, high replication rate, good reproducibility and easy to use. In the process of practical application, researchers should understand the species differences between experimental animals and human beings, the formation mechanism of MASLD model and pathological changes are very different from those of human beings when they suffer from the disease and they need to select appropriate models according to specific research purposes and needs and consider the complications of the MASLD model. The TCM syndrome models, guided by TCM theory and based on the basic principle of evidence-based treatment, has obvious advantages in the treatment of fatty liver, but the formation principle, method and judgement index of the model are difficult to be standardized and need to be further verified (Fig. 6). Future research should establish a unified standard for the models and on the basis of improving the existing models, more efforts should be devoted to exploring new modelling methods, so that the experimental models can be more closely related to the characteristics of human MASLD, with a view to carrying out more in-depth research on the disease, elucidating its pathogenesis and an improved chance of preventing and treating MASLD.

Due to the complex pathogenesis of MASLD, there is no specific clinical drug and the use of a single treatment is limited. In the light of the current research results, individualized dietary control and lifestyle changes are the basic treatment for MASLD. A healthy lifestyle with good dietary habits, active treatment of the primary disease and selection of appropriate medication as an adjuvant therapy are conducive to intervening and controlling the development of MASLD and preventing further deterioration of the disease. Currently, effective therapeutic agents for MASLD are focused on multiple potential targets, with multi-target, multi-pathway therapeutic measures and targeted agents for key aspects of the target genes or metabolic pathways will also be applied in the clinic. However, various drugs are still in the research stage and in the future, clinical attention should not only be paid to the efficacy and safety of such drugs, but also need to further evaluate the effects of combining drugs with different mechanisms, so as to ultimately provide patients with a reasonable individualized medication plan (Fig. 6). In addition to synthetic drugs, natural medicines (TCM, Chinese medicine compounds), are characterized by multiple components and multiple targets of action, so the development of new natural medicines with multiple targets of action, maintaining a healthy composition of intestinal flora and promoting energy metabolism of the body, play a therapeutic role in MASLD from another angle.

Availability of data and materials

Not applicable.

Authors' contributions

KL was responsible for data curation and writing the original draft. CC, LW and ZS were responsible for formal analysis and methodology. JL, MA and QL were responsible for conceptualization and investigation. HH, QM, XW and RW were responsible for supervision, funding acquisition, writing, reviewing and editing. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Abbreviations:

ALT

alanine aminotransferase

AST

aspartate aminotransferase

CDAA

choline deficient amino acid-defined

DXM

dexamethasone

ER

endoplasmic reticulum

FFA

free fatty acid

IR

insulin resistance

MCD

methionine choline deficiency

MSG

monosodium glutamate

mTOR

mammalian target of rapamycin

NO

nitric oxide

OA

oleic acid

PHH

primary human hepatocytes

ROS

reactive oxygen species

SOD

superoxide dismutase

TG

triglyceride

THR-α

thyroid hormone receptor-α

TNF-α

tumor necrosis factor-α

Acknowledgements

Not applicable.

Funding

The present study was supported by National Natural Science Foundation of China (grant nos 82360682 and 82360825), Major Science and Technology Research Projects of Nanchang City [grant no. Hongkezi(2023)137-2], Training Project of Ganpo Juncai Support Plan for High level and High skilled Leading Talent in 2024 (grant no. 20240003188), 'Chunhui Plan' Collaborative Research Project of Ministry of Education of the People's Republic of China (grant nos. H ZKY20220385 and 202200200), Jiangxi Provincial Natural Science Foundation (grant nos. 20242BAB26172, 20224BAB206104 and 20224BAB206112) and Jiangxi University of Chinese Medicine Science and Technology Innovation Team Development Program (grant no. CXTD-22002).

References

1 

Ludwig J, Viggiano TR, McGill DB and Oh BJ: Nonalcoholic steatohepatitis: Mayo clinic experiences with a hitherto unnamed disease. Mayo Clin Proc. 55:434–438. 1980. View Article : Google Scholar : PubMed/NCBI

2 

Dong W, Jiang H, Li Y, Lv L, Gong Y, Li B, Wang H and Zeng H: Interpretable machine learning analysis of immunoinflammatory biomarkers for predicting CHD among NAFLD patients. Cardiovasc Diabetol. 24:2632025. View Article : Google Scholar : PubMed/NCBI

3 

Charu V, Liang JW, Mannalithara A, Kwong A, Tian L and Kim WR: Benchmarking clinical risk prediction algorithms with ensemble machine learning for the noninvasive diagnosis of liver fibrosis in NAFLD. Hepatology. 80:11842024. View Article : Google Scholar : PubMed/NCBI

4 

Gawrieh S, Vilar-Gomez E, Wilson LA, Pike F, Kleiner DE, Neuschwander-Tetri BA, Diehl AM, Dasarathy S, Kowdley KV, Hameed B, et al: Increases and decreases in liver stiffness measurement are independently associated with the risk of liver-related events in NAFLD. J Hepatol. 81:600–608. 2024. View Article : Google Scholar : PubMed/NCBI

5 

Marjot T: The endothelium as the gatekeeper of insulin's action on metabolic tissues: Implications for MASLD and MASH. J Hepatol. 83:808–810. 2025. View Article : Google Scholar : PubMed/NCBI

6 

Nadeau KJ, Mayer-Davis EJ, Gubitosi-Klug R, Zeitler PS, Kahn SE and Dabelea D; SEARCH TODAY, RISE and DISCOVERY study groups: Youth-onset type 2 diabetes: What we've learned from key youth-onset type 2 diabetes studies, what we still don't know, and why it is important. Diabetes Care. 48:1136–1149. 2025. View Article : Google Scholar : PubMed/NCBI

7 

The Lancet Gastroenterology Hepatology: Redefining non-alcoholic fatty liver disease: What's in a name? Lancet Gastroenterol Hepatol. 5:4192020. View Article : Google Scholar : PubMed/NCBI

8 

Lazarus JV, Newsome PN, Francque SM, Kanwal F, Terrault NA and Rinella ME: Reply: A multi-society Delphi consensus statement on new fatty liver disease nomenclature. Hepatology. 79:E93–E94. 2024. View Article : Google Scholar

9 

Zhang F, Zhang X, Lv B, Sun R, Zhao T and He N: Changes in epidemiology and treatment outcomes after the academic renaming from NAFLD to MAFLD/MASLD. Chin Hepatol. 7:900–903. 2025.In Chinese.

10 

Bae JC: No More NAFLD: The term is now MASLD. Endocrinol Metab (Seoul). 39:92–94. 2024. View Article : Google Scholar : PubMed/NCBI

11 

Drygalski K: Pharmacological treatment of MASLD: Contemporary treatment and future perspectives. Int J Mol Sci. 26:65182025. View Article : Google Scholar : PubMed/NCBI

12 

Li H, Zhang Z, Jiang X, Li L, Hu T, Wang B and Deng Z: Meta-analysis and data mining of Chinese herbal medicines for the treatment of metabolic dysfunction-associated steatotic liver disease. Acad J Guangzhou Med Univ. 53:17–26. 2025.In Chinese.

13 

Younossi ZM, Golabi P, Paik JM, Henry A, Dongen CV and Henry L: The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): A systematic review. Hepatology. 77:1335–1347. 2023. View Article : Google Scholar : PubMed/NCBI

14 

Zhou F, Zhou J, Wang W, Zhang XJ, Ji YX, Zhang P, She ZG, Zhu L, Cai J and Li H: Unexpected rapid increase in the burden of NAFLD in China from 2008 to 2018: A systematic review and meta-analysis. Hepatology. 70:1119–1133. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Benedict M and Zhang X: Non-alcoholic fatty liver disease: An expanded review. World J Gastroenterol. 9:715–732. 2017.

16 

Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L and Wymer M: Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 64:73–84. 2016. View Article : Google Scholar

17 

Fang YL, Chen H, Wang CL and Liang L: Pathogenesis of non-alcoholic fatty liver disease in children and adolescence: from 'two hit theory' to 'multiple hit model'. World J Gastroenterol. 24:2974–2983. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Liu X and Gao H: Research progress of non-alcoholic fatty liver disease. J Hubei Univ Sci Technol (Med Sci). 33:364–368. 2019.In Chinese.

19 

Tilg H and Moschen AR: Evolution of inflammation in nonalcoholic fatty liver disease: The multiple parallel hits hypothesis. Hepatology. 52:1836–1846. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Buzzetti E, Pinzani M and Tsochatzis EA: The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism. 65:1038–1048. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Mariana MV and Cortez-Pinto H: Non-alcoholic fatty liver disease: What the clinician needs to know. World J Gastroenterol. 20:12956–12980. 2014. View Article : Google Scholar

22 

Fang C, Cai X, Hayashi S, Hao S, Sakiyama H, Wang X, Yang Q, Akira S, Nishiguchi S, Fujiwara N, et al: Caffeine-stimulated muscle IL-6 mediates alleviation of non-alcoholic fatty liver disease. Biochim Biophys Acta Mol Cell Biol Lipids. 1864:271–280. 2019. View Article : Google Scholar

23 

Horton JD, Goldstein JL and Brown MS: SREBPs: Activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest. 109:1125–1131. 2002. View Article : Google Scholar : PubMed/NCBI

24 

Thanapirom K and Tsochatzis EA: Non-alcoholic fatty liver disease (NAFLD) and the quest for effective treatments. Hepatobiliary Surg Nutr. 8:77–79. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Wilkinson AL, Qurashi M and Shetty S: The role of sinusoidal endothelial cells in the axis of inflammation and cancer within the liver. Front Physiol. 11:9902020. View Article : Google Scholar : PubMed/NCBI

26 

Furuta K, Guo Q, Hirsova P and Ibrahim SH: Emerging roles of liver sinusoidal endothelial cells in nonalcoholic steatohepatitis. Biology (Basel). 9:3952020.PubMed/NCBI

27 

Nasiri-Ansari N, Androutsakos T, Flessa CM, Kyrou I, Siasos G, Randeva HS, Kassi E and Papavassiliou AG: Endothelial cell dysfunction and nonalcoholic fatty liver disease (NAFLD): A concise review. Cells. 11:25112022. View Article : Google Scholar : PubMed/NCBI

28 

Chen G, Cai X and Lu L: Role of hepatic sinusoidal endothelial cells in nonalcoholic steatohepatitis. Chin Hepatol. 29:998–1002. 2024.In Chinese.

29 

Hammoutene A and Rautou PE: Role of liver sinusoidal endothelial cells in non-alcoholic fatty liver disease. Hepatology. 70:1278–1291. 2019. View Article : Google Scholar

30 

Liu J, Ren CQ, Feng YL and He JR: Relationship between liver fibrosis and insulin resistance in patients with type 2 diabetes and nonalcoholic fatty liver disease. Chin J Clin Res. 34:443–448. 2021.In Chinese.

31 

Wang Y, Xu D, Xu L and Wu Y: Peripheral blood TLR4, NF-κB mRNA in patients with type 2 diabetes mellitus complicated with non-alcoholic fatty liver disease and their correlations with insulin resistance. Lab Med Clin. 19:925–929. 2022.In Chinese.

32 

Dashti N, Williams DL and Alaupovic P: Effects of oleate and insulin on the production rates and cellular mRNA concentrations of apolipoproteins in HepG2 cells. J Lipid Res. 30:1365–1373. 1989. View Article : Google Scholar : PubMed/NCBI

33 

Li X, Ni X and Hua J: Effect of hepatocyte fatty degeneration induced by free fatty acid on macrophage polarization. J Clin Hepatol. 37:385–389. 2021.In Chinese.

34 

Zhang HB and Wang Y: Research progress of non-alcoholic fatty liver disease. Hainan Med J. 28:1651–1653. 2017.In Chinese.

35 

Ye Y and Yu J: Adiponectin level, its gene polymorphism and nonalcoholic fatty liver disease. Basic Clin Med. 34:1285–1288. 2014.In Chinese.

36 

Zou WS, Chen J and Zhang H: Expression and significance of osteopontin, leptin, visfatin and adiponectin in bile and gall-bladder mucosa. China J Mod Med. 29:30–35. 2019.In Chinese.

37 

Shah SV and Fonseca VA: Iron and diabetes revisited. Diabetes Care. 34:1676–1677. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Zhan X: Research on the relationship between the iron metabolism parameters level and TCM syndrome types in nonalcoholic fatty liver disease. Fujian Univ Tradit Chin Med. 2020.In Chinese.

39 

Nelson JE, Wilson L, Brunt EM, Yeh MM, Kleiner DE, Unalp-Arida A and Kowdley KV; Nonalcoholic Steatohepatitis Clinical Research Network: Relationship between the pattern of hepatic iron deposition and histological severity in nonalcoholic fatty liver disease. Hepatology. 53:448–457. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Baumeister SE, Völzke H, Marschall P, John U, Schmidt CO, Flessa S and Alte D: Impact of fatty liver disease on health care utilization and costs in a general population: A 5-year observation. Gastroenterology. 134:85–94. 2008. View Article : Google Scholar

41 

Pagano C, Soardo G, Esposito W, Fallo F, Basan L, Donnini D, Federspil G, Sechi LA and Vettor R: Plasma adiponectin is decreased in nonalcoholic fatty liver disease. Eur J Endocrinol. 152:113–118. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Cohen ED and Fisher Edward A: Lipoprotein metabolism, dyslipidemia, and nonalcoholic fatty liver disease. Semin Liver Dis. 33:380–388. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Wang YY, Liang FX, Lu W, Song YJ, Ren JF, Zhou YD, Huang XX and Yang SR: Effects of electroacupuncture on insulin sensitivity and ediponectin/resistin gene expression in adipose tissue of obese insulin resistant rats. J Basic Chin Med. 27:1444–1450. 2021.In Chinese.

44 

Xu MX, Wang M and Yang WW: Gold-quercetin nanoparticles prevent metabolic endotoxemia-induced kidney injury by regulating TLR4/NF-kappaB signaling and Nrf2 pathway in high fat diet fed mice. Int J Nanomedicine. 12:327–345. 2017. View Article : Google Scholar :

45 

Mu H, Zhang Z, Liang C and Liu N: Study on the effect and its mechanism of carvedilol on leptin-induced activation and proliferation of human hepatic stellate cells. China Pharm. 28:2620–2624. 2017.In Chinese.

46 

Begriche K, Massart J, Robin MA, Bonnet F and Fromenty B: Mitochondrial adaptations and dysfunctions in nonalcoholic fatty liver disease. Hepatology. 58:1497–1507. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Scott FL, Stec B, Pop C, Dobaczewska MK, Lee JJ, Monosov E, Robinson H, Salvesen GS, Schwarzenbacher R and Riedl SJ: The Fas-FADD death domain complex structure unravels signalling by receptor clustering. Nature. 457:1019–1022. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Arroyave-Ospina JC, Wu Z, Geng Y and Moshage H: Role of oxidative stress in the pathogenesis of non-alcoholic fatty liver disease: Implications for prevention and therapy. Antioxidants (Basel). 10:1742021. View Article : Google Scholar : PubMed/NCBI

49 

Dong S, Liu P and Sun MY: Role of 'two-hit' in non-alcoholic fatty liver disease. J Clin Hepatol. 28:551–555. 2012.In Chinese.

50 

Liu Y: Oxidative stress and hepatic protection. Chin J Gastroenterol Hepatol. 20:594–597. 2011.In Chinese.

51 

Lamkanfi M, Sarkar A, Vande Walle L, Vitari AC, Amer AO, Wewers MD, Tracey KJ, Kanneganti TD and Dixit VM: Inflammasome-dependent release of the alarmin HMGB1 in endotoxemia. Immunology. 185:4385–4392. 2010.

52 

Mridha AR, Haczeyni F, Yeh MM, Haigh WG, Ioannou GN, Barn V, Ajamieh H, Adams L, Hamdorf JM, Teoh NC and Farrell GC: TLR9 is up-regulated in human and murine NASH: Pivotal role in inflammatory recruitment and cell survival. Clin Sci (Lond). 131:2145–2159. 2017. View Article : Google Scholar : PubMed/NCBI

53 

Sharifnia T, Antoun J, Verriere TG, Suarez G, Wattacheril J, Wilson KT, Peek RM Jr, Abumrad NN and Flynn CR: Hepatic TLR4 signaling in obese NAFLD. Am J Physiol Gastrointest Liver Physiol. 309:G270–G278. 2015. View Article : Google Scholar : PubMed/NCBI

54 

Rocha DM, Caldas AP, Oliveira LL, Bressan J and Hermsdorff HH: Saturated fatty acids trigger TLR4-mediated inflammatory response. Atherosclerosis. 244:211–215. 2016. View Article : Google Scholar

55 

Glass CK and Olefsky JM: Inflammation and lipid signaling in the etiology of insulin resistance. Cell Metab. 15:635–645. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Seimon TA, Nadolski MJ, Liao XH, Magallon J, Nguyen M, Feric NT, Koschinsky ML, Harkewicz R, Witztum JL, Tsimikas S, et al: Atherogenic lipids and lipo proteins trigger CD36-TLR2-dependent apoptosis in macrophages un dergoing endoplasmic reticulum stress. Cell Metab. 12:467–482. 2010. View Article : Google Scholar : PubMed/NCBI

57 

Miura K and Ohnishi H: Role of gut microbiota and toll-like receptors in nonalcoholic fatty liver disease. World J Gastroenterol. 20:7381–7391. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Ashraf NU and Sheikh TA: Endoplasmic reticulum stress and oxidative stress in the pathogenesis of non-alcoholic fatty liver disease. Free Radic Res. 49:1405–1418. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Amir M and Czaja MJ: Autophagy in nonalcoholic steatohepatitis. Expert Rev Gastroenterol Hepatol. 5:159–166. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Gäbele E, Dostert K, Hofmann C, Wiest R, Schölmerich J, Hellerbrand C and Obermeier F: DSS induced colitis increases portal LPS levels and enhances hepatic inflammation and fibrogenesis in experimental NASH. Hepatology. 55:1391–1399. 2011. View Article : Google Scholar

61 

Simonen P, Kotronen A, Hallikainen M, Sevastianova K, Makkonen J, Hakkarainen A, Lundbom N, Miettinen TA, Gylling H and Yki-Järvinen H: Cholesterol synthesis is increased and absorption decreased in non-alcoholic fatty liver disease independent of obesity. Hepatology. 54:153–159. 2011. View Article : Google Scholar

62 

Enjoji M, Yasutake K, Kohjima M and Nakamuta M: Nutrition and nonalcoholic fatty liver disease: The significance of cholesterol. Int J Hepatol. 2012:9258072012. View Article : Google Scholar : PubMed/NCBI

63 

Subramanian S, Goodspeed L, Wang S, Kim J, Zeng LX, Ioannou GN, Haigh WG, Yeh MM, Kowdley KV, O'Brien KD, et al: Dietary cholesterol exacerbates hepatic steatosis and inflammation in obese LDL receptor deficient mice. J Lipid Res. 52:1626–1635. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Ersoy BA, Maner-Smith KM, Li Y, Alpertunga I and Cohen DE: Thioesterase-mediated control of cellular calcium homeostasis enables hepatic ER stress. J Clin Invest. 128:141–156. 2018. View Article : Google Scholar :

65 

Ghemrawi R, Battaglia-Hsu F and Arnold C: Endoplasmic reticulum stress in metabolic disorders. Cells. 7:632018. View Article : Google Scholar : PubMed/NCBI

66 

Henkel AS: Unfolded protein response sensors in hepatic lipid metabolism and nonalcoholic fatty liver disease. Semin Liver Dis. 38:320–332. 2018. View Article : Google Scholar : PubMed/NCBI

67 

DeZwaan-McCabe D, Sheldon RD, Gorecki MC, Guo DF, Gansemer ER, Kaufman RJ, Rahmouni K, Gillum MP, Taylor EB, Teesch LM and Rutkowski DT: ER Stress inhibits liver fatty acid oxidation while unmitigated stress leads to anorexia-induced lipolysis and both liver and kidney steatosis. Cell Rep. 19:1794–1806. 2017. View Article : Google Scholar : PubMed/NCBI

68 

Maillo C, Martín J, Sebastián D, Hernández-Alvarez M, García-Rocha M, Reina O, Zorzano A, Fernandez M and Méndez R: Circadian- and UPR-dependent control of CPEB4 mediates a translational response to counteract hepatic steatosis under ER stress. Nat Cell Biol. 19:94–105. 2017. View Article : Google Scholar : PubMed/NCBI

69 

Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP and Ron D: Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science. 287:664–666. 2000. View Article : Google Scholar : PubMed/NCBI

70 

Li Y, Huang B, Jiang X, Chen W, Zhang J, Wei Y, Chen Y, Lian M, Bian Z, Miao Q, et al: Mucosal-associated invariant T cells improve nonalcoholic fatty liver disease through regulating macrophage polarization. Front Immunol. 9:19942018. View Article : Google Scholar : PubMed/NCBI

71 

Ma J, Zhou Q and Li H: Gut Microbiota and nonalcoholic fatty liver disease: Insights on mechanisms and therapy. Nutrients. 9:11242017. View Article : Google Scholar : PubMed/NCBI

72 

Harte AL, da Silva NF, Creely SJ, McGee KC, Billyard T, Youssef-Elabd EM, Tripathi G, Ashour E, Abdalla MS, Sharada HM, et al: Elevated endotoxin levels in non-alcoholic fatty liver disease. J Inflammation (Lond). 7:152010. View Article : Google Scholar

73 

Ley RE, Turnbaugh PJ, Klein S and Gordon JI: Microbial ecology: Human gut microbes associated with obesity. Nature. 444:1022–1023. 2006. View Article : Google Scholar : PubMed/NCBI

74 

Janssen AWF, Houben T, Katiraei S, Dijk W, Boutens L, van der Bolt N, Wang Z, Brown JM, Hazen SL, Mandard S, et al: Modulation of the gut microbiota impacts nonalcoholic fatty liver disease: A potential role for bile acids. J Lipid Res. 58:1399–1416. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Le Roy T, Llopis M, Lepage P, Bruneau A, Rabot S, Bevilacqua C, Martin P, Philippe C, Walker F, Bado A, et al: Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice. Gut. 62:1787–1794. 2013. View Article : Google Scholar

76 

Hu H, Lin A, Kong M, Yao X, Yin M, Xia H, Ma J and Liu H: Intestinal microbiome and NAFLD: Molecular insights and therapeutic perspectives. World J Gastroenterol. 20:142–158. 2020. View Article : Google Scholar

77 

Rennert C, Heil T, Schicht G, Stilkerich A, Seidemann L, Kegel-Hübner V, Seehofer D and Damm G: Prolonged lipid accumulation in cultured primary human hepatocytes rather leads to ER stress than oxidative stress. Int J Mol Sci. 21:70972020. View Article : Google Scholar : PubMed/NCBI

78 

Wesolowski SR, Kasmi KC, Jonscher KR and Friedman JE: Developmental origins of NAFLD: A womb with a clue. Nat Rev Gastroenterol Hepatol. 14:81–96. 2017. View Article : Google Scholar :

79 

Arab JP, Karpen SJ, Dawson PA, Arrese M and Trauner M: Bile acids and nonalcoholic fatty liver disease: Molecular insights and therapeutic perspectives. Hepatology. 65:350–362. 2017. View Article : Google Scholar

80 

Jiang C, Xie C, Li F, Zhang L, Nichols RG, Krausz KW, Cai J, Qi Y, Fang ZZ, Takahashi S, et al: Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J Clin Invest. 125:386–402. 2015. View Article : Google Scholar :

81 

Thomas C, Gioiello A, Noriega L, Strehle A, Oury J, Rizzo G, Macchiarulo A, Yamamoto H, Mataki C, Pruzanski M, et al: TGR5 mediated bile acid sensing controls glucose homeostasis. Cell Metab. 10:167–177. 2009. View Article : Google Scholar : PubMed/NCBI

82 

Zhou T, Zheng Y, Liang H and Tang J: Progress of bile acid metabolism in chronic liver disease. J Integr Tradit Chin West Med Hepatol. 30:173–175. 2020.In Chinese.

83 

Watanabe M, Houten SM, Mataki C, Christoffolete MA, Kim BW, Sato H, Messaddeq N, Harney JW, Ezaki O, Kodama T, et al: Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature. 439:484–489. 2006. View Article : Google Scholar : PubMed/NCBI

84 

Wang F, Wang YY, Zhang D, Zhao XR and Liu K: Research on curative effects of regulating bile acid metabolism and intestinal flora in treatment of nonalcoholic fatty liver disease. China Med Pharm. 9:36–40. 452019.In Chinese.

85 

Jiang LY and Xiao XH: Research progress on correlation between gut microbiota and non-alcoholic fatty liver disease. J Clin Pathol Res. 36:2060–2065. 2016.

86 

Carr RM and Reid AE: FXR agonists as therapeutic agents for non-alcoholic fatty liver disease. Curr Atheroscler Rep. 17:5002015. View Article : Google Scholar : PubMed/NCBI

87 

Liu K, Xu SY, Xu B, Chen D and Li N: The protective role of early autophagy in nonalcoholic fatty liver. Beijing Med J. 33:947–950. 2011.In Chinese.

88 

Wang XL, Xiong JR, Zhao QM, Jiang W and Li P: The dual role of autophagy in disease. Prog Mod Biomed. 14:4997–5000. 2014.In Chinese.

89 

Yang L, Li P, Fu S, Calay ES and Hotamisligil GS: Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance. Cell Metab. 11:467–478. 2010. View Article : Google Scholar : PubMed/NCBI

90 

Ma DW, Ha J, Yoon KS, Kang I, Choi TG and Kim SS: Innate immune system in the pathogenesis of non-alcoholic fatty liver disease. Nutrients. 15:20682023. View Article : Google Scholar : PubMed/NCBI

91 

Kazankov K, Jørgensen SMD, Thomsen KL, Møller HJ, Vilstrup H, George J, Schuppan D and Grønbæk H: The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Nat Rev Gastroenterol Hepatol. 16:145–159. 2019. View Article : Google Scholar

92 

Tilg H, Effenberger M and Adolph TE: A role for il-1 inhibitors in the treatment of non-alcoholic fatty liver disease (NAFLD)? Expert Opin Investig Drugs. 29:1032020. View Article : Google Scholar

93 

Hayden MS and Ghosh S: NF-κB, the first quarter-century: Remarkable progress and outstanding questions. Genes Dev. 26:203–234. 2012. View Article : Google Scholar : PubMed/NCBI

94 

Garcia-Ruiz C, Mato JM, Vance D, Kaplowitz N and Fernández-Checa JC: Acid sphingomyelinase-ceramide system in steatohepatitis: A novel target regulating multiple pathways. J Hepatol. 62:2192015. View Article : Google Scholar

95 

Fantuzzi G: Adipose tissue, adipokines, and inflammation. J Allergy Clin Immunol. 115:911–919; quiz 920. 2005. View Article : Google Scholar : PubMed/NCBI

96 

Tong Q, Wang XL, Li SB, Yang GL, Jin S, Gao ZY and Liu XB: Combined detection of il-6 and il-8 is beneficial to the diagnosis of early-stage esophageal squamous cell cancer: A preliminary study based on the screening of serum markers using protein chips. OncoTargets Ther. 11:5777–5787. 2018. View Article : Google Scholar

97 

Tikhomirova AS, Kislyakov VA, Baykova IE and Nikitin IG: Clinical-morphological parallels of the PNPLA3 gene polymorphism in patients with nonalcoholic fatty liver disease. Ter Arkh. 90:85–88. 2018.

98 

Dai G, Liu P, Li X, Zhou X and He S: Association between PNPLA3 rs738409 polymorphism and nonalcoholic fatty liver disease (NAFLD) susceptibility and severity: a meta-analysis. Medicine (Baltimore). 98:e143242019. View Article : Google Scholar : PubMed/NCBI

99 

Dongiovanni P, Petta S, Maglio C, Fracanzani AL, Pipitone R, Mozzi E, Motta BM, Kaminska D, Rametta R, Grimaudo S, et al: Transmembrane 6 superfamily member 2 gene variant disentangles nonalcoholic steatohepatitis from cardiovascular disease. Hepatology. 61:506–514. 2015. View Article : Google Scholar

100 

Wu S, Zheng B, Liu T, Zhu Z, Gu W and Liu Q: 17beta-hydroxysteroid dehydrogenase 3 deficiency due to novel compound heterozygous variants of HSD17B3 gene in a sib pair. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 38:787–790. 2021.In Chinese. PubMed/NCBI

101 

Dai LL, Li SD, Ma YC, Tang JR, Lv JY, Zhang YQ, Miao YL, Ma YQ, Li CM, Chu YY, et al: MicroRNA-30b regulates insulin sensitivity by targeting SERCA2b in non-alcoholic fatty liver disease. Liver Int. 39:1504–1513. 2019. View Article : Google Scholar : PubMed/NCBI

102 

Yu J, Peng J, Luan Z, Zheng F and Su W: MicroRNAs as a novel tool in the diagnosis of liver lipid dysregulation and fatty liver disease. Molecules. 24:2302019. View Article : Google Scholar : PubMed/NCBI

103 

Dongiovanni P, Meroni M, Longo M, Fargion S and Fracanzani AL: miRNA signature in NAFLD: A turning point for a non-invasive diagnosis. Int J Mol Sci. 19:39662018. View Article : Google Scholar : PubMed/NCBI

104 

Zhang M, Sun W, Zhou M and Tang Y: MicroRNA-27a regulates hepatic lipid metabolism and alleviates NAFLD via repressing FAS and SCD1. Sci Rep. 7:144932017. View Article : Google Scholar : PubMed/NCBI

105 

Szabo G and Csak T: Role of microRNAs in NAFLD/NASH. Dig Dis Sci. 61:1314–1324. 2016. View Article : Google Scholar : PubMed/NCBI

106 

López-Riera M, Conde I, Quintas G, Pedrola L, Zaragoza A, Perez-Rojas J, Salcedo M, Benlloch S, Castell JV and Jover R: Non-invasive prediction of NAFLD severity: A comprehensive, independent validation of previously postulated serum microRNA biomarkers. Sci Rep. 8:106062018. View Article : Google Scholar : PubMed/NCBI

107 

Guo Y, Xiong Y, Sheng Q, Zhao S, Wattacheril J and Flynn CR: A micro-RNA expression signature for human NAFLD progression. J Gastroenterol. 51:1022–1030. 2016. View Article : Google Scholar : PubMed/NCBI

108 

Luo ZL, Tang LJ, Wang T, Dai RW, Ren JD, Cheng L, Xiang K and Tian FZ: Effects of treatment with hydrogen sulfide on methionine-choline deficient diet-induced non-alcoholic steatohepatitis in rats. J Gastroenterol Hepatol. 29:215–222. 2014. View Article : Google Scholar

109 

Wu D, Zheng N, Qi K, Cheng H, Sun Z, Gao B, Zhang Y, Pang W, Huangfu C, Ji S, et al: Exogenous hydrogen sulfide mitigates the fatty liver in obese mice through improving lipid metabolism and antioxidant potential. Med Gas Res. 5:12015. View Article : Google Scholar : PubMed/NCBI

110 

Salehi-Sahlabadi A, Sadat S, Beigrezaei S, Pourmasomi M, Feizi A, Ghiasvand R, Hadi A, Clark CCT and Miraghajani M: Dietary patterns and risk of non-alcoholic fatty liver disease. BMC Gastroenterol. 21:412021. View Article : Google Scholar : PubMed/NCBI

111 

Liu H and Ye H: Research progress on the relationship between sleep, circadian rhythm, intestinal flora and non-alcoholic fatty liver disease. Mod Pract Med. 31:565–568. 2019.In Chinese.

112 

Wei YT, Lee PY, Lin CY, Chen HJ, Lin CC, Wu JS, Chang YF, Wu CL and Guo HR: Non-alcoholic fatty liver disease among patients with sleep disorders: A nationwide study of Taiwan. BMC Gastroenterol. 20:322020. View Article : Google Scholar : PubMed/NCBI

113 

Marin-Alejandre BA, Abete I, Cantero I, Riezu-Boj JI, Milagro FI, Monreal JI, Elorz M, Herrero JI, Benito-Boillos A, Quiroga J, et al: Association between sleep disturbances and liver status in obese subjects with nonalcoholic fatty liver disease: A comparison with healthy controls. Nutrients. 11:3222019. View Article : Google Scholar : PubMed/NCBI

114 

Ai Y, Yang X, Pan X and Ye J: Role of circadian clock genes in the development and progression of nonalcoholic fatty liver disease. J Clin Hepatol. 35:2327–2330. 2019.In Chinese.

115 

Reinke H and Asher G: Circadian clock control of liver metabolic functions. Gastroenterology. 150:573–580. 2016. View Article : Google Scholar

116 

Wen Y: Relationship between social mental factors and non-alcoholic fatty liver disease: A case-control study. China J Health Psychol. 26:565–568. 2018.In Chinese.

117 

Cho IY, Chang Y, Sung E, Kang JH, Wild SH, Byrne CD, Shin H and Ryu S: Depression and increased risk of non-alcoholic fatty liver disease in individuals with obesity. Epidemiol Psychiatr Sci. 30:e232021. View Article : Google Scholar : PubMed/NCBI

118 

Youssef NA, Abdelmalek MF, Binks M, Guy CD, Omenetti A, Smith AD, Diehl AM and Suzuki A: Associations of depression, anxiety and antidepressants with histological severity of nonalcoholic fatty liver disease. Liver Int. 33:1062–1070. 2013. View Article : Google Scholar : PubMed/NCBI

119 

Peluso I, Manafikhi H, Reggi R and Palmery M: Effects of red wine on postprandial stress: Potential implication in non-alcoholic fatty liver disease development. Eur J Clin Nutr. 54:497–507. 2015. View Article : Google Scholar

120 

Yu D, Shu XO, Xiang YB, Li H, Yang G, Gao YT, Zheng W and Zhang X: Higher dietary choline intake is associated with lower risk of nonalcoholic fatty liver in normal-weight Chinese women. J Nutr. 144:2034–2040. 2014. View Article : Google Scholar : PubMed/NCBI

121 

Kado A, Inoue Y, Moriya M, Tsutsumi K, Ikeuchi T, Okushin K, Yotsuyanagi K, Koike H and Fujishiro M: Triglyceride level and soft drink consumption predict nonalcoholic fatty liver disease in nonobese male adolescents. Hepatol Res. 53:497–510. 2023. View Article : Google Scholar : PubMed/NCBI

122 

Khazaei Y, Dehghanseresht N, Ebrahimi Mousavi S, Nazari M, Salamat S, Asbaghi O and Mansoori A: Association between protein intake from different animal and plant origins and the risk of non-alcoholic fatty liver disease: A case-control study. Clin Nutr Res. 12:29–39. 2023. View Article : Google Scholar : PubMed/NCBI

123 

Lanaspa MA, Kuwabara M, Andres-Hernando A, Li N, Cicerchi C, Jensen T, Orlicky DJ, Roncal-Jimenez CA, Ishimoto T, Nakagawa T, et al: High salt intake causes leptin resistance and obesity in mice by stimulating endogenous fructose production and metabolism. Proc Natl Acad Sci USA. 115:3138–3143. 2018. View Article : Google Scholar : PubMed/NCBI

124 

Liang YF, Kuang HY, Qin W, Zhu WB, Liu NJ, Zuo J and Han S: Correlation between salt intake and non-alcoholic fatty liver disease in type 2 diabetes mellitus patients in northern China. Chin J Mult Organ Dis Elderly. 18:765–768. 2019.In Chinese.

125 

Chen S, Teoh NC, Chitturi S and Farrell GC: Coffee and non-alcoholic fatty liver disease: Brewing evidence for hepatoprotection? J Gastroenterol Hepatol. 29:435–441. 2014. View Article : Google Scholar

126 

Sinha RA, Farah BL, Singh BK, Siddique MM, Li Y, Wu YJ, Ilkayeva OR, Gooding J, Ching J, Zhou J, et al: Caffeine stimulates hepatic lipid metabolism by the autophagy-lysosomal pathway in mice. Hepatology. 59:1366–1380. 2014. View Article : Google Scholar

127 

Chen B, Sun L, Zeng G, Shen Z, Wang K, Yin L, Xu F, Wang P, Ding Y, Nie Q, et al: Gut bacteria alleviate smoking-related NASH by degrading gut nicotine. Nature. 610:562–568. 2022. View Article : Google Scholar : PubMed/NCBI

128 

Jung HS, Chang Y, Kwon MJ, Sung E, Yun KE, Cho YK, Shin H and Ryu S: Smoking and the risk of non-alcoholic fatty liver disease: A cohort study. Am J Gastroenterol. 114:453–463. 2019. View Article : Google Scholar

129 

Ma H, Zhao J and Xu J: The correlation between heavy metal ions in blood and metabolic dysfunction-associated steatotic liver disease from 1999 to 2018 based on NHANES data. Front Public Health. 12:15129012025. View Article : Google Scholar : PubMed/NCBI

130 

Salman M, Kamel MA, El-Nabi SEH, Ismail AHA, Ullah S, Al-Ghamdi A, Hathout HMR and El-Garawani IM: The regulation of HBP1, SIRT1, and SREBP-1c genes and the related microRNAs in non-alcoholic fatty liver rats: The association with the folic acid anti-steatosis. PLoS One. 17:e02654552022. View Article : Google Scholar : PubMed/NCBI

131 

Zhou Q, Xu W, Wang L and Duan Y: Polychlorinated biphenyl 118 induced nonalcoholic fatty liver disease in rats. J Nanjing Med Univ (Nat Sci). 39:659–663. 2019.In Chinese.

132 

Stephen Robert JM, Peddha MS and Srivastava AK: Effect of silymarin and quercetin in a miniaturized scaffold in wistar rats against non-alcoholic fatty liver disease. ACS Omega. 6:20735–20745. 2021. View Article : Google Scholar : PubMed/NCBI

133 

Zhang M, Yuan Y, Wang Q, Li X, Men J and Lin M: The Chinese medicine Chai Hu Li Zhong Tang protects against non-alcoholic fatty liver disease by activating AMPKα. Biosci Rep. 38:BSR201806442018. View Article : Google Scholar

134 

Lin M, Deng G, Ou Y, Chen Z, Huang J, Kang M and Li J: Establishment of non-alcoholic fatty liver disease model in young rats induced by high-fat diet. J Med Theor Pract. 32:2321–2323. 23262019.In Chinese.

135 

Jin Y, Xing W, Lyu A, Zhang J, Yang W, Xu Y, Wang Q and Li J: Comparison of CYP enzyme activities in rat models of nonalcoholic fatty liver established by different methods. Chin J Comp Med. 28:75–83. 2018.In Chinese.

136 

Chen XQ, Xu LS, Li DF and Deng WP: Effects of glucagon-like peptide-1 analog on CDAA diet-induced models of nonalcoholic fatty liver fibrosis in rats. J Evid-Based Med. 17:163–170. 2017.In Chinese.

137 

Yan D, Li SY and Wei YY: Study on establishment of animal model with diabetes complicated fatty liver induced by streptozocin and nicotinamide. Chin J Clin Pharmacol. 31:1846–1848. 2015.In Chinese.

138 

Li X and Li Y: Discussion on nonalcoholic fatty liver disease animal model. Chin Community Doctors. 33:5–8. 2017.In Chinese.

139 

Yong HY, Larrouy-Maumus G, Zloh M, Smyth R, Ataya R, Benton CM and Munday MR: Early detection of metabolic changes in drug-induced steatosis using metabolomics approache. RSC Adv. 10:41047–41057. 2020. View Article : Google Scholar : PubMed/NCBI

140 

Che X and Han J: Effect of FGF21 on TLR4/p38MAPK signaling pathway in nonalcoholic fatty liver diseases of rats. J Med Res. 47:130–135. 2018.In Chinese.

141 

Hakkak R, Spray B, Børsheim E and Korourian S: Diet containing soy protein concentrate with low and high isoflavones for 9 weeks protects against non-alcoholic fatty liver steatosis using obese zucker rats. Front Nutr. 9:9135712022. View Article : Google Scholar : PubMed/NCBI

142 

Matsumoto Y, Fujita S, Yamagishi A, Shirai T, Maeda Y, Suzuki T, Kobayashi KI, Inoue J and Yamamoto Y: Brown rice inhibits development of nonalcoholic fatty liver disease in obese zucker (fa/fa) rats by increasing lipid oxidation via activation of retinoic acid synthesis. J Nutr. 151:2705–2713. 2021. View Article : Google Scholar : PubMed/NCBI

143 

Xia QS, Gao Y, Wen-Bin W, Wu F, Dong H, Xu LJ, Fang K, Hu ML, Yuan F, Lu FE and Gong J: Ban-xia-xie-xin-tang ameliorates hepatic steatosis by regulating cidea and cidec expression in HFD-fed mice. Phytomedicine. 105:1543512022. View Article : Google Scholar : PubMed/NCBI

144 

Shi Y, Laba PC, Zhang DY, Weng SQ, Liu XY and Wang HQ: Effects of bifidobacteria on high-fat diet-induced non-alcoholic fatty liver disease in C57BL/6 mice. Chin J Clin Med. 29:473–480. 2022.In Chinese.

145 

Yu H, Yi X, Gao X, Ji J, Liu Z, Xia G, Li C, Zhang X and Shen X: Tilapia-head chondroitin sulfate protects against nonalcoholic fatty liver disease via modulating the gut-liver axis in High-Fat-Diet-Fed C57BL/6 mice. Foods. 11:9222022. View Article : Google Scholar : PubMed/NCBI

146 

De Minicis S, Agostinelli L, Rychlicki C, Sorice GP, Saccomanno S, Candelaresi C, Giaccari A, Trozzi L, Pierantonelli I, Mingarelli E, et al: HCC development is associated to peripheral insulin resistance in a mouse model of NASH. PLoS One. 9:e00971362014. View Article : Google Scholar

147 

Li XC, Zhang XY, Xu S, Li H and Zhang YH: Establishment of a non-alcoholic fatty liver model in C57BL/6J mice. In: Proceedings of the Symposium on Veterinary Toxicology and Feed Toxicology and the 4th National Congress of the Veterinary Toxicology Committee of the Chinese Society of Toxicology; Beijing. pp. 94–95. 2013, In Chinese.

148 

Feng Z, Pang L, Chen S, Wei J, Huang Q, Wang Y and Lin X: The protective effect of Melissa officinalis on dexamethasone-induced nonalcoholic fatty liver in C57BL/6J mice. J Chin Med Mater. 44:961–965. 2021.In Chinese.

149 

Asgharpour A, Cazanave SC, Pacana T, Seneshaw M, Vincent R, Banini BA, Kumar DP, Daita K, Min HK, Mirshahi F, et al: A diet-induced animal model of non-alcoholic fatty liver disease and hepatocellular cancer. J Hepatol. 65:579–588. 2016. View Article : Google Scholar : PubMed/NCBI

150 

Ma H, Zhu G, Zhang X, Yang R, Xie Y, Liu X and Wang B: An establishment method of a mouse model of non-alcoholic fatty liver disease. J Anhui Agric Sci. 49:80–84. 2021.In Chinese.

151 

Jahn D, Dorbath D, Kircher S, Nier A, Bergheim I, Lenaerts K, Hermanns HM and Geier A: Beneficial effects of vitamin d treatment in an obese mouse model of non-alcoholic steatohepatitis. Nutrients. 11:772019. View Article : Google Scholar : PubMed/NCBI

152 

Wang JJ, Fang HL, Li CW, Chen FC, Tang WJ, Jiang XY, Wu HY and Chen DL: Establishment of non-alcoholic fatty liver model in mice. J Clin Rehabilit Tissue Eng Res. 15:4395–4399. 2011.In Chinese.

153 

Liu T, Tan F, Long X, Pan Y, Mu J, Zhou X, Yi R and Zhao X: Improvement effect of lotus leaf flavonoids on carbon tetrachloride-induced liver injury in mice. Biomedicines. 8:412020. View Article : Google Scholar : PubMed/NCBI

154 

Malloy VL, Perrone CE, Mattocks DA, Ables GP, Caliendo NS, Orentreich DS and Orentreich N: Methionine restriction prevents the progression of hepatic steatosis in leptin-deficient obese mice. Metabolism. 62:1651–1661. 2013. View Article : Google Scholar : PubMed/NCBI

155 

Son YJ, Jung DS, Shin JM, Kim M, Yoo G and Nho CW: Yellow loosestrife (lysimachia vulgaris var. davurica) ameliorates liver fibrosis in db/db mice with methionine and choline-deficient diet-induced nonalcoholic steatohepatitis. BMC Complement Med and Ther. 21:442021. View Article : Google Scholar

156 

Yu W, Zhang J, Liu Y, Liu F, Shao H and Han G: Improvement effect of mussel polysaccharide α-d-glucan on non-alcoholic fatty liver apolipoprotein E knockout mice induced by high fat diet. Sci Technol Food Ind. 43:369–376. 2022.In Chinese.

157 

Song L, Tang SQ, Tong JW, Cai P, Liu JX, Ren XJ and Huang W: Study on the mechanism of Xuefu Zhuyu decoction in preventing and treating NAFLD. Chin J Integr Tradit West Med. 40:1103–1106. 2020.In Chinese.

158 

Yang P, Wang Y, Tang W, Sun W, Ma Y, Lin S, Jing J, Jiang L, Shi H, Song Z and Yu L: Western diet induces severe nonalcoholic steatohepatitis, ductular reaction, and hepatic fibrosis in liver CGI-58 knockout mice. Sci Rep. 10:47012020. View Article : Google Scholar : PubMed/NCBI

159 

Piguet AC, Saran U, Simillion C, Keller I, Terracciano L, Reeves HL and Dufour JF: Regular exercise decreases liver tumors development in hepatocyte-specific PTEN-deficient mice independently of steatosis. J Hepatol. 62:1296–1303. 2015. View Article : Google Scholar : PubMed/NCBI

160 

Semba T, Nishimura M, Nishimura S, Ohara O, Ishige T, Ohno S, Nonaka K, Sogawa K, Satoh M, Sawai S, et al: The FLS (fatty liver Shionogi) mouse reveals local expressions of lipocalin-2, CXCL1 and CXCL9 in the liver with non-alcoholic steatohepatitis. BMC Gastroenterol. 13:1202013. View Article : Google Scholar : PubMed/NCBI

161 

Li JZ, Huang Y, Karaman R, Ivanova PT, Brown HA, Roddy T, Castro-Perez J, Cohen JC and Hobbs HH: Chronic overexpression of PNPLA3I148M in mouse liver causes hepatic steatosis. J Clin Invest. 122:4130–4144. 2012. View Article : Google Scholar : PubMed/NCBI

162 

Yin HQ, Kim M, Kim JH, Kong G, Lee MO, Kang KS, Yoon BI, Kim HL and Lee BH: Hepatic gene expression profiling and lipid homeostasis in mice exposed to steatogenic drug, tetracycline. Toxicol Sci. 94:206–216. 2006. View Article : Google Scholar : PubMed/NCBI

163 

Li Z, Jin H, Oh SY and Ji GE: Anti-obese effects of two lactobacilli and two bifidobacteria on ICR mice fed on a high fat diet. Biochem Biophys Res Commun. 480:222–227. 2016. View Article : Google Scholar : PubMed/NCBI

164 

Yu Z, He Y, Luo W and Li L: Establishment of AKT gene-mediated non-alcoholic fatty liver models in mice. Acta Med Univ Sci Technol Huazhong. 45:170–175. 2016.In Chinese.

165 

López-Lemus UA, Garza-Guajardo R, Barboza-Quintana O, Rodríguez-Hernandez A, García-Rivera A, Madrigal-Pérez VM, Guzmán-Esquivel J, García-Labastida LE, Soriano-Hernández AD, Martínez-Fierro ML, et al: Association between nonalcoholic fatty liver disease and severe male reproductive organ impairment (germinal epithelial loss): study on a mouse model and on human patients. Am J Mens Health. 12:639–648. 2018. View Article : Google Scholar : PubMed/NCBI

166 

Musolino V, Gliozzi M, Scarano F, Bosco F, Scicchitano M, Nucera S, Carresi C, Ruga S, Zito MC, Maiuolo J, et al: Bergamot polyphenols improve dyslipidemia and pathophysiological features in a mouse model of non-alcoholic fatty liver disease. Sci Rep. 10:25652020. View Article : Google Scholar : PubMed/NCBI

167 

Chow JD, Jones ME, Prelle K, Simpson ER and Boon WC: A selective estrogen receptor α agonist ameliorates hepatic steatosis in the male aromatase knockout mouse. J Endocrinol. 210:323–334. 2011. View Article : Google Scholar : PubMed/NCBI

168 

Komatsu G, Nonomura T, Sasaki M, Ishida Y, Arai S and Miyazaki T: AIM-deficient mouse fed a high-trans-fat, high-cholesterol diet: A new animal model for nonalcoholic fatty liver disease. Exp Anim. 68:147–158. 2019. View Article : Google Scholar

169 

Nguyen TTP, Kim DY, Lee YG, Lee YS, Truong XT, Lee JH, Song DK, Kwon TK, Park SH, Jung CH, et al: SREBP-1c impairs ULK1 sulfhydration-mediated autophagic flux to promote hepatic steatosis in high-fat-diet-fed mice. Mol Cell. 81:3820–3832.e7. 2021. View Article : Google Scholar : PubMed/NCBI

170 

Deguise MO, Pileggi C, De Repentigny Y, Beauvais A, Tierney A, Chehade L, Michaud J, Llavero-Hurtado M, Lamont D, Atrih A, et al: SMN depleted mice offer a robust and rapid onset model of nonalcoholic fatty liver disease. Cell Mol Gastroenterol Hepatol. 12:354–377.e3. 2021. View Article : Google Scholar : PubMed/NCBI

171 

Cano A, Buque X, Martínez-Uña M, Aurrekoetxea I, Menor A, García-Rodríguez JL, Lu SC, Martínez-Chantar ML, Mato JM, Ochoa B and Aspichueta P: Methionine adenosyltransferase 1A gene deletion disrupts hepatic very low-density lipoprotein assembly in mice. Hepatology. 54:1975–1986. 2011. View Article : Google Scholar : PubMed/NCBI

172 

Heintz MM, Kumar R, Rutledge MM and Baldwin WS: Cyp2b-nullmale mice are susceptible to diet-induced obesity and perturbations in lipid homeostasis. J Nutr Biochem. 70:125–137. 2019. View Article : Google Scholar : PubMed/NCBI

173 

Edmunds LR, Xie B, Mills AM, Huckestein BR, Undamatla R, Murali A, Pangburn MM, Martin J, Sipula I, Kaufman BA, et al: Liver-specific Prkn knockout mice are more susceptible to diet-induced hepatic steatosis and insulin resistance. Mol Metab. 41:1010512020. View Article : Google Scholar : PubMed/NCBI

174 

Yang W, Yan X, Chen R, Xin X, Ge S, Zhao Y, Yan X and Zhang J: Smad4 deficiency in hepatocytes attenuates NAFLD progression via inhibition of lipogenesis and macrophage polarization. Cell Death Dis. 16:582025. View Article : Google Scholar : PubMed/NCBI

175 

Yang YQ, Feng J, Bai X, Shen ZM, Liu XW, Gao J, Xie JY, Hu JH and Gao C: Establishment of a Microtus fortis model of non-alcoholic fatty liver. Acta Lab Anim Sci Sin. 21:34–38. 2013.In Chinese.

176 

Yu Z, Wang H, Gao G, Ke L, Zhou J, Rao P and Yu C: Effects of freshwater clam soup nanoparticles on non-alcoholic fatty liver disease of Meriones unguieulataus. J Chin Inst Food Sci Technol. 21:116–120. 2021.In Chinese.

177 

Bhathena J, Kulamarva A, Martoni C, Urbanska AM, Malhotra M, Paul A and Prakash S: Diet-induced metabolic hamster model of nonalcoholic fatty liver disease. Diabetes Metab Syndr Obes. 4:195–203. 2011.PubMed/NCBI

178 

Cui CX, Deng JN, Li Y, Liu YY, Fan JY, Mu HN, Sun HY, Wang YH and Han JY: Silibinin capsules improves high fat diet-induced nonalcoholic fatty liver disease in hamsters through modifying hepatic de novo lipogenesis and fatty acid oxidation. J Ethnopharmacol. 208:24–35. 2017. View Article : Google Scholar : PubMed/NCBI

179 

Zhao CZ, Jiang W, Zhu YY, Wang CZ, Zhong WH, Wu G, Chen J, Zhu MN, Wu QL, Du XL, et al: Highland barley monascus purpureus went extract ameliorates high-fat, high-fructose, high-cholesterol diet induced nonalcoholic fatty liver disease by regulating lipid metabolism in golden hamsters. J Ethnopharmacol. 286:1149222022. View Article : Google Scholar

180 

Skat-Rørdam J, Pedersen K, Skovsted GF, Gregersen I, Vangsgaard S, Ipsen DH, Latta M, Lykkesfeldt J and Tveden-Nyborg P: Vitamin C deficiency may delay diet-induced NASH regression in the guinea pig. Antioxidants (Basel). 11:692021. View Article : Google Scholar

181 

Jin M, Zheng L, Wei Y, Cheng J, Zhang D, Yan S, Qin H, Wang Q, Ci X and Feng H: Enterobacter cloacae aggravates metabolic disease by inducing inflammation and lipid accumulation. Environ Toxicol Pharmacol. 90:1038192022. View Article : Google Scholar : PubMed/NCBI

182 

Ipsen DH, SkatRørdam J, Tsamouri MM, Latta M, Lykkesfeldt J and TvedenNyborg P: Molecular drivers of non-alcoholic steatohepatitis are sustained in mild-to-late fibrosis progression in a guinea pig model. Mol Genet Genomics. 294:649–661. 2019. View Article : Google Scholar : PubMed/NCBI

183 

Pedersen K, Ipsen DH, Skat-Rrdam J, Lykkesfeldt J and Tveden-Nyborg P: Dietary long-chain fatty acids accelerate metabolic dysfunction in guinea pigs with non-alcoholic steatohepatitis. Nutrients. 15:24452023. View Article : Google Scholar : PubMed/NCBI

184 

Wang Y, Zhang P, Su X, Yu Q, Chen Y, Guan H, Liu E and Fan J: Establishment of a novel nonalcoholic fatty liver disease model using cholesterol-fed rabbits with reference to the potential role of endoplasmic reticulum stress. Mol Med Rep. 18:2898–2904. 2018.PubMed/NCBI

185 

Sturzeneker MCS, de Noronha L, Olandoski M, Wendling LU and Precoma DB: Ramipril significantly attenuates the development of non-alcoholic steatohepatitis in hyperlipidaemic rabbits. Am J Cardiovasc Dis. 9:8–17. 2019.PubMed/NCBI

186 

Nguyen TN, Podkowa AS, Tam AY, Arnold EC, Miller RJ, Park TH, Do MN and Oelze ML: Characterizing fatty liver in vivo in rabbits, using quantitative ultrasound. Ultrasound Med Biol. 45:2049–2062. 2019. View Article : Google Scholar : PubMed/NCBI

187 

Zheng W, Liu YL, Shang HB, Zhang Y, Ma DW, Hou N, Wang J, Sun XT, Peng Y, Pan L, et al: Characterization of spontaneously-developed non-alcoholic fatty liver disease in aged rhesus monkeys. Diabetol Metab Syndr. 10:682018. View Article : Google Scholar : PubMed/NCBI

188 

Cydylo MA, Davis AT and Kavanagh K: Fatty liver promotes fibrosis in monkeys consuming high fructose. Obesity (Silver Spring). 25:290–293. 2017. View Article : Google Scholar : PubMed/NCBI

189 

Kramer JA, Grindley J, Crowell AM, Makaron L, Kohli R, Kirby M, Mansfield KG and Wachtman LM: The common marmoset as a model for the study of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Vet Pathol. 52:404–413. 2015. View Article : Google Scholar

190 

Hamid H, Zhang JY, Li WX, Liu C, Li ML, Zhao LH, Ji C and Ma QG: Interactions between the cecal microbiota and non-alcoholic steatohepatitis using laying hens as the model. Poult Sci. 98:2509–2521. 2019. View Article : Google Scholar : PubMed/NCBI

191 

Zhu L, Liao R, Huang J, Yan H, Xiao C, Yang Y, Wang H and Yang C: The miR-216/miR-217 Cluster regulates lipid metabolism in laying hens with fatty liver syndrome via PPAR/SREBP signaling pathway. Front Vet Sci. 9:9138412022. View Article : Google Scholar : PubMed/NCBI

192 

Tian WH, Wang Z, Yue YX, Li H, Li ZJ, Han RL, Tian YD, Kang XT and Liu XJ: Mir-34a-5p increases hepatic triglycerides and total cholesterol levels by regulating acsl1 protein expression in laying hens. Int J Mol Sci. 20:44202019. View Article : Google Scholar : PubMed/NCBI

193 

Song J, Shi X, Li X, Liang Q, Zeng L, Li G, Yan Y, Xu G and Zheng J: Associations of the T329S polymorphism in flavin-containing monooxygenase 3 with atherosclerosis and fatty liver syndrome in 90-week-old hens. Front Vet Sci. 9:8686022022. View Article : Google Scholar : PubMed/NCBI

194 

Delgado ME, Cárdenas BI, Farran N and Fernandez M: Metabolic reprogramming of liver fibrosis. Cells. 10:36042021. View Article : Google Scholar : PubMed/NCBI

195 

Mazza G, Al-Akkad W and Rombouts K: Engineering in vitro models of hepatofibrogenesis. Adv Drug Deliv Rev. 121:147–157. 2017. View Article : Google Scholar : PubMed/NCBI

196 

Soret PA, Magusto J, Housset C and Gautheron J: In vitro and in vivo models of non-alcoholic fatty liver disease: A critical appraisal. J Clin Med. 10:362020. View Article : Google Scholar : PubMed/NCBI

197 

Zhong Y, Yu JS, Wang X, Binas B and Yoo HH: Chemical-based primary human hepatocyte monolayer culture for the study of drug metabolism and hepato-toxicity: comparison with the spheroid model. FASEB J. 35:e213792021. View Article : Google Scholar

198 

Kanasaki A, Iida T, Murao K, Shirouchi B and Sato M: D-Allulose enhances uptake of HDL-cholesterol into rat's primary hepatocyte via SR-B1. Cytotechnology. 72:295–301. 2020. View Article : Google Scholar : PubMed/NCBI

199 

Ng IC, Zhang L, Shen NNYY, Soong YT, Ng CW, Koh PKS, Zhou Y and Yu H: Isolation of primary rat hepatocytes with multiparameter perfusion control. J Vis Exp. e622892021.

200 

Xie Y, Yao J, Jin W, Ren L and Li X: Induction and maturation of hepatocyte-like cells in vitro: Focus on technological advances and challenges. Front Cell Dev Biol. 9:7659802021. View Article : Google Scholar : PubMed/NCBI

201 

Oseini AM, Cole BK, Issa D, Feaver RE and Sanyal AJ: Translating scientific discovery: The need for preclinical models of nonalcoholic steatohepatitis. Hepatol Int. 12:6–16. 2018. View Article : Google Scholar : PubMed/NCBI

202 

Zhao S, Rong C, Zhang S, Liu Y and Chen J: Protective effects of polysaccharides of Auricularia auricula polysaccharide on ethanol-induced liver injury. Jiangsu Agric Sci. 45:142–144. 2017.In Chinese.

203 

Yu QQ, Meng MR, Liu YY, Cheng YX and Chen Y: Preventive effect and underlying mechanism of dihydrocumin on NAFLD in oleic acid-treated HepG2 cells. Chin Tradit Herb Drugs. 49:1092–1099. 2018.In Chinese.

204 

Mann DA: Human induced pluripotent stem cell-derived hepatocytes for toxicology testing. Expert Opin Drug Metab Toxicol. 11:1–5. 2015. View Article : Google Scholar

205 

Chen Y and Ma K: NLRC4 inflammasome activation regulated by TNF-α promotes inflammatory responses in nonalcoholic fatty liver disease. Biochem Biophys Res Commun. 511:524–530. 2019. View Article : Google Scholar : PubMed/NCBI

206 

Giraudi PJ, Becerra VJB, Marina V, Chavez-Tapiaa NC, Tiribelli C and Rosso N: The importance of the interaction between hepatocyte and hepatic stellate cells in fibrogenesis induced by fatty accumulation. Exp Mol Pathol. 98:85–92. 2015. View Article : Google Scholar

207 

Feaver RE, Cole BK, Lawson MJ, Hoang SA, Marukian S, Blackman BR, Figler RA, Sanyal AJ, Wamhoff BR and Dash A: Development of an in vitro human liver system for interrogating nonalcoholic steatohepatitis. JCI Insight. 1:e909542016. View Article : Google Scholar : PubMed/NCBI

208 

Salerno S, Campana C, Morelli S, Drioli E and De Bartolo L: Human hepatocytes and endothelial cells in organotypic membrane systems. Biomaterials. 32:8848–8859. 2011. View Article : Google Scholar : PubMed/NCBI

209 

Ebrahimkhani MR, Neiman JA, Raredon MS, Hughes DJ and Griffith LG: Bioreactor technologies to support liver function in vitro. Adv Drug Deliv Rev. 69:132–157. 2014. View Article : Google Scholar : PubMed/NCBI

210 

Kamei KI, Yoshioka M, Terada S, Tokunaga Y and Chen Y: Three-dimensional cultured liver-on-a-Chip with mature hepatocyte-like cells derived from human pluripotent stem cells. Biomed Microdevices. 21:732019. View Article : Google Scholar : PubMed/NCBI

211 

Kostrzewski T, Maraver P, Ouro-Gnao L, Levi A, Snow S, Miedzik A, Rombouts K and Hughes D: A microphysiological system for studying nonalcoholic steatohepatitis. Hepatol Commun. 4:77–91. 2020. View Article : Google Scholar : PubMed/NCBI

212 

Kostrzewski T, Cornforth T, Snow SA, Ouro-Gnao L, Rowe C, Large EM and Hughes DJ: Three-dimensional perfused human in vitro model of non-alcoholic fatty liver disease. World J Gastroenterol. 23:204–215. 2017. View Article : Google Scholar : PubMed/NCBI

213 

Gaskell H, Sharma P, Colley HE, Murdoch C, Williams DP and Webb SD: Characterization of a functional C3A liver spheroid model. Toxicol Res (Camb). 5:1053–1065. 2016. View Article : Google Scholar : PubMed/NCBI

214 

Rose S, Ezan F, Cuvellier M, Bruyère A, Legagneux V, Langouët S and Baffet G: Generation of proliferating human adult hepatocytes using optimized 3D culture conditions. Sci Rep. 11:5152021. View Article : Google Scholar : PubMed/NCBI

215 

Vorrink SU, Zhou Y, Ingelman-Sundberg M and Lauschke VM: Prediction of drug-induced hepatotoxicity using long-term stable primary hepatic 3D spheroid cultures in chemically defined conditions. Toxicol Sci. 163:655–665. 2018. View Article : Google Scholar : PubMed/NCBI

216 

Messner S, Fredriksson L, Lauschke VM, Roessger K, Escher C, Bober M, Kelm JM, Ingelman-Sundberg M and Moritz W: Transcriptomic, proteomic, and functional long-term characterization of multicellular three-dimensional human liver microtissues. Appl In Vitro Toxicol. 4:1–12. 2018. View Article : Google Scholar : PubMed/NCBI

217 

Rubiano A, Indapurkar A, Yokosawa R, Miedzik A, Rosenzweig B, Arefin A, Moulin CM, Dame K, Hartman N, Volpe DA, et al: Characterizing the reproducibility in using a liver microphysiological system for assaying drug toxicity, metabolism, and accumulation. Clin Transl Sci. 14:1049–1061. 2021. View Article : Google Scholar : PubMed/NCBI

218 

Hu H, Gehart H, Artegiani B, López-Iglesias C, Dekkers F, Basak O, van Es J, Chuva de Sousa Lopes SM, Begthel H, Korving J, et al: Long-term expansion of functional mouse and human hepatocytes as 3D organoids. Cell. 175:1591–1606. e192018. View Article : Google Scholar : PubMed/NCBI

219 

Guan Y, Xu D, Garfin PM, Ehmer U, Hurwitz M, Enns G, Michie S, Wu M, Zheng M, Nishimura T, et al: Human hepatic organoids for the analysis of human genetic diseases. J Clin Invest Insight. 2:e949542017.

220 

Wang Y, Wang H, Deng P, Tao T, Liu H and Wu S, Chen W and Wu S: Modeling human nonalcoholic fatty liver disease (NAFLD) with an organoids-on-a-chip system. ACS Biomater Sci Eng. 6:5734–5743. 2020. View Article : Google Scholar : PubMed/NCBI

221 

Fernandes R, Luo X, Tsao CY, Payne GF, Ghodssi R, Rubloffde GW and Bentley WE: Biological nanofactories facilitate spatially selective capture and manipulation of quorum sensing bacteria in a bioMEMS device. Lab Chip. 10:1128–1134. 2010. View Article : Google Scholar : PubMed/NCBI

222 

Kang D, Hong G, An S, Jang I, Yun WS, Shim JH and Jin S: Bioprinting of multiscaled hepatic lobules within a highly vascularized construct. Small. 16:e19055052020. View Article : Google Scholar : PubMed/NCBI

223 

Mazzocchi A, Devarasetty M, Huntwork R, Soker S and Skardal A: Optimization of collagen type I-hyaluronan hybrid bioink for 3D bioprinted liver microenvironments. Biofabrication. 11:0150032018. View Article : Google Scholar : PubMed/NCBI

224 

Do A, Zahrawi F and Mehal WZ: Therapeutic landscape of metabolic dysfunction-associated steatohepatitis (MASH). Nat Rev Drug Discov. 24:171–189. 2025. View Article : Google Scholar

225 

Dai Q, Ain Q, Seth N, Rooney M and Zipprich A: Liver sinusoidal endothelial cells: Friend or foe in metabolic dysfunction-associated steatotic liver disease/metabolic dysfunction-associated steatohepatitis. Dig Liver Dis. 57:493–503. 2025. View Article : Google Scholar : PubMed/NCBI

226 

Kuchay MS, Choudhary NS and Ramos-Molina B: Pathophysiological underpinnings of metabolic dysfunction-associated steatotic liver disease. Am J Physiol Cell Physiol. 328:C1637–C1666. 2025. View Article : Google Scholar : PubMed/NCBI

227 

Sun HS, Nie CC, Li P, Liu Y and Gao L: Pharmacological treatment of non-alcoholic fatty liver disease in rats induced by high-fat diet combined with chronic stress. Chin J Gerontol. 40:177–181. 2020.In Chinese.

228 

Cheng F, Ma C, Wang X, Zhai C, Wang G, Xu X, Mu J, Li C, Wang Z, Zhang X, et al: Effect of traditional Chinese medicine formula sinisan on chronic restraint stress-induced nonalcoholic fatty liver disease: A rat study. BMC Complement Altern Med. 17:2032017. View Article : Google Scholar : PubMed/NCBI

229 

Cardoso A, Ferreira SC, Araújo MA, Mendes R, do Carmo LO, Moreira L, Ferreira L, Couto CA, Reis IA, Anastácio LR and Faria LC: Association between body composition, eating behavior, dietary intake, and metabolic dysfunction-associated steatotic liver disease in liver transplant recipients. Clin Nutr. 52:154–161. 2025. View Article : Google Scholar : PubMed/NCBI

230 

Zhong Q, Zhou R, Huang YN, Huang RD, Li FR, Chen HW, Wei YF, Liu K, Cao BF, Liao KY, et al: Frailty and risk of metabolic dysfunction-associated steatotic liver disease and other chronic liver diseases. J Hepatol. 82:427–437. 2025. View Article : Google Scholar

231 

Kineman RD, Del Rio-Moreno M and Waxman DJ: Liver-specific actions of GH and IGF1 that protect against MASLD. Nat Rev Endocrinol. 21:105–117. 2025. View Article : Google Scholar

232 

Liu M, Zhang H, Niu M and Shen Y: Protective effects of decoction for strengthening spleen, tonifying qi, activating blood in rats with type 2 diabetes and non-alcoholic fatty liver disease. Mod J Integr Tradit Chin West Med. 31:3246–3251. 32682022.In Chinese.

233 

Shi H: Effect of HYRG capsule on glucose and lipid metabolism-related enzymes oxidiation products in rats with qi and blood stasis type nonalcohlic fatty liver disease. Lanzhou: Gansu Univ Chin Med; 2019, In Chinese.

234 

Chitturi S, Wong VW, Chan WK, Wong GL, Wong SK, Sollano J, Ni YH, Liu CJ, Lin YC, Lesmana LA, et al: The Asia pacific working party on non-alcoholic fatty liver disease guidelines 2017-Part 2: Management and special groups. J Gastroenterol Hepatol. 33:86–98. 2018. View Article : Google Scholar

235 

Gjorgjieva M, Sobolewski C, Dolicka D, Correia de Sousa M and Foti M: MiRNAs and NAFLD: From pathophysiology to therapy. Gut. 68:2065–2079. 2019. View Article : Google Scholar : PubMed/NCBI

236 

Hanin G, Yayon N, Tzur Y, Haviv R, Bennett ER, Udi S, Krishnamoorthy YR, Kotsiliti E, Zangen R, Efron B, et al: MiRNA-132 induces hepatic steatosis and hyperlipidaemia by synergistic multitarget suppression. Gut. 67:1124–1134. 2018. View Article : Google Scholar

237 

Huang TT, Zhou YR, Zhang XJ and Liu SS: Advances in the treatment of nonalcoholic fatty liver disease. Med Rev. 21:1406–1409. 2015.In Chinese.

238 

Liu JL and Li SH: Efficacy and safety of evolocumab and alirocumab in reducing lipids and cardiovascular events. J Evid-Based Med. 15:334–338. 2015.In Chinese.

239 

Steffens D, Bramlage P, Scheeff C, Kasner M, Hassanein A, Friebel J and Rauch-Kröhnert U: PCSK9 inhibitors and cardiovascular outcomes. Expert Opin Biol Ther. 20:35–47. 2020. View Article : Google Scholar

240 

Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Packard CJ, Bayes-Genis A, Cikes M, Fruchart GC, et al: 2019 ESC/EAS guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J. 41:111–188. 2020. View Article : Google Scholar

241 

Yerevanian A and Soukas AA: Metformin: Mechanisms in human obesity and weight loss. Curr Obes Rep. 8:156–164. 2019. View Article : Google Scholar : PubMed/NCBI

242 

Cui H and Zhang X: Occurrence and clinical management of nonalcoholic fatty liver disease in obesity patients: A literature review. J Pediatr Endocrinol Metab. 33:579–584. 2020. View Article : Google Scholar : PubMed/NCBI

243 

Adorini L, Pruzanski M and Shapiro D: Farnesoid X receptor targeting to treat nonalcoholic steatohepatitis. Drug Discov Today. 17:988–997. 2012. View Article : Google Scholar : PubMed/NCBI

244 

Mudaliar S, Henry RR, Sanyal AJ, Morrow L, Marschall HU, Kipnes M, Adorini L, Sciacca CI, Clopton P, Castelloe E, et al: Efficacy and safety of the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty liver disease. Gastroenterology. 145:574–582.e1. 2013. View Article : Google Scholar : PubMed/NCBI

245 

Sundelin EIO, Gormsen LC, Heebøll S, Vendelbo MH, Jakobsen S, Munk OL, Feddersen S, Brøsen K, Hamilton-Dutoit SJ, Pedersen SB, et al: Hepatic exposure of metformin in patients with non-alcoholic fatty liver disease. Br J Pharmacol. 85:1761–1770. 2019.

246 

Knudsen LB and Lau J: The discovery and development of liraglutide and semaglutide. Front Endocrinol (Lausanne). 10:1552019. View Article : Google Scholar : PubMed/NCBI

247 

López-Vicario C, González-Périz A, Rius B, Morán-Salvador E, García-Alonso V, Lozano JJ, Bataller R, Cofán M, Kang JX, Arroyo V, et al: Molecular interplay between Δ5/Δ6 desaturases and long-chain fatty acids in the pathogenesis of non-alcoholic steatohepatitis. Gut. 63:344–355. 2014. View Article : Google Scholar

248 

Liu BB and Xu KS: Drug therapy for non-alcoholic fatty liver disease. Chin J Pract Intern Med. 39:214–217. 2019.In Chinese.

249 

Abenavoli L, Falalyeyeva T, Boccuto L, Tsyryuk O and Kobyliak N: Obeticholic acid: A new era in the treatment of nonalcoholic fatty liver disease. Pharmaceuticals. 11:1042018. View Article : Google Scholar : PubMed/NCBI

250 

Bell DSH and Jerkins T: In praise of pioglitazone: An economically efficacious therapy for type 2 diabetes and other manifestations of the metabolic syndrome. Diabetes Obes Metab. 25:3093–3102. 2023. View Article : Google Scholar : PubMed/NCBI

251 

Sumida Y and Yoneda M: Current and future pharmacological therapies for NAFLD/NASH. J Gastroenterol. 53:362–376. 2018. View Article : Google Scholar :

252 

Chen F, Fei X, Wang J, Shen Z, Liu W, Shao Z, Chen Y and Zhu Z: Effects of Qi-Ho expectorant formula on serum leptin and lipocalin in patients with non-alcoholic fatty liver disease. Lishizhen Med Mater Med Res. 26:843–844. 2015.In Chinese.

253 

Lewis JD, Habel LA, Quesenberry CP, Strom BL, Peng T, Hedderson MM, Ehrlich SF, Mamtani R, Bilker W, Vaughn DJ, et al: Pioglitazone use and risk of bladder cancer and other common cancers in persons with diabetes. JAMA. 314:265–277. 2015. View Article : Google Scholar : PubMed/NCBI

254 

Harrison SA, Alkhouri N, Davison BA, Sanyal A, Edwards C, Colca JR, Lee BH, Loomba R, Cusi K, Kolterman O, et al: Insulin sensitizer MSDC-0602K in non-alcoholic steatohepatitis: A randomized, double blind, placebo-controlled phase IIb study. J Hepatol. 72:613–626. 2020. View Article : Google Scholar

255 

Chen Z, Tian R, She Z, Cai J and Li H: Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radic Biol Med. 152:116–141. 2020. View Article : Google Scholar : PubMed/NCBI

256 

Nagashimada M and Ota T: Role of vitamin E in nonalcoholic fatty liver disease. IUBMB Life. 71:516–522. 2019. View Article : Google Scholar

257 

Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM, Bass NM, Neuschwander-Tetri BA, Lavine JE, Tonascia J, Unalp A, et al: Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N Engl J Med. 362:1675–1685. 2010. View Article : Google Scholar : PubMed/NCBI

258 

Lavine JE: Vitamin E treatment of nonalcoholic steatohepatitis in children: A pilot study. J Pediatr. 136:734–738. 2000. View Article : Google Scholar : PubMed/NCBI

259 

Liu YL and Zhang QZ: An excerpt of the Asia-Pacific Working Party on nonalcoholic fatty liver disease guidelines 2017. J Clin Hepatol. 33:2278–2282. 2017.In Chinese.

260 

Wah Kheong C, Nik Mustapha NR and Mahadeva S: A randomized trial of silymarin for the treatment of nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol. 15:1940–1949.e8. 2017. View Article : Google Scholar

261 

Zhu X, Wu M, Wang H, Li H, Lin J, Peng Y, Hu Y, Li C and Ding Y: Safety, tolerability, and pharmacokinetics of the novel pan-phosphodiesterase inhibitor ZSP1601 in healthy subjects: A double-blinded, placebo-controlled first-in-human single-dose and multiple-dose escalation and food effect study. Expert Opin Investig Drugs. 30:579–589. 2021. View Article : Google Scholar : PubMed/NCBI

262 

Hu Y, Li H, Zhang H, Chen X, Chen J, Xu Z, You H, Dong R, Peng Y, Li J, et al: ZSP1601, a novel pan-phosphodiesterase inhibitor for the treatment of NAFLD, A randomized, placebo-controlled phase Ib/IIa trial. Nat Commun. 14:64092023. View Article : Google Scholar : PubMed/NCBI

263 

Xiang M, Wang PX, Wang AB, Zhang XJ, Zhang Y, Zhang P, Mei FH, Chen MH and Li H: Targeting hepatic TRAF1-ASK1 signaling to improve inflammation, insulin resistance, and hepatic steatosis. J Hepatol. 64:1365–1377. 2016. View Article : Google Scholar : PubMed/NCBI

264 

Budas G, Karnik S, Jonnson T, Shafizadeh T, Watkins S, Breckenridge D and Tumas D: Reduction of liver steatosis and fibrosis with an ASK1 inhibitor in a murine model of NASH is accompanied by improvements in cholesterol, bile acid and lipid metabolism. J Hepatol. 64:S0702016. View Article : Google Scholar

265 

Harrison SA, Wong VW, Okanoue T, Bzowej N, Vuppalanchi R, Younes Z, Kohli A, Sarin S, Caldwell SH, Alkhouri N, et al: Selonsertib for patients with bridging fibrosis or compensated cirrhosis due to NASH: Results from randomized phase III STELLAR trials. J Hepatol. 73:26–39. 2020. View Article : Google Scholar : PubMed/NCBI

266 

Lefere S, Devisscher L and Tacke F: Targeting CCR2/5 in the treatment of nonalcoholic steatohepatitis (NASH) and fibrosis: opportunities and challenges. Expert Opin Investig Drugs. 29:89–92. 2020. View Article : Google Scholar : PubMed/NCBI

267 

Kruger AJ, Fuchs BC, Masia R, Holmes JA, Salloum S, Sojoodi M, Ferreira DS, Rutledge SM, Caravan P, Alatrakchi N, et al: Prolonged cenicriviroc therapy reduces hepatic fibrosis despite steatohepatitis in a diet-induced mouse model of nonalcoholic steatohepatitis. Hepatol Commun. 2:529–545. 2018. View Article : Google Scholar : PubMed/NCBI

268 

Ratziu V, Sanyal A, Harrison SA, Wong VW, Francque S, Goodman Z, Aithal GP, Kowdley KV, Seyedkazemi S, Fischer L, et al: Cenicriviroc treatment for adults with nonalcoholic steatohepatitis and fibrosis: Final analysis of the phase 2b centaur study. Hepatology. 72:892–905. 2020. View Article : Google Scholar : PubMed/NCBI

269 

Anstee QM, Neuschwander-Tetri BA, Wong VW, Abdelmalek MF, Younossi ZM, Yuan J, Pecoraro ML, Seyedkazemi S, Fischer L, Bedossa P, et al: Cenicriviroc for the treatment of liver fibrosis in adults with nonalcoholic steatohepatitis: Aurora phase 3 study design. Contemp Clin Trials. 89:1059222020. View Article : Google Scholar

270 

Pawlak M, Lefebvre P and Staels B: Molecular mechanism of PPARα action and its impact on lipid metabolism, inflammation and fibrosis in non-alcoholic fatty liver disease. J Hepatol. 62:720–733. 2015. View Article : Google Scholar

271 

Ratziu V, Harrison SA, Francque S, Bedossa P, Lehert P, Serfaty L, Romero-Gomez M, Boursier J, Abdelmalek M, Caldwell S, et al: Elafibranor, an agonist of the peroxisome proliferator-activated receptor-α and-δ, induces resolution of nonalcoholic steatohepatitis without fibrosis worsening. Gastroenterology. 150:1147–1159.e5. 2016. View Article : Google Scholar

272 

Wang J, Sun F, Yang JJ and Lu C: Effect of DNMT3A regulation of Drp1 on activated proliferation and migration capacity of hepatic stellate cells. Chin Pharmacol Bull. 38:1542–1547. 2022.In Chinese.

273 

Chalasani N, Abdelmalek MF, Garcia-Tsao G, Vuppalanchi R, Alkhouri N, Rinella M, Noureddin M, Pyko M, Shiffman M, Sanyal A, et al: Effects of belapectin, an inhibitor of Galectin-3, in patients with nonalcoholic steatohepatitis with cirrhosis and portal hypertension. Gastroenterology. 158:1334–1345.e5. 2020. View Article : Google Scholar

274 

Bi Z, Li G, Chen G, Yang Z, Long F, Luo L, Yang B and Tang D: Exploring the correlation between intestinal flora dysbiosis and hepatocellular carcinoma based on the theory of righteousness and evil in Chinese medicine. Mod J Integr Tradit Chin West Med. 31:1229–1234. 2022.In Chinese.

275 

Said I, Ahad H and Said A: Gut microbiome in non-alcoholic fatty liver disease associated hepatocellular carcinoma: Current knowledge and potential for therapeutics. World J Gastrointest Oncol. 14:947–958. 2022. View Article : Google Scholar : PubMed/NCBI

276 

Liu Q, Liu Y, Li F, Gu Z, Liu M, Shao T, Zhang L, Zhou G, Pan C, He L, et al: Probiotic culture supernatant improves metabolic function through FGF21-adiponectin pathway in mice. J Nutr Biochem. 75:1082562020. View Article : Google Scholar

277 

Malaguarnera M, Vacante M, Antic T, Giordano M, Chisari G, Acquaviva R, Mastrojeni S, Malaguarnera G, Mistretta A, Li Volti G and Galvano F: Bifidobacterium longum with fructo-oligosaccharides in patients with nonalcoholic steatohepatitis. Dig Dis Sci. 57:545–553. 2012. View Article : Google Scholar

278 

Wang H, Chen Q, Li H and Zhang Z: Adiponectin alleviates LPS-induced inflammatory response of microglia through A20/NLRP3/caspase-1 pathway. J Xi'an Jiaotong Univ (Med Sci). 42:529–533. 2021.In Chinese.

279 

Fernandes P, Hashiguchi T, Fujii M and Yoneyama H: Anti-NASH effects of solithromycin in NASH-HCC mouse model. Gastroenterology. 146:145–146. 2014. View Article : Google Scholar

280 

Kannt A, Wohlfart P, Madsen AN, Veidal SS, Feigh M and Schmoll D: Activation of thyroid hormone receptor-β improved disease activity and metabolism independent of body weight in a mouse model of non-alcoholic steatohepatitis and fibrosis. Br J Pharmacol. 178:2412–2423. 2021. View Article : Google Scholar : PubMed/NCBI

281 

Mavromati M and Jornayvaz FR: Hypothyroidism-associated dyslipidemia: Potential molecular mechanisms leading to NAFLD. Int J Mol Sci. 22:127972021. View Article : Google Scholar : PubMed/NCBI

282 

Hu L, Gu Y, Liang J, Ning M, Yang J, Zhang Y, Qu H, Yang Y, Leng Y and Zhou B: Discovery of highly potent and selective thyroid hormone receptor β agonists for the treatment of nonalcoholic steatohepatitis. J Med Chem. 66:3284–3300. 2023. View Article : Google Scholar : PubMed/NCBI

283 

Karim G and Bansal MB: Resmetirom: An orally administered, small-molecule, liver-directed, β-selective THR agonist for the treatment of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. Touch Rev Endocrinol. 19:60–70. 2023.

284 

Harrison SA, Bedossa P, Guy CD, Schattenberg JM, Loomba R, Taub R, Labriola D, Moussa SE, Neff GW, Rinella ME, et al: A phase 3, randomized, controlled trial of resmetirom in NASH with liver fibrosis. N Engl J Med. 390:497–509. 2024. View Article : Google Scholar : PubMed/NCBI

285 

Loomba R, Neutel J, Mohseni R, Bernard D, Severance R, Dao M, Saini S, Margaritescu C, Homer K, Tran B, et al: VK2809, a novel liver-directed thyroid receptor beta agonist, significantly reduces liver fat with both low and high doses in patients with non-alcoholic fatty liver disease: A phase 2 randomized, placebo-controlled trial. J Hepatology. 70:e150–e151. 2019. View Article : Google Scholar

286 

Leng YR, Zhang MH, Luo JG and Zhang H: Pathogenesis of NASH and promising natural products. Chin J Nat Med. 19:12–27. 2021.PubMed/NCBI

287 

Chen M, Xie Y, Gong S, Wang Y, Yu H, Zhou T, Huang F, Guo X, Zhang H, Huang R, et al: Traditional Chinese medicine in the treatment of nonalcoholic steatohepatitis. Pharmacol Res. 172:1058492021. View Article : Google Scholar : PubMed/NCBI

288 

Dai X, Feng J, Chen Y, Huang S, Shi X, Liu X and Sun Y: Traditional Chinese medicine in nonalcoholic fatty liver disease: molecular insights and therapeutic perspectives. Chin Med. 16:682021. View Article : Google Scholar : PubMed/NCBI

289 

Deng Y, Liu H, Wu X, Cheng L, He M, Wang T and Ye L: Research progress on the hepatoprotective effects of polydatin. Chin J Comp Med. 31:136–140. 2021.In Chinese.

290 

Pan L, Lv B, Jiang X, Wang T, Ma X, Chang N, Wang X and Gao X: Identification of NF-κB inhibitors following shenfu injection and bioactivity-integrated UPLC/Q-TOF-MS and screening for related anti-inflammatory targets in vitro and in silico. J Ethnopharmacol. 19:658–667. 2016.

291 

Li L, Wang YL, Qin HY, Qu YY, Yang TS, Wang ZY and Xie JR: Research progress on berberine in treatment of nonalcoholic fatty liver disease by regulating gut-liver axis. Chin Tradit Herb Drugs. 52:1501–1509. 2021.In Chinese.

292 

Liu YL, Zhang QZ, Wang YR, Fu LN, Han JS, Zhang J and Wang BM: Astragaloside IV Improves high-fat diet-induced hepatic steatosis in nonalcoholic fatty liver disease rats by regulating inflammatory factors level via TLR4/NF-κB signaling pathway. Front Pharmacol. 11:6050642020. View Article : Google Scholar

293 

Zhao ZM, Wu JZ, Yao Z, Li YJ, Hou W, Chen WH and Shi AH: Effects of Cassia glycosides on the rats with non-alcoholic fatty liver disease through Toll-like receptor 4 and nuclear factor-κB. Chin J Clin Pharmacol. 35:2863–2867. 2019.In Chinese.

294 

Paudel P, Jung HA and Choi JS: Anthraquinone and naphthopyrone glycosides from cassia obtusifolia seeds mediate hepatoprotection via Nrf2-mediated HO-1 activation and MAPK modulation. Arch Pharm Res. 41:677–689. 2018. View Article : Google Scholar : PubMed/NCBI

295 

Zhang Y, Cheng S, Zhou F, Zhang L, Su Z, Zhao D, Chen Y and Jia Y: Effects of geniposide on inflammation and oxidative stress of ApoE knockout mice with atherosclerosis and none-alcoholic fatty liver disease. Tradit Chin Drug Res Clin Pharmacol. 26:581–586. 2015.In Chinese.

296 

Lee JH, Jung IR, Choi SE, Lee SM, Lee SJ, Han SJ, Kim HJ, Kim DJ, Lee KW and Kang Y: Toxicity generated through inhibition of pyruvate carboxylase and carnitine palmitoyl transferase-1 is similar to high glucose/palmitate-induced glucolipotoxicity in INS-1 beta cells. Mol Cell Endocrinol. 383:48–59. 2014. View Article : Google Scholar

297 

Xu QM, Gao Y, Li ZM, Wei RM, Ma XH, Jin L and Zhang KF: The effect of gentiopicroside on TLR-4/NF-κB and AMPK/Nrf2 in non-alcoholic fatty liver disease. Nat Prod Res Dev. 32:1652–1658. 2020.In Chinese.

298 

Molteni M, Gemma S and Rossetti C: The role of toll-like receptor 4 in infectious and noninfectious inflammation. Mediators Inflamm. 2016:69789362016. View Article : Google Scholar : PubMed/NCBI

299 

Wang QB, Xu FP, Wei CX, Peng J and Dong XD: Research progress on free radicals in the human body. Zhonghua Liu Xing Bing Xue Za Zhi. 37:1175–1182. 2016.In Chinese. PubMed/NCBI

300 

Huang Z, Wei Y, Wang Z, Lai PL, Zeng BH, Xu JJ and Yang YL: Effects of total glucosides paeony on TLR4 and JNK protein expression in inflammatory pathway of NAFLD rats. J Hunan Normal Univ (Med Sci). 15:23–27. 2018.In Chinese.

301 

Shen S, Wang K, Zhi Y, Shen W and Huang L: Gypenosides improves nonalcoholic fatty liver disease induced by high-fat diet induced through regulating LPS/TLR4 signaling pathway. Cell Cycle. 19:3042–3053. 2020. View Article : Google Scholar : PubMed/NCBI

302 

Li X, Fang C, Xu Y, Xu B and Zheng X: Effect of hesperidin on CYP2E1 expression in nonalcoholic fatty liver rats induced by high fructose diet. Mod J Integr Tradit Chin West Med. 29:2635–2639. 2020.In Chinese.

303 

Park S, Choi Y, Um SJ, Yoon SK and Park T: Oleuropein attenuates hepatic steatosis induced by high-fat diet in mice. J Hepatol. 54:984–993. 2011. View Article : Google Scholar

304 

Chen M, Liu G, Fang Z, Gao W, Song Y, Lei L, Du X and Li X: Buddleoside alleviates nonalcoholic steatohepatitis by targeting the AMPK-TFEB signaling pathway. Autophagy. 21:1316–1334. 2025. View Article : Google Scholar : PubMed/NCBI

305 

Yin Y, Liu H, Zheng Z, Lu R and Jiang Z: Genistein can ameliorate hepatic inflammatory reaction in nonalcoholic steatohepatitis rats. Biomed Pharmacother. 111:1290–1296. 2019. View Article : Google Scholar : PubMed/NCBI

306 

Liu X, Sun R, Li Z, Lv P, Sun X, Olson MA and Gong Y: Luteolin alleviates non-alcoholic fatty liver disease in rats via restoration of intestinal mucosal barrier damage and microbiota imbalance involving in gut-liver axis. Arch Biochem Biophys. 711:1090192021. View Article : Google Scholar : PubMed/NCBI

307 

Kou X, Hong M, Pan F, Huang X, Meng Q, Zhang Y and Ke Q: Inhibitory effects of nobiletin-mediated interfacial instability of bile salt emulsified oil droplets on lipid digestion. Food Chem. 444:1387512024. View Article : Google Scholar : PubMed/NCBI

308 

He TT, Qian YL and Chen WM: Study on constitution classification of TCM for 260 nonalcoholic fatty liver disease patients. Inner Mong J Tradit Chin Med. 37:1–2. 2018.In Chinese.

309 

Pan YT, Xu FY, Yu XZ and Shang WB: Research progress on therapeutic targets of active components in Chinese herbs for treatment of nonalcoholic fatty liver disease. Zhongguo Zhong Yao Za Zhi. 42:1109–1112. 2017.In Chinese. PubMed/NCBI

310 

Liang ZQ, Bai JH, Zhao RH, Liu YH, Li LM and Lin QX: Effect of ginkgo flavone on nonalcoholic fatty liver disease in mice and the correlation analysis on NF-κB in the action mechanism. Nat Prod Res Dev. 28:277–282. 2016.In Chinese.

311 

Huang L, Ding W, Wang MQ, Wang ZG, Chen HH, Chen W, Yang Q, Lu TN, Yang Q and He JM: Tanshinone IIA ameliorates non-alcoholic fatty liver disease through targeting peroxisome proliferator-activated receptor gamma and toll-like receptor 4. J Int Med Res. 47:5239–5255. 2019. View Article : Google Scholar : PubMed/NCBI

312 

Zheng Y, Wang J, Wang J, Xie H and Zhao T: Effect of curcumol on the fenestrae of liver sinusoidal endothelial cells based on NF-κB signaling pathway. Evid Based Complement Alternat Med. 2020:85906382020. View Article : Google Scholar

313 

Han LP, Sun B, Li CJ, Xie Y and Chen LM: Effect of celastrol on toll-like receptor 4-mediated inflammatory response in free fatty acid-induced HepG2 cells. Int J Mol Med. 42:2053–2061. 2018.PubMed/NCBI

314 

Ye Z, Zhu K, He Y, Chen L and Zhang J: Mechanism of asiatic acid improving nonalcoholic fatty liver disease by regulating TLR4/NF-κB/NLRP3 signaling pathway. China Food Addit. 35:201–207. 2024.In Chinese.

315 

Malekinejad H, Zeynali-Moghaddam S, Rezaei-Golmisheh A, Alenabi A, Malekinejad F, Alizadeh A and Shafie-Irannejad V: Lupeol attenuated the NAFLD and PCOS-induced metabolic, oxidative, hormonal, histopathological, and molecular injuries in mice. Res Pharm Sci. 18:551–565. 2023. View Article : Google Scholar : PubMed/NCBI

316 

Brusotti G, Montanari R, Capelli D, Cattaneo G, Laghezza A, Tortorella P, Loiodice F, Peiretti F, Bonardo B, Paiardini A, et al: Betulinic acid is a PPARγ antagonist that improves glucose uptake, promotes osteogenesis and inhibits adipogenesis. Sci Rep. 7:57772017. View Article : Google Scholar

317 

Hou L and Xiao Y: Study on the distribution of Chinese medicine evidence elements in non-alcoholic fatty liver disease. Mod J Integr Tradit Chin West Med. 24:2116–2118. 2015.In Chinese.

318 

Zhang L, Zhao Y, Li Z, Guo D and Fu S: Correlative study between ultrasonic echo intensity parameters and Traditional Chinese Medicine syndromes of fatty liver. Chin J Ultrasound Med. 28:55–58. 2012.In Chinese.

319 

He F, Zhang X and Wen X: Effects of crataegolic acid on inflammatory response and oxidative stress in non-alcoholic fatty liver disease model mice induced by high-fat diet. China Pharm. 30:901–905. 2019.In Chinese.

320 

Zhou X, Ren Q, Wang B, Fang G, Ling Y and Li X: Alisol A 24-Acetate Isolated from the alismatis rhizoma improves hepatic lipid deposition in hyperlipidemic mice by ABCA1/ABCG1 pathway. J Nanosci Nanotechnol. 19:5496–5502. 2019. View Article : Google Scholar : PubMed/NCBI

321 

Ho C, Gao Y, Zheng D, Liu Y, Shan S, Fang B, Zhao Y, Song D, Zhang Y and Li Q: Alisol A attenuates high-fat-diet-induced obesity and metabolic disorders via the AMPK/ACC/SREBP1c pathway. J Cell Mol Med. 23:5108–5118. 2019. View Article : Google Scholar : PubMed/NCBI

322 

Li X, Chen XX, Xu Y, Xu XB, Wu WF, Zhao Q and Hu JN: Construction of Glycogen-Based Nanoparticles loaded with resveratrol for the alleviation of high-fat diet-induced nonalcoholic fatty liver disease. Biomacromolecules. 23:409–423. 2022. View Article : Google Scholar

323 

Feng D, Zou J, Su D, Mai H, Zhang S, Li P and Zheng X: Curcumin prevents high-fat diet-induced hepatic steatosis in ApoE−/− mice by improving intestinal barrier function and reducing endotoxin and liver TLR4/NF-κB inflammation. Nutr Metab (Lond). 16:792019. View Article : Google Scholar

324 

Zhang M, Zhang Y, Tang L, Gong Z, Han L and Wang D: Gastrodin inhibits non-alcoholic fatty liver disease via mediating SREBP1c signaling pathway. Chin J Exp Tradit Med Formul. 30:70–77. 2024.In Chinese.

325 

Xiao J, Zhang R, Zhou Q, Liu L, Huang F, Deng Y, Ma Y, Wei Z, Tang X and Zhang M: Lychee (litchi chinensis sonn.) pulp phenolic extract provides protection against alcoholic liver injury in mice by alleviating intestinal microbiota dysbiosis, intestinal barrier dysfunction and liver inflammation. J Agric Food Chem. 65:9675–9684. 2017. View Article : Google Scholar : PubMed/NCBI

326 

Aithal GP, Thomas JA, Kaye PV, Lawson A, Ryder SD, Spendlove I, Austin AS, Freeman JG, Morgan L and Webber J: Randomized, placebo-controlled trial of pioglitazone in nondiabetic subjects with nonalcoholic steatohepatitis. Gastroenterology. 135:1176–1184. 2008. View Article : Google Scholar : PubMed/NCBI

327 

Castellino G, Nikolic D, Magán-Fernández A, Malfa GA, Chianetta R, Patti AM, Amato A, Montalto G, Toth PP, Banach M, Cicero AFG and Rizzo M: Altilix® supplement containing chlorogenic acid and luteolin improved hepatic and cardiometabolic parameters in subjects with metabolic syndrome: A 6 month randomized, double-blind, placebo-controlled study. Nutrients. 11:25802019. View Article : Google Scholar

328 

Tan Y, Kim J, Cheng J, Ong M, Lao WG, Jin XL, Lin YG, Xiao L, Zhu XQ and Qu XQ: Green tea polyphenols ameliorate non-alcoholic fatty liver disease through upregulating AMPK activation in high fat fed Zucker fatty rats. World J Gastroenterol. 23:3805–3814. 2017. View Article : Google Scholar : PubMed/NCBI

329 

Chen X, Ren Y, Kong W and Wang Y: Effect of salvianolic acid B on oxidative stress in a cell model of nonalcoholic fatty liver disease. J Clin Hepatol. 34:2175–2181. 2018.In Chinese.

330 

Zhang X, Li T, Liu S, Wu Q, Lin Z, Pan Y and Alitongbieke G: Mechanism of berberine improving non-alcoholic fatty liver induced by high-fat diet in mice. Chin J Bioprocess Eng. 19:657–661. 2021.In Chinese.

331 

Kim R, Kim SB, Cho EH, Park SH, Park SB, Hong SK and Chae G: CD44 expression in patients with combined hepatocellular cholangiocarcinoma. Ann Surg Treat Res. 89:9–16. 2015. View Article : Google Scholar : PubMed/NCBI

332 

Sun T, Xue M, Yang J, Pei Z, Zhang N, Qin K and Liang H: Metabolic regulation mechanism of fucoidan via intestinal microecology in diseases. J Sci Food Agric. 101:4456–4463. 2021. View Article : Google Scholar : PubMed/NCBI

333 

Cani PD and Delzenne NM: The role of the gut microbiota in energy metabolism and metabolic disease. Curr Pharm Des. 15:1546–1558. 2009. View Article : Google Scholar : PubMed/NCBI

334 

Zhang DD, Zhang JG, Wang YZ, Liu Y, Liu GL and Li XY: Per-Arnt-Sim kinase (PASK): An emerging regulator of mammalian glucose and lipid metabolism. Nutrients. 7:7437–7450. 2015. View Article : Google Scholar : PubMed/NCBI

335 

Rahban M, Zolghadri S, Salehi N, Ahmad F, Haertlé T, Rezaei-Ghaleh N, Sawyer L and Saboury AA: Thermal stability enhancement: fundamental concepts of protein engineering strategies to manipulate the flexible structure. Int J Biol Macromol. 214:642–644. 2022. View Article : Google Scholar : PubMed/NCBI

336 

Zhong M, Yan Y, Yuan H, A R, Xu G, Cai F, Yang Y, Wang Y and Zhang W: Astragalus mongholicus polysaccharides ameliorate hepatic lipid accumulation and inflammation as well as modulate gut microbiota in NAFLD rats. Food Funct. 13:7287–7301. 2022. View Article : Google Scholar : PubMed/NCBI

337 

Wang L, Li W, Li Y, Chen G, Zhao L, Li W, Wang S, Wang C, Feng Y and Zhang Y: Dried tangerine peel polysaccharide (DTPP) alleviates hepatic steatosis by suppressing TLR4/MD-2-mediated inflammation and endoplasmic reticulum stress. Bioorg Chem. 147:1073692024. View Article : Google Scholar : PubMed/NCBI

338 

Yu J, Fan Y, Yang Y, He D, Song B and Yao Z: Influence of dendrobium nobile polysaccharide on the expression of TLR4 and HO-1 in rats with nonalcoholic fatty liver disease. West J Tradit Chin Med. 34:25–29. 2021.In Chinese.

339 

Liu HP, Zhang RJ, Cao HK, Gao Y and Wang YW: Study on effects of Polygala fallax polysaccharide on nonalcoholic fatty liver in rats by PPAR-γ and TLR-4/NF-κB signaling pathway. J Chin Med Mater. 47:1247–1252. 2024.In Chinese.

340 

Zhang K and Jin L: Protective effect and mechanism of dicliptera chinensis polysaccharides on non-alcoholic fatty liver disease in rats based on PPAR-γ and TLR-4/NF-κB signaling pathway. Pharmacol Clin Chin Mater Med. 35:103–107. 2019.In Chinese.

341 

Zhao H, Gao X, Liu Z, Zhang L, Fang X, Sun J, Zhang Z and Sun Y: Sodium alginate prevents non-alcoholic fatty liver disease by modulating the gut-liver axis in high-fat diet-fed rats. Nutrients. 14:48462022. View Article : Google Scholar : PubMed/NCBI

342 

Liu X, Su J, Shi Y, Guo Y, Suheryani I, Zhao S, Deng Y, Meng W, Chen Y, Sun L and Dai R: Herbal Formula, Baogan Yihao (BGYH), prevented dimethylnitrosamine(DMN)-indu ced liver injury in rats. Drug Dev Res. 78:155–163. 2017. View Article : Google Scholar : PubMed/NCBI

343 

Xie R, Lin D, Yang L and Li F: Protective effect and mechanism of zingerone on rats with nonalcoholic fatty liver disease. J Shenyang Pharm Univ. 39:684–689. 2022.In Chinese.

344 

Lv YL, Liu CC, Liu H, Xu KY, Qiao L and Bao JF: Effects of rhein on TLR4 signaling pathway in NAFLD rats. Chin J Health Lab Tec. 28:2580–2584. 2018.In Chinese.

345 

Wei J, Zhen YZ, Cui J, He FL, Shen T, Hu G, Ren XH and Lin YJ: Rhein lysinate decreases inflammation and adipose infiltration in KK/HlJ diabetic mice with nonalcoholic fatty liver disease. Arch Pharm Res. 39:960–969. 2016. View Article : Google Scholar : PubMed/NCBI

346 

Dong X, Fu J, Yin X, Cao S, Li X, Lin L, Huyiligeqi and Ni J: Emodin: a review of its pharmacology, toxicity and pharmacokinetics. Phytother Res. 30:1207–1218. 2016. View Article : Google Scholar : PubMed/NCBI

347 

Ye J, Zheng J, Tian X, Xu B, Yuan F, Wang B, Yang Z and Huang F: Fucoxanthin attenuates free fatty acid-induced nonalcoholic fatty liver disease by regulating lipid metabolism/oxidative stress/inflammation via the AMPK/Nrf2/TLR4 signaling pathway. Mar Drugs. 20:2252022. View Article : Google Scholar : PubMed/NCBI

348 

Borstlap WAA, Musters GD, Stassen LPS, van Westreenen HL, Hess D, van Dieren S, Festen S, van der Zaag EJ, Tanis PJ and Bemelman WA: Vacuum-assisted early transanal closure of leaking low colorectal anastomoses: The CLEAN study. Surg Endosc. 32:315–327. 2018. View Article : Google Scholar :

349 

Yu S, Jiang J, Li Q, Liu X, Wang Z, Yang L and Ding L: Schisantherin a alleviates non-alcoholic fatty liver disease by restoring intestinal barrier function. Front Cell Infect Microbiol. 12:8550082022. View Article : Google Scholar : PubMed/NCBI

350 

Gu L, Zhang Y, Zhang S, Zhao H, Wang Y, Kan D, Zhang Y, Guo L, Lv J, Hao Q, et al: Coix lacryma-jobi seed oil reduces fat accumulation in nonalcoholic fatty liver disease by inhibiting the activation of the p-AMPK/SePP1/apoER2 pathway. J Oleo Sci. 70:685–696. 2021. View Article : Google Scholar : PubMed/NCBI

351 

Pan MX, Zheng CY, Deng YJ, Tang KR, Nie H, Xie JQ, Liu DD, Tu GF, Yang QH and Zhang YP: Hepatic protective effects of shenling baizhu powder, a herbal compound, against inflammatory damage via TLR4/NLRP3 signalling pathway in rats with nonalcoholic fatty liver disease. J Integr Med. 19:428–438. 2021. View Article : Google Scholar : PubMed/NCBI

352 

Du N, Yang J, Xu D, Zhang S and Shi Z: Fuzi Lizhong decoction modulates bile acid metabolism in nonalcoholic fatty liver disease viathe FXR-FGF15 signaling pathway. Med J West China. 36:1576–1581. 2024.In Chinese.

353 

Liu H, Xu J, Li H, Zhang L and Xu P: Network pharmacology-based investigation to explore the effect and mechanism of erchen decoction against the nonalcoholic fatty liver disease. Anat Rec (Hoboken). 304:2605–2619. 2021. View Article : Google Scholar : PubMed/NCBI

354 

Zhang Y, Zhou G, Chen Z, Guan W, Zhang J, Bi M, Wang F, You X, Liao Y, Zheng S, et al: Si-Wu-Tang alleviates nonalcoholic fatty liver disease via blocking tlr4-jnk and caspase-8-gsdmd signaling pathways. Evid Based Complement Alternat Med. 2020:87864242020. View Article : Google Scholar : PubMed/NCBI

355 

Su L, Lin J, Liu X, Ye X and Chen J: Clinical study on Xiaoyao San prescription combined with silymarin capsules for non-alcoholic fatty liver disease with syndrome of liver depression and spleen deficiency. New Chin Med. 56:42–46. 2024.In Chinese.

356 

Feng W, Su A, Huang X and Wang C: Mechanism of Huangqisan regulating autophagy by AMPK/mTOR signaling pathway against hepatic steatosis. Chin J Exp Tradit Med Formul. 29:21–30. 2023.In Chinese.

357 

Xia M, Zhang Z, Shen H and Kang J: Study on clinical regression of non-alcoholic fatty liver disease by nourishing yin and softening liver method. J New Chin Med. 48:52–54. 2016.In Chinese.

358 

Lu S, Lu S, Chen Y, Shen T and Li Y: Qinghua formula for the treatment of non-alcoholic fatty liver disease with phlegm internal obstruction type: A clinical study. World Sci Technol - Mod Tradit Chin Med. 21:1759–1765. 2019.In Chinese.

359 

Xie WN, Peng HB and Li Y, Huang LP, Chen SX, Wu YF and Li Y: Liver with liver stagnation and spleen deficiency syndrome and intestinal microflora. Chin J Exp Tradit Med Formul. 27:129–137. 2021.In Chinese.

360 

Xu L, Fu J, Fang F, Chen LZ and Zhuang GF: Effect of modfied Yinchen Wuling San in treating non-alcoholic fatty liver disease with moisture and heat implication and on intestinal microflora. Chin J Exp Tradit Med Formul. 25:127–132. 2019.In Chinese.

361 

Zhan YH, Wang XH and Wang F: Regulatory effect of modified Banxia Xiexintang on insulin resistance of patients with nonalcoholic fatty liver. Chin J Exp Tradit Med Formul. 27:117–122. 2021.In Chinese.

362 

Wan Y, Zhang Z, Song G and Ouyang J: Clinical effect of Yishen Tiaogan recipe combined with probiotics on nonalcoholic fatty liver disease. Chin Arch Tradit Chin Med. 38:10–13. 2020.In Chinese.

363 

Chen L, Huang Q, Liu S and Lu Z: Clinical observation of Wuling powder combined with polyene phosphatidylcholine on patients with nonalcoholic fatty liver disease. Chin Arch Tradit Chin Med. 38:190–193. 2020.In Chinese.

364 

Mian F and Yang R: Study on the effects of activating blood and eliminating lipids formula on TNF-α, TGF and FFA in patients with non-alcoholic fatty liver disease. World Latest Med Inf. 18:136–137. 2018.In Chinese.

365 

Zhang Y, Ni Y, Zhang J, Li J and Liu H: Effectiveness of spleen-strengthening and fat-eliminating formula in treating patients with type 2 diabetes mellitus combined with nonalcoholic fatty liver disease and its effect on HMGB1 and TLR4 levels. Lishizhen Med Mater Med Res. 35:261–264. 2024.In Chinese.

366 

Wang S, Ning M, Wang Y, Yang X and Li M: Study on the therapeutic effect and mechanism of anemarrhenae-hawthorn drink on non-alcoholic fatty liver in rats. Anhui Med Pharm J. 27:1927–1932. 2023.In Chinese.

367 

Wang S, Xu WX, Shen MY, Li C, La XJ, Li JA, Qi YJ, Wang XP and Wu Z: Effect of Tongping Zhigan formula on liver tissue inflammation and TLR4/MyD88/NF-κB pathway in non-alcoholic fatty liver mice. J Hebei Tradit Chin Med Pharmacol. 39:1–6. 2024.In Chinese.

368 

Ding Y, Ou X, Li C, Li X and Lian S: An investigation of the mechanism of Qinggan Jiangzhuo particles in the treatment of nonalcoholic fatty liver disease based on the TLR4/NF-κB signaling pathway. Jilin J Chin Med. 41:1489–1492. 2021.In Chinese.

369 

Chen Z, Xiang S, Yang M, Xia F and Zhou B: Study on the molecular mechanism of liver-protecting and lipid-cleansing tablets for the treatment of non-alcoholic fatty liver disease based on network pharmacology. J Chin Med Mater. 43:1462–1468. 2020.In Chinese.

370 

Ye M, Xue J, Li F, Yang Y, He J and Tang Y: Study on regulation and mechanism of Huazhi Fugan granules on TLR4/NF-κB pathway and fatty iron death in non-alcoholic fatty liver. Mod J Integr Tradit Chin West Med. 30:3307–3312. 2021.In Chinese.

371 

Guo HH, Shen HR, Zhang HJ, Wang LL, Han YX and Jiang JD: Dengzhan shengmai inhibits nonalcoholic fatty liver disease via regulating intestinal microenvironment. Acta Pharm Sin. 57:3524–3534. 2022.In Chinese.

372 

Liao Y, Chi X, Yang Y, Wu Y, Zhan Y and Song Y: Effects of Zhibitai on the expressions of ERK and TLRs in liver tissues of NAFLD rats. Pharmacol Clin Chin Mater Med. 35:130–134. 2019.In Chinese.

373 

Yang JF, Xue JB, Tao YY, Huang K, Lu J and Liu CH: Mechanism of Kuhuang Granules in treating non-alcoholic fatty liver disease based on network pharmacology and experimental validation. J Integr Tradit Chin West Med Hepatol. 32:58–62. 2022.In Chinese.

374 

Winn NC, Liu Y, Rector RS, Parks EJ, Ibdah JA and Kanaley JA: Energy-matched moderate and high intensity exercise training improves nonalcoholic fatty liver disease risk independent of changes in body mass or abdominal adiposity-a-randomized trial. Metabolism. 78:128–140. 2018. View Article : Google Scholar

375 

Hannah WN Jr and Harrison SA: Lifestyle and dietary interventions in the management of nonalcoholic fatty liver disease. Dig Dis Sci. 61:1365–1374. 2016. View Article : Google Scholar : PubMed/NCBI

376 

Fogacci F, Rizzoli E, Giovannini M, Bove M, D'Addato S, Borghi C and Cicero AFG: Effect of dietary supplementation with eufortyn colesterolo plus on serum lipids, endothelial reactivity, indexes of non-alcoholic fatty liver disease and systemic inflammation in healthy subjects with polygenic hypercholesterolemia: The ANEMONE study. Nutrients. 14:20992022. View Article : Google Scholar

377 

Fatemeh H, Abdollah H, Bizhan H, Seyed-Saeed S and Kambiz AA: An energy-restricted high-protein diet supplemented with beta-cryptoxanthin alleviated oxidative stress and inflammation in nonalcoholic fatty liver disease: A randomized controlled trial. Nutr Res. 73:15–26. 2020. View Article : Google Scholar

378 

Yang M, Xue J, Li F, Dong L and Fu Q: Effect of Chinese medicine starvation-free fasting therapy on glucolipid metabolism and liver fat deposition in patients with nonalcoholic fatty liver disease. China Med Herald. 17:135–138. 2020.In Chinese.

379 

Yang R, Wan L, Zhu H and Peng Y: The effect of 12 week-maximum fat oxidation intensity (FATmax) exercise on microvascular function in obese patients with nonalcoholic fatty liver disease and its mechanism. Gen Physiol Biophys. 42:251–262. 2023. View Article : Google Scholar : PubMed/NCBI

380 

Li H, Sun P, Chen Y and Jia H: Research on the intervention of NAFLD by Baduanjin. J Chengdu Sport Univ. 44:79–83. 902018.In Chinese.

381 

Vahedi H, Atefi M, Entezari MH and Hassanzadeh A: The effect of sesame oil consumption compared to sunflower oil on lipid profile, blood pressure, and anthropometric indices in women with non-alcoholic fatty liver disease: A randomized double-blind controlled trial. Trials. 23:5512022. View Article : Google Scholar : PubMed/NCBI

382 

Guo W, Li Z, Anagnostopoulos G, Kong WT, Zhang S, Chakarov S, Shin A, Qian J, Zhu Y, Bai W, et al: Notch signaling regulates macrophage-mediated inflammation in metabolic dysfunction-associated steatotic liver disease. Immunity. 57:2310–2327.e6. 2024. View Article : Google Scholar : PubMed/NCBI

383 

Palomer X, Wang JR, Escalona C, Wu S, Wahli W and Vázquez-Carrera M: Targeting AMPK as a potential treatment for hepatic fibrosis in MASLD. Trends Pharmacol Sci. 46:551–566. 2025. View Article : Google Scholar : PubMed/NCBI

384 

Nóvoa E, da Silva Lima N, Gonzalez-Rellan MJ, Chantada-Vazquez MDP, Verheij J, Rodriguez A, Esquinas-Roman EM, Fondevila MF, Koning M, Fernandez U, et al: Mitochondrial antiviral signaling protein enhances MASLD progression through the ERK/TNFα/NFκβ pathway. Hepatology. 81:1535–1552. 2025. View Article : Google Scholar

385 

Yu Y, Wang Q, Huang X and Li Z: GA receptor targeted chitosan oligosaccharide polymer nanoparticles improve non-alcoholic fatty liver disease by inhibiting ferroptosis. Int J Biol Macromol. 278:1347792024. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Ma Q, Liu K, Chang C, Wang L, Shen Z, Li J, Adu M, Lin Q, Huang H, Wu X, Wu X, et al: Metabolic dysfunction‑associated steatotic liver disease: Pathogenesis, model and treatment (Review). Int J Mol Med 56: 227, 2025.
APA
Ma, Q., Liu, K., Chang, C., Wang, L., Shen, Z., Li, J. ... Wei, R. (2025). Metabolic dysfunction‑associated steatotic liver disease: Pathogenesis, model and treatment (Review). International Journal of Molecular Medicine, 56, 227. https://doi.org/10.3892/ijmm.2025.5668
MLA
Ma, Q., Liu, K., Chang, C., Wang, L., Shen, Z., Li, J., Adu, M., Lin, Q., Huang, H., Wu, X., Wei, R."Metabolic dysfunction‑associated steatotic liver disease: Pathogenesis, model and treatment (Review)". International Journal of Molecular Medicine 56.6 (2025): 227.
Chicago
Ma, Q., Liu, K., Chang, C., Wang, L., Shen, Z., Li, J., Adu, M., Lin, Q., Huang, H., Wu, X., Wei, R."Metabolic dysfunction‑associated steatotic liver disease: Pathogenesis, model and treatment (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 227. https://doi.org/10.3892/ijmm.2025.5668
Copy and paste a formatted citation
x
Spandidos Publications style
Ma Q, Liu K, Chang C, Wang L, Shen Z, Li J, Adu M, Lin Q, Huang H, Wu X, Wu X, et al: Metabolic dysfunction‑associated steatotic liver disease: Pathogenesis, model and treatment (Review). Int J Mol Med 56: 227, 2025.
APA
Ma, Q., Liu, K., Chang, C., Wang, L., Shen, Z., Li, J. ... Wei, R. (2025). Metabolic dysfunction‑associated steatotic liver disease: Pathogenesis, model and treatment (Review). International Journal of Molecular Medicine, 56, 227. https://doi.org/10.3892/ijmm.2025.5668
MLA
Ma, Q., Liu, K., Chang, C., Wang, L., Shen, Z., Li, J., Adu, M., Lin, Q., Huang, H., Wu, X., Wei, R."Metabolic dysfunction‑associated steatotic liver disease: Pathogenesis, model and treatment (Review)". International Journal of Molecular Medicine 56.6 (2025): 227.
Chicago
Ma, Q., Liu, K., Chang, C., Wang, L., Shen, Z., Li, J., Adu, M., Lin, Q., Huang, H., Wu, X., Wei, R."Metabolic dysfunction‑associated steatotic liver disease: Pathogenesis, model and treatment (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 227. https://doi.org/10.3892/ijmm.2025.5668
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team