Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (666)

Search Parameters:
Keywords = stellate cell

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
25 pages, 2693 KiB  
Article
Adipokine and Hepatokines in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): Current and Developing Trends
by Salvatore Pezzino, Stefano Puleo, Tonia Luca, Mariacarla Castorina and Sergio Castorina
Biomedicines 2025, 13(8), 1854; https://doi.org/10.3390/biomedicines13081854 - 30 Jul 2025
Viewed by 309
Abstract
Background/Objectives: Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a major global health challenge characterized by complex adipose–liver interactions mediated by adipokines and hepatokines. Despite rapid field evolution, a comprehensive understanding of research trends and translational advances remains fragmented. This study systematically maps the [...] Read more.
Background/Objectives: Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a major global health challenge characterized by complex adipose–liver interactions mediated by adipokines and hepatokines. Despite rapid field evolution, a comprehensive understanding of research trends and translational advances remains fragmented. This study systematically maps the scientific landscape through bibliometric analysis, identifying emerging domains and future clinical translation directions. Methods: A comprehensive bibliometric analysis of 1002 publications from 2004 to 2025 was performed using thematic mapping, temporal trend evaluation, and network analysis. Analysis included geographical and institutional distributions, thematic cluster identification, and research paradigm evolution assessment, focusing specifically on adipokine–hepatokine signaling mechanisms and clinical implications. Results: The United States and China are at the forefront of research output, whereas European institutions significantly contribute to mechanistic discoveries. The thematic map analysis reveals the motor/basic themes residing at the heart of the field, such as insulin resistance, fatty liver, metabolic syndrome, steatosis, fetuin-A, and other related factors that drive innovation. Basic clusters include metabolic foundations (obesity, adipose tissue, FGF21) and adipokine-centered subjects (adiponectin, leptin, NASH). New themes focus on inflammation, oxidative stress, gut microbiota, lipid metabolism, and hepatic stellate cells. Niche areas show targeted fronts such as exercise therapies, pediatric/novel adipokines (chemerin, vaspin, omentin-1), and advanced molecular processes that focus on AMPK and endoplasmic-reticulum stress. Temporal analysis shows a shift from single liver studies to whole models that include the gut microbiota, mitochondrial dysfunction, and interactions between other metabolic systems. The network analysis identifies nine major clusters: cardiovascular–metabolic links, adipokine–inflammatory pathways, hepatokine control, and new therapeutic domains such as microbiome interventions and cellular stress responses. Conclusions: In summary, this study delineates current trends and emerging areas within the field and elucidates connections between mechanistic research and clinical translation to provide guidance for future research and development in this rapidly evolving area. Full article
(This article belongs to the Special Issue Advances in Hepatology)
Show Figures

Figure 1

26 pages, 2576 KiB  
Review
Exploring Cirrhosis: Insights into Advances in Therapeutic Strategies
by Magdalena Wiacek, Anna Adam, Rafał Studnicki and Igor Z. Zubrzycki
Int. J. Mol. Sci. 2025, 26(15), 7226; https://doi.org/10.3390/ijms26157226 - 25 Jul 2025
Viewed by 189
Abstract
Cirrhosis remains a significant global health burden, responsible for nearly 4% of annual deaths worldwide. Despite progress in antiviral therapies and public health measures, its prevalence has plateaued, particularly in regions affected by viral hepatitis, alcohol misuse, and metabolic syndrome. This review presents [...] Read more.
Cirrhosis remains a significant global health burden, responsible for nearly 4% of annual deaths worldwide. Despite progress in antiviral therapies and public health measures, its prevalence has plateaued, particularly in regions affected by viral hepatitis, alcohol misuse, and metabolic syndrome. This review presents a comprehensive synthesis of the multifactorial drivers of cirrhosis, including hepatocyte injury, liver stellate cell activation, and immune-mediated inflammation. The emphasis is on the central role of metabolic dysfunction, characterized by mitochondrial impairment, altered lipid and glucose metabolism, hormonal imbalance, and systemic inflammation, in exacerbating disease progression. While current therapies may slow the progression of early-stage disease, they are very often ineffective in reversing established fibrosis. Emerging molecular strategies offer promising alternatives by targeting key pathogenic pathways. These include AMPK activators (e.g., metformin, AICAR), FGF21 analogs, and mitochondria-targeted agents (e.g., MitoQ, urolithin A, NAD+ precursors) to restore bioenergetic balance and reduce oxidative stress. Other approaches, such as mesenchymal stem cell therapy, inflammasome inhibition, and hormonal modulation, aim to suppress fibrogenesis and restore liver homeostasis. The integration of systems biology and multi-omics profiling supports patient stratification and precision medicine. This review highlights a shift toward mechanism-based interventions that have the potential to alter cirrhosis outcomes and improve patient survival. Full article
(This article belongs to the Special Issue Cirrhosis: From Molecular Mechanisms to Therapeutic Strategies)
Show Figures

Graphical abstract

17 pages, 1229 KiB  
Review
The Role of PAR2 in MASLD Progression and HCC Development
by Pietro Guerra, Patrizia Pontisso and Andrea Martini
Int. J. Mol. Sci. 2025, 26(15), 7076; https://doi.org/10.3390/ijms26157076 - 23 Jul 2025
Viewed by 199
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has recently become the leading cause of chronic liver disease and can progress to hepatocellular carcinoma (HCC) through multiple pathogenic mechanisms. Protease-activated receptor 2 (PAR2) is a G-protein-coupled receptor activated by proteases such as trypsin, tryptase or [...] Read more.
Metabolic dysfunction-associated steatotic liver disease (MASLD) has recently become the leading cause of chronic liver disease and can progress to hepatocellular carcinoma (HCC) through multiple pathogenic mechanisms. Protease-activated receptor 2 (PAR2) is a G-protein-coupled receptor activated by proteases such as trypsin, tryptase or coagulation factors VII and Xa. Recent studies have shown that PAR2 expression is increased in the liver of patients with MASLD or liver fibrosis. Its activation is linked to metabolic dysfunction through several pathways, including SREBP1c activation, AMPK inhibition and Akt-induced insulin resistance. Inhibition of PAR2 has been effective in reducing MASLD progression in different animal models. Notably, PAR2 blockade has also been effective in more advanced stages of the disease by dampening chronic inflammation and fibrogenesis through the inhibition of hepatic stellate cell activation and of TGF-β and SerpinB3 production. PAR2 also plays a role in cancer development, promoting tumour proliferation, angiogenesis and expression of immune checkpoint inhibitors (like PD-L1, CD47 and CD24). Due to its multifaceted involvement in liver disease, PAR2 is emerging as a key therapeutic target in this clinical context. This review aims to summarise current knowledge on PAR2′s role in MASLD and its potential as a therapeutic target. Full article
(This article belongs to the Special Issue Obesity and Cancer Risk: Molecular Mechanisms and Perspectives)
Show Figures

Figure 1

17 pages, 707 KiB  
Review
Liver Regeneration as a Model for Studying Cellular Plasticity in Mammals: The Roles of Hepatocytes and Cholangiocytes
by Andrey Elchaninov, Polina Vishnyakova, Valeria Glinkina, Timur Fatkhudinov and Gennady Sukhikh
Cells 2025, 14(15), 1129; https://doi.org/10.3390/cells14151129 - 22 Jul 2025
Viewed by 228
Abstract
In most countries, liver disease is one of the most common pathologic conditions among the population. In this regard, the development of new methods to treat liver diseases is not possible without understanding the mechanisms of regeneration of this organ. A characteristic reaction [...] Read more.
In most countries, liver disease is one of the most common pathologic conditions among the population. In this regard, the development of new methods to treat liver diseases is not possible without understanding the mechanisms of regeneration of this organ. A characteristic reaction of the liver to certain damaging factors is a pronounced cellular plasticity; this primarily concerns hepatocytes and cholangiocytes. This property is also characteristic of Ito stellate cells and macrophages. In this study, we focus on the plasticity of hepatocytes and cholangiocytes. We consider such manifestations of plasticity as the ability to enter the mitotic cycle, as well as transdifferentiation. The contribution of each type of plasticity to liver regeneration is considered, as well as the molecular mechanisms providing the cellular plasticity of hepatocytes and cholangiocytes. Full article
(This article belongs to the Special Issue Cellular Differentiation in Health and Disease)
Show Figures

Figure 1

16 pages, 4308 KiB  
Article
Single-Cell Transcriptomic Analysis of Different Liver Fibrosis Models: Elucidating Molecular Distinctions and Commonalities
by Guofei Deng, Xiaomei Liang, Yuxi Pan, Yusheng Luo, Zizhen Luo, Shaoxuan He, Shuai Huang, Zhaopeng Chen, Jiancheng Wang and Shuo Fang
Biomedicines 2025, 13(8), 1788; https://doi.org/10.3390/biomedicines13081788 - 22 Jul 2025
Viewed by 321
Abstract
Background: Liver fibrosis, a consequence of various chronic liver diseases, is characterized by excessive accumulation of extracellular matrix (ECM), leading to impaired liver function and potentially progressing to cirrhosis or hepatocellular carcinoma. The molecular mechanisms underlying liver fibrosis are complex and not [...] Read more.
Background: Liver fibrosis, a consequence of various chronic liver diseases, is characterized by excessive accumulation of extracellular matrix (ECM), leading to impaired liver function and potentially progressing to cirrhosis or hepatocellular carcinoma. The molecular mechanisms underlying liver fibrosis are complex and not fully understood. In vivo experiments are essential for studying the molecular mechanisms of the disease. However, the diverse principles behind mouse modeling techniques for liver fibrosis can complicate the elucidation of specific fibrotic mechanisms. Methods: Five distinct liver fibrosis models were utilized: CONTROL, NASH (non-alcoholic steatohepatitis), BDL (bile duct ligation), TAA (thioacetamide), and CCl4 (carbon tetrachloride). Patents for these drugs were reviewed using Patentscope® and Worldwide Espacenet®. ScRNA-seq was performed to analyze and compare the cellular and molecular differences in these models. Results: The analysis revealed that, particularly in the drug-induced fibrosis models, hepatic stellate cells (HSCs), Kupffer cells, and T-cell subsets exhibit distinct regulatory patterns and dynamic remodeling processes across different liver fibrosis models. These findings highlight the heterogeneity of immune responses and extracellular matrix (ECM) remodeling in various models, providing important insights into the complex mechanisms underlying liver fibrosis. Conclusions: The study enhances our understanding of liver fibrosis development and provides valuable insights for selecting the most representative animal models in future research. This comprehensive analysis underscores the importance of model-specific immune responses and ECM remodeling in liver fibrosis. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

16 pages, 2958 KiB  
Article
MK2 Inhibition as a Novel Treatment for Fibrosis in Primary Sclerosing Cholangitis via an IL-22-Dependent Mechanism
by Cody S. Howe and Ellen J. Beswick
Cells 2025, 14(13), 1031; https://doi.org/10.3390/cells14131031 - 5 Jul 2025
Viewed by 419
Abstract
Primary sclerosing cholangitis (PSC) is a chronic liver disease characterized by bile duct inflammation and fibrosis, leading to cirrhosis and liver failure. Current therapies are limited to symptom management, with no approved treatments targeting fibrosis. We have identified the MAP kinase-activated protein kinase [...] Read more.
Primary sclerosing cholangitis (PSC) is a chronic liver disease characterized by bile duct inflammation and fibrosis, leading to cirrhosis and liver failure. Current therapies are limited to symptom management, with no approved treatments targeting fibrosis. We have identified the MAP kinase-activated protein kinase 2 (MK2) pathway as a potential therapeutic target for treating PSC due to its role in promoting inflammatory cytokine production and activation of fibroblasts. Thus, MDR2 knockout mice were treated therapeutically with MK2 inhibitors, which led to significantly reduced hepatic inflammation and fibrosis. Liver enzymes, collagen 1A1, and fibronectin were decreased in serum with MK2 inhibitor treatment. Furthermore, the production of IL-6, TNFα, CXCL5, collagen 1A1, and fibronectin was decreased in liver tissues and liver stellate cells, whereas the production of IL-10, G-CSF, and IL-22 was increased. MDR2KO mice treated with IL-22 also showed improvements in inflammation and fibrosis, along with increased IL-10 and G-CSF production. Taken together, we identified both a direct mechanism of MK2 regulation of fibrotic factors and an indirect cytokine-mediated mechanism whereby the levels of IL-22, IL-10, and G-CSF were increased with MK2 inhibition and contributed to decreased levels of fibrotic factors. These data suggest that the MK2 pathway is a promising treatment target for PSC. Full article
(This article belongs to the Special Issue Fibrosis in Chronic Inflammatory Diseases)
Show Figures

Graphical abstract

12 pages, 7448 KiB  
Article
An Old New Friend: Folliculo-Stellate Cells in Pituitary Neuroendocrine Tumors
by Valeria-Nicoleta Nastase, Iulia Florentina Burcea, Roxana Ioana Dumitriu-Stan, Amalia Raluca Ceausu, Flavia Zara, Catalina Poiana and Marius Raica
Cells 2025, 14(13), 1019; https://doi.org/10.3390/cells14131019 - 3 Jul 2025
Viewed by 398
Abstract
Pituitary neuroendocrine tumors (PitNETs) represent a complex pathology based on numerous incompletely elucidated molecular mechanisms. Beyond tumor cells, analyzing the tumor microenvironment may help identify novel prognostic markers and therapies. A key component of this environment is the folliculo-stellate (FS) cell. We examined [...] Read more.
Pituitary neuroendocrine tumors (PitNETs) represent a complex pathology based on numerous incompletely elucidated molecular mechanisms. Beyond tumor cells, analyzing the tumor microenvironment may help identify novel prognostic markers and therapies. A key component of this environment is the folliculo-stellate (FS) cell. We examined FS cells in 77 PitNETs obtained by transsphenoidal surgery, using glial fibrillary acidic protein (GFAP) as an immunohistochemical marker. Immunohistochemistry for anterior pituitary hormones and transcription factors was performed to accurately classify the tumors. Our study included 19 somatotroph, 16 mammosomatotroph, 5 plurihormonal PIT-1 positive, 7 corticotroph, 14 gonadotroph, 11 unusual plurihormonal, and 5 null cell PitNETs. FS cells were observed in 55 of the cases, distributed isolated, in small groups or diffuse networks. A considerable number of tumors immunopositive for more than one hormone (including associations between GH/PRL, but also unusual combinations like GH/ACTH) also contained FS cells (p < 0.01), suggesting their involvement in tumor lineages differentiation. In 27 tumors, GFAP-positive cells clustered in highly vascularized areas. Additionally, in 11 of these cases a direct interaction between endothelial cells and FS cells was noted, sustaining their potential role in tumor angiogenesis. Given their complexity, FS cells may be crucial for understanding tumorigenesis mechanisms. Full article
Show Figures

Figure 1

23 pages, 8906 KiB  
Article
9-cis-Retinoic Acid Improves Disease Modelling in iPSC-Derived Liver Organoids
by Mina Kazemzadeh Dastjerd, Vincent Merens, Ayla Smout, Rebeca De Wolf, Christophe Chesné, Catherine Verfaillie, Stefaan Verhulst and Leo A. van Grunsven
Cells 2025, 14(13), 983; https://doi.org/10.3390/cells14130983 - 26 Jun 2025
Viewed by 777
Abstract
Liver fibrosis majorly impacts global health, necessitating the development of in vitro models to study disease mechanisms and develop drug therapies. Relevant models should at least include hepatocytes and hepatic stellate cells (HSCs) and ideally use three-dimensional cultures to mimic in vivo conditions. [...] Read more.
Liver fibrosis majorly impacts global health, necessitating the development of in vitro models to study disease mechanisms and develop drug therapies. Relevant models should at least include hepatocytes and hepatic stellate cells (HSCs) and ideally use three-dimensional cultures to mimic in vivo conditions. Induced pluripotent stem cells (iPSCs) allow for patient-specific liver modelling, but current models based on iPSC-derived hepatocytes (iHepatocytes) and HSCs (iHSCs) still lack key functions. We developed organoids of iHepatocytes and iHSCs and compared them to HepaRG and primary HSC organoids. RNA sequencing analysis comparison of these cultures identified a potential role for the transcription factor RXRA in hepatocyte differentiation and HSC quiescence. Treating cells with the RXRA ligand 9-cis-retinoic acid (9CRA) promoted iHepatocyte metabolism and iHSC quiescence. In organoids, 9CRA enhanced fibrotic response to TGF-β and acetaminophen, highlighting its potential for refining iPSC-based liver fibrosis models to more faithfully replicate human drug-induced liver injury and fibrotic conditions. Full article
(This article belongs to the Special Issue Organoids as an Experimental Tool)
Show Figures

Graphical abstract

14 pages, 3481 KiB  
Article
Effect of 3-HBI on Liver Fibrosis via the TGF-β/SMAD2/3 Pathway on the Human Hepatic Stellate Cell Model
by Chavisa Khongpiroon, Watunyoo Buakaew, Paul J. Brindley, Saranyapin Potikanond, Krai Daowtak, Yordhathai Thongsri, Pachuenp Potup and Kanchana Usuwanthim
Int. J. Mol. Sci. 2025, 26(13), 6022; https://doi.org/10.3390/ijms26136022 - 23 Jun 2025
Viewed by 765
Abstract
Liver fibrosis can progress to irreversible cirrhosis if the underlying causes remain, and this can in turn develop into hepatocellular carcinoma (HCC). Despite these adverse outcomes, liver fibrosis can be reversed. Consequently, research has focused on substances that target liver fibrosis to prevent [...] Read more.
Liver fibrosis can progress to irreversible cirrhosis if the underlying causes remain, and this can in turn develop into hepatocellular carcinoma (HCC). Despite these adverse outcomes, liver fibrosis can be reversed. Consequently, research has focused on substances that target liver fibrosis to prevent or reduce its progression. This study deals with the potential anti-fibrotic action of 3-hydroxy-β-ionone (3-HBI), a bioactive compound found in many plants. To assess the putative effects of 3-HBI, pro-inflammatory cytokine production and the expression of genes and proteins associated with the TGF-β/SMAD2/3 pathway were monitored following exposure to 3-HBI. Initially, cells of the human hepatic stellate cell line LX-2 were treated with TGF-β1 to simulate fibrogenesis. Following the exposure of activated LX-2 cells to 3-HBI, the production of pro-fibrotic substances was significantly reduced. Molecular docking studies revealed that 3-HBI exhibited a high binding affinity for key proteins in the TGF-β/SMAD2/3 pathway. Analyses using qRT-PCR and Western blotting revealed that 3-HBI suppressed the expression of TIMP1, MMP2, MMP9, COL1A1, COL4A1, SMAD2, SMAD3, SMAD4, MMP2, and ACTA2. Together, these findings demonstrate that 3-HBI inhibited the activation of LX-2 cells and significantly reduced the proinflammatory responses triggered by TGF-β1. Accordingly, we confirmed the noteworthy potential of 3-HBI as a therapeutic agent to prevent and treat liver fibrosis, effected by its modulation of the TGF-β/SMAD2/3 signaling pathway. Full article
(This article belongs to the Special Issue Molecular Advances in Liver Fibrosis)
Show Figures

Figure 1

13 pages, 1240 KiB  
Article
Insulin Modulates NK Cell Activity in Liver Fibrosis MASH Patients via the STING Pathway
by Johnny Amer, Ahmad Salhab, Amiram Ariel and Rifaat Safadi
Cells 2025, 14(13), 941; https://doi.org/10.3390/cells14130941 - 20 Jun 2025
Viewed by 602
Abstract
Background: The STING (Stimulator of Interferon Genes) pathway plays a vital role in the body’s innate immune defense system, primarily involved in DNA sensing and type I interferon production. While STING is well-established in various immune cells, its role in natural killer (NK) [...] Read more.
Background: The STING (Stimulator of Interferon Genes) pathway plays a vital role in the body’s innate immune defense system, primarily involved in DNA sensing and type I interferon production. While STING is well-established in various immune cells, its role in natural killer (NK) cells, particularly within the context of liver fibrosis, remains inadequately explored. Aim: The current study investigates the relationship between STING expression, NK cell activity, and insulin receptor (IR) signaling in patients with metabolic dysfunction-associated steatohepatitis (MASH). Methods: Peripheral NK cells were isolated from healthy controls and MASH patients with varying stages of liver fibrosis (early: F1/F2; advanced: F3/F4). The expressions of STING, IR, NK cell activation markers (CD107a, NKp46), and NK cell inhibitory markers (LAIR-1, Siglec-7) were assessed using flow cytometry. NK cell cytotoxicity against primary hepatic stellate cells (pHSCs) was evaluated through apoptosis assays. STING agonists (2′3′-cGAMP and DMXAA) were used to stimulate NK cells, and their effects on STING expression, NK cell activation, and cytotoxicity were measured. Additionally, the impact of insulin signaling on STING expression and NK cell function was examined. Results: Our results demonstrate that STING expression in NK cells correlates with disease severity in liver fibrosis. NK cells from MASH patients with advanced fibrosis (F3/F4) showed inhibited STING protein levels that were statistically comparable to healthy NK cells and accompanied by impaired cytotoxicity and decreased IFN-γ production. In contrast, NK cells from early fibrosis (F1/F2) exhibited higher STING expression and better functional activity. STING agonist treatment (2′3′-cGAMP) restored STING expression and enhanced NK cell activity across all fibrosis stages. Furthermore, insulin treatment and combined insulin and 2′3′-cGAMP treatment synergistically upregulated both IR and STING expressions, leading to improved NK cell function and increased cytotoxicity, particularly in advanced fibrosis. Conclusion: Our results highlight the potential of targeting STING and insulin signaling pathways as a therapeutic approach in restoring NK cell function and enhance immune surveillance in liver fibrosis. Full article
(This article belongs to the Special Issue Inflammation in Target Organs)
Show Figures

Graphical abstract

17 pages, 1642 KiB  
Review
Defenestration of Liver Sinusoidal Endothelial Cells: The Trigger of Liver Fibrosis
by Juntao Zhou, Jianqiao Wang, Lijuan Zhang, Chengliang Zhang and Cheng Tian
Pharmaceuticals 2025, 18(6), 893; https://doi.org/10.3390/ph18060893 - 14 Jun 2025
Viewed by 792
Abstract
Liver fibrosis is a common pathological manifestation of various chronic liver diseases, distinguished by the excessive accumulation of the extracellular matrix. If unresolved, liver fibrosis can progress to cirrhosis or hepatocellular carcinoma. Fenestrae are important structures of liver sinusoidal endothelial cells (LSECs) regulating [...] Read more.
Liver fibrosis is a common pathological manifestation of various chronic liver diseases, distinguished by the excessive accumulation of the extracellular matrix. If unresolved, liver fibrosis can progress to cirrhosis or hepatocellular carcinoma. Fenestrae are important structures of liver sinusoidal endothelial cells (LSECs) regulating hepatic substance exchange, immune response and hemodynamics. The loss of this structure is usually accompanied by dysfunction of LSECs, which disrupts normal liver physiology by impairing hepatic substance exchange, compromising liver microcirculation, and activating hepatic stellate cells (HSCs). This cascade of events ultimately contributes to the onset and development of liver fibrosis. Oxidative stress, impairment of the NO signaling pathway, actin–myosin complex remodeling and pathological angiogenesis are considered to be the main mechanisms underlying LSEC defenestration. Recently, research on the treatment of LSEC defenestration has made notable progress, and findings suggest a potential value in the application of anti-fibrotic therapies. This article expounds the important correlation between defenestration of LSECs and liver fibrosis, while also reviews therapeutic agents and approaches targeting this pathological process. Full article
(This article belongs to the Special Issue Pharmacotherapy of Liver Fibrosis and Hepatitis: Recent Advances)
Show Figures

Graphical abstract

12 pages, 1781 KiB  
Article
Improved Translational Relevance of In Vitro Fibrosis Models by Integrating IOX2-Mediated Hypoxia-Mimicking Pathways
by Manuel A. González Hernández, Jennifer Venhorst, Lars Verschuren, Karin Toet, Martien P. M. Caspers, Martine C. Morrison, Beatrice Coornaert, Gerard J. P. van Westen and Roeland Hanemaaijer
Biomedicines 2025, 13(6), 1448; https://doi.org/10.3390/biomedicines13061448 - 12 Jun 2025
Viewed by 440
Abstract
Background/Objectives: Preclinical models of liver fibrosis only partially mimic human disease processes. Particularly, traditional transforming growth factor beta 1 (TGFβ1)-induced hepatic stellate cell (HSC) models lack relevant processes, including hypoxia-induced pathways. Here, the ability of a hypoxia-mimicking compound (IOX2) to more accurately [...] Read more.
Background/Objectives: Preclinical models of liver fibrosis only partially mimic human disease processes. Particularly, traditional transforming growth factor beta 1 (TGFβ1)-induced hepatic stellate cell (HSC) models lack relevant processes, including hypoxia-induced pathways. Here, the ability of a hypoxia-mimicking compound (IOX2) to more accurately reflect the human fibrotic phenotype on a functional level was investigated. Methods: Human primary HSCs were stimulated (TGFβ1 +/− IOX2), and the cell viability and fibrotic phenotype were determined. The latter was assessed as protein levels of fibrosis markers—collagen, TIMP-1, and Fibronectin. Next-generation sequencing (NGS), differential expression analyses (DESeq2), and Ingenuity Pathway Analysis (IPA) were performed for mechanistic evaluation and biological annotation. Results: Stimulation with TGFβ1 + IOX2 significantly increased fibrotic marker levels. Also, fibrosis-related pathways were activated, and hypoxia-related genes and collagen modifications, such as crosslinking, increased dose-dependently. Comparative analysis with human fibrotic DEGs showed improved disease representation in the HSC model in the presence of IOX2. Conclusions: In conclusion, the HSC model better recapitulated liver fibrosis by IOX2 administration. Therefore, hypoxia-mimicking compounds hold promise for enhancing the translational value of in vitro fibrosis models, providing valuable insights in liver fibrosis pathogenesis and potential therapeutic strategies. Full article
(This article belongs to the Special Issue Novel Insights into Liver Metabolism)
Show Figures

Graphical abstract

22 pages, 5650 KiB  
Article
Uridine, a Therapeutic Nucleoside, Exacerbates Alcoholic Liver Disease via SRC Kinase Activation: A Network Toxicology and Molecular Dynamics Perspective
by Zhenyu Liu, Zhihao Wang, Jie Wang, Shiquan Xu and Tong Zhang
Int. J. Mol. Sci. 2025, 26(12), 5473; https://doi.org/10.3390/ijms26125473 - 7 Jun 2025
Viewed by 903
Abstract
This study looked into the underlying mechanisms and causal relationship between alcoholic liver disease (ALD) and the blood metabolite uridine using a variety of analytical methods, such as Mendelian randomization and molecular dynamics simulations. We discovered uridine to be a possible hepatotoxic agent [...] Read more.
This study looked into the underlying mechanisms and causal relationship between alcoholic liver disease (ALD) and the blood metabolite uridine using a variety of analytical methods, such as Mendelian randomization and molecular dynamics simulations. We discovered uridine to be a possible hepatotoxic agent aggravating ALD by using Mendelian randomization (MR) analysis with genome-wide association study (GWAS) data from 1416 ALD cases and 217,376 controls, as well as with 1091 blood metabolites and 309 metabolite concentration ratios as exposure factors. According to network toxicology analysis, uridine interacts with important targets such as SRC, FYN, LYN, ADRB2, and GSK3B. The single-cell RNA sequencing analysis of ALD tissues revealed that SRC was upregulated in hepatocytes and activated hepatic stellate cells. Subsequently, we determined the stable binding between uridine and SRC through molecular docking and molecular dynamics simulation (RMSD = 1.5 ± 0.3 Å, binding energy < −5.0 kcal/mol). These targets were connected to tyrosine kinase activity, metabolic reprogramming, and GPCR signaling by Gene Ontology (GO) and KEGG studies. These findings elucidate uridine’s role in ALD progression via immunometabolic pathways, offering novel therapeutic targets for precision intervention. These findings highlight the necessity of systems biology frameworks in drug safety evaluation, particularly for metabolites with dual therapeutic and toxicological roles. Full article
(This article belongs to the Special Issue Network Pharmacology: An Emerging Field in Drug Discovery)
Show Figures

Figure 1

23 pages, 1229 KiB  
Review
Ventricular Arrhythmias and Myocardial Infarction: Electrophysiological and Neuroimmune Mechanisms
by Meng Zheng, Zhen Zhou, Ke-Qiong Deng, Hanyu Zhang, Ziyue Zeng, Yongkang Zhang, Bo He, Huanhuan Cai and Zhibing Lu
Biomedicines 2025, 13(6), 1290; https://doi.org/10.3390/biomedicines13061290 - 23 May 2025
Cited by 1 | Viewed by 1148
Abstract
Ventricular arrhythmias (VAs) after myocardial infarction (MI) are still one of the most important causes of cardiovascular death, though patients receive timely vascular recanalization and drug treatment. And it requires further exploring the mechanism and new therapeutics of VAs induced by MI. Here, [...] Read more.
Ventricular arrhythmias (VAs) after myocardial infarction (MI) are still one of the most important causes of cardiovascular death, though patients receive timely vascular recanalization and drug treatment. And it requires further exploring the mechanism and new therapeutics of VAs induced by MI. Here, we review the electrophysiological and neuroimmune mechanisms of VAs induced by MI. Immune cells are regulated by combining with neuroendocrine molecules released by the sympathetic nervous system (SNS), and, in turn, they modulate SNS both at the paraventricular nucleus of the hypothalamus and stellate ganglion by releasing cytokines or chemokines. In addition, ‘life essentials’ such as sleep, physiological health, and exercise can also influence cardiovascular health through neuroimmune mechanisms. Those factors and mechanisms provide us with new perspectives for understanding the occurrence and maintenance of VAs after MI. Exploring the crosstalk between electrophysiology and neuroimmunology will contribute to finding new therapeutics for VAs after MI. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

20 pages, 1742 KiB  
Review
Developments and Applications of Liver-on-a-Chip Technology—Current Status and Future Prospects
by Joseph Mugaanyi, Jing Huang, Jiongze Fang, Arthur Musinguzi, Caide Lu and Zaozao Chen
Biomedicines 2025, 13(6), 1272; https://doi.org/10.3390/biomedicines13061272 - 22 May 2025
Viewed by 1412
Abstract
Background/Objectives: Liver-on-a-chip (LiOC) technology is increasingly recognized as a transformative platform for modeling liver biology, disease mechanisms, drug metabolism, and toxicity screening. Traditional two-dimensional (2D) in vitro models lack the complexity needed to replicate the liver’s unique microenvironment. This review aims to [...] Read more.
Background/Objectives: Liver-on-a-chip (LiOC) technology is increasingly recognized as a transformative platform for modeling liver biology, disease mechanisms, drug metabolism, and toxicity screening. Traditional two-dimensional (2D) in vitro models lack the complexity needed to replicate the liver’s unique microenvironment. This review aims to summarize recent advancements in LiOC systems, emphasizing their potential in biomedical research and translational applications. Methods: This narrative review synthesizes findings from key studies on the development and application of LiOC platforms. We explored innovations in material science and bioengineering, including microfluidic design, 3D printing, stem cell– and tissue-derived liver organoid integration, and co-culture strategies. Commercially available LiOC systems and their regulatory relevance were also evaluated. Results: LiOC systems have evolved from simple PDMS-based chips to complex, multicellular constructs incorporating hepatocytes, endothelial cells, Kupffer cells, and hepatic stellate cells. Recent studies demonstrate their superior ability to replicate liver-specific architecture and functions. Applications span cancer research, drug toxicity assessment (e.g., drug-induced liver injury prediction with >85% sensitivity), disease modeling, and regenerative medicine. Several platforms have gained FDA recognition and are in active use for preclinical drug testing. Conclusions: LiOC technology offers a more physiologically relevant alternative to traditional models and holds promise for reducing reliance on animal studies. While challenges remain, such as vascularization and long-term function, ongoing advancements are paving the way toward clinical and pharmaceutical integration. The technology is poised to play a key role in personalized medicine and next-generation therapeutic development. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

Back to TopTop