1
|
Gan L, Jiang Q, Huang D, Wu X, Zhu X, Wang L, Xie W, Huang J, Fan R, Jing Y, Tang G, Li XD, Guo J, Yin S. A natural small molecule alleviates liver fibrosis by targeting apolipoprotein L2. Nat Chem Biol 2025; 21:80-90. [PMID: 39103634 DOI: 10.1038/s41589-024-01704-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 07/16/2024] [Indexed: 08/07/2024]
Abstract
Liver fibrosis is an urgent clinical problem without effective therapies. Here we conducted a high-content screening on a natural Euphorbiaceae diterpenoid library to identify a potent anti-liver fibrosis lead, 12-deoxyphorbol 13-palmitate (DP). Leveraging a photo-affinity labeling approach, apolipoprotein L2 (APOL2), an endoplasmic reticulum (ER)-rich protein, was identified as the direct target of DP. Mechanistically, APOL2 is induced in activated hepatic stellate cells upon transforming growth factor-β1 (TGF-β1) stimulation, which then binds to sarcoplasmic/ER calcium ATPase 2 (SERCA2) to trigger ER stress and elevate its downstream protein kinase R-like ER kinase (PERK)-hairy and enhancer of split 1 (HES1) axis, ultimately promoting liver fibrosis. As a result, targeting APOL2 by DP or ablation of APOL2 significantly impairs APOL2-SERCA2-PERK-HES1 signaling and mitigates fibrosis progression. Our findings not only define APOL2 as a novel therapeutic target for liver fibrosis but also highlight DP as a promising lead for treatment of this symptom.
Collapse
Affiliation(s)
- Lu Gan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Qiwei Jiang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dong Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xueji Wu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinying Zhu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lei Wang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Xie
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jialuo Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Runzhu Fan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yihang Jing
- Greater Bay Biomedical InnoCenter, Shenzhen Bay Laboratory (SZBL), Shenzhen, China
| | - Guihua Tang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiang David Li
- Greater Bay Biomedical InnoCenter, Shenzhen Bay Laboratory (SZBL), Shenzhen, China
- Department of Chemistry, University of Hong Kong, Hong Kong, China
| | - Jianping Guo
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Sheng Yin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
2
|
Bloom PP, Chung RT. The future of clinical trials of gut microbiome therapeutics in cirrhosis. JHEP Rep 2025; 7:101234. [PMID: 39717506 PMCID: PMC11663965 DOI: 10.1016/j.jhepr.2024.101234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 12/25/2024] Open
Abstract
The last two decades have witnessed an explosion of microbiome research, including in hepatology, with studies demonstrating altered microbial composition in liver disease. More recently, efforts have been made to understand the association of microbiome features with clinical outcomes and to develop therapeutics targeting the microbiome. While microbiome therapeutics hold much promise, their unique features pose certain challenges for the design and conduct of clinical trials. Herein, we will briefly review indications for microbiome therapeutics in cirrhosis, currently available microbiome therapeutics, and the biological pathways targeted by these therapies. We will then focus on the best practices and important considerations for clinical trials of gut microbiome therapeutics in cirrhosis.
Collapse
Affiliation(s)
- Patricia P. Bloom
- University of Michigan, Division of Gastroenterology, Ann Arbor, MI, USA
| | - Raymond T. Chung
- Massachusetts General Hospital, Division of Gastroenterology, Boston, MA, USA
| |
Collapse
|
3
|
Antwi MB, Lefere S, Clarisse D, Koorneef L, Heldens A, Onghena L, Decroix K, Fijalkowska D, Thommis J, Hellemans M, Hoorens A, Geerts A, Devisscher L, De Bosscher K. PPARα-ERRα crosstalk mitigates metabolic dysfunction-associated steatotic liver disease progression. Metabolism 2024:156128. [PMID: 39743041 DOI: 10.1016/j.metabol.2024.156128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/12/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD), the most prevalent liver disease worldwide, continues to rise. More effective therapeutic strategies are urgently needed. We investigated how targeting two key nuclear receptors involved in hepatic energy metabolism, peroxisome proliferator-activated receptor alpha (PPARα) and estrogen-related receptor alpha (ERRα), ameliorates MASLD. METHODS The PPARα agonist pemafibrate and/or ERRα inverse agonist C29 were administered in a short- and long-term Western diet plus fructose model, and a diabetic-background streptozotocin-Western diet model (STZ-WD). Liver and adipose tissue morphology, histological samples, serum metabolites, RNA and protein levels were analysed and scanning electron microscopy was performed. In addition, we performed cell-based assays and immunohistochemistry and immunofluorescence stainings with light and super-resolution confocal microscopy of healthy, MASLD and MASH human livers. RESULTS The ligand combinations' efficacy was highlighted by reduced liver steatosis across all mouse models, alongside improvements in body weight, inflammation, and fibrosis in both long-term models. Additionally, tumour formation was prevented in the STZ-WD mice model. Cell-based assays demonstrated that ERRα inhibits PPARα's activity, explaining why ERRα blockage improves inflammatory and lipid metabolism gene profiles and enhances lipid-lowering effects. Complementary RNA sequencing and shotgun proteomics, combined with enrichment analysis, jointly identified downregulated serum amyloid A1/A2 as essential components underlying the combination treatment's effectiveness. MASLD/MASH patient livers showed reduced PPARα and increased ERRα levels supporting disrupted NR crosstalk in the hepatocyte nucleus. CONCLUSION Our study supports that dual nuclear receptor targeting, which simultaneously increases PPARα and diminishes ERRα activity, may represent a viable novel strategy against MASLD. IMPACT AND IMPLICATIONS Our research introduces a novel therapeutic strategy against MASLD by simultaneously increasing PPARα activity while diminishing ERRα activity. With PPARα agonists already tested in phase III clinical trials, ERRα ligands/modulators need further (clinical) development to make our findings applicable to both MASLD patients and physicians.
Collapse
Affiliation(s)
- Milton Boaheng Antwi
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium; Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Hepatology Research Unit, Department Internal Medicine and Pediatrics, Liver Research Center, Ghent University, Belgium; Department for Basic and Applied Medical Sciences, Gut-Liver Immunopharmacology unit, Ghent University, Ghent, Belgium
| | - Sander Lefere
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Hepatology Research Unit, Department Internal Medicine and Pediatrics, Liver Research Center, Ghent University, Belgium
| | - Dorien Clarisse
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Lisa Koorneef
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Anneleen Heldens
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Hepatology Research Unit, Department Internal Medicine and Pediatrics, Liver Research Center, Ghent University, Belgium
| | - Louis Onghena
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Hepatology Research Unit, Department Internal Medicine and Pediatrics, Liver Research Center, Ghent University, Belgium
| | - Kylian Decroix
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Department for Basic and Applied Medical Sciences, Gut-Liver Immunopharmacology unit, Ghent University, Ghent, Belgium
| | - Daria Fijalkowska
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Jonathan Thommis
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Madeleine Hellemans
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Anne Hoorens
- Department of Pathology, Ghent University Hospital, Ghent University, 9000 Ghent, Belgium
| | - Anja Geerts
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Hepatology Research Unit, Department Internal Medicine and Pediatrics, Liver Research Center, Ghent University, Belgium
| | - Lindsey Devisscher
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium; Department for Basic and Applied Medical Sciences, Gut-Liver Immunopharmacology unit, Ghent University, Ghent, Belgium
| | - Karolien De Bosscher
- Translational Nuclear Receptor Research, UGent Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent, Belgium.
| |
Collapse
|
4
|
Ezhilarasan D, Karthikeyan S, Najimi M, Vijayalakshmi P, Bhavani G, Jansi Rani M. Preclinical liver toxicity models: Advantages, limitations and recommendations. Toxicology 2024; 511:154020. [PMID: 39637935 DOI: 10.1016/j.tox.2024.154020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Experimental animal models are crucial for elucidating the pathophysiology of liver injuries and for assessing new hepatoprotective agents. Drugs and chemicals such as acetaminophen, isoniazid, valproic acid, ethanol, carbon tetrachloride (CCl4), dimethylnitrosamine (DMN), and thioacetamide (TAA) are metabolized by the CYP2E1 enzyme, producing hepatotoxic metabolites that lead to both acute and chronic liver injuries. In experimental settings, acetaminophen (centrilobular necrosis), carbamazepine (centrilobular necrosis and inflammation), sodium valproate (necrosis, hydropic degeneration and mild inflammation), methotrexate (sinusoidal congestion and inflammation), and TAA (centrilobular necrosis and inflammation) are commonly used to induce various types of acute liver injuries. Repeated and intermittent low-dose administration of CCl4, TAA, and DMN activates quiescent hepatic stellate cells, transdifferentiating them into myofibroblasts, which results in abnormal extracellular matrix production and fibrosis induction, more rapidly with DMN and CCL4 than TAA (DMN > CCl4 > TAA). Regarding toxicity and mortality, CCl4 is more toxic than DMN and TAA (CCl4 > DMN > TAA). Models used to induce metabolic dysfunction-associated liver disease (MAFLD) vary, but MAFLD's multifactorial nature driven by factors like obesity, fatty liver, dyslipidaemia, type II diabetes, hypertension, and cardiovascular disease makes it challenging to replicate human metabolic dysfunction-associated steatohepatitis accurately. From an experimental point of view, the degree and pattern of liver injury are influenced by various factors, including the type of hepatotoxic agent, exposure duration, route of exposure, dosage, frequency of administration, and the animal model utilized. Therefore, there is a pressing need for standardized protocols and regulatory guidelines to streamline the selection of animal models in preclinical studies.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India.
| | - Sivanesan Karthikeyan
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Paramasivan Vijayalakshmi
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India; Department of Pharmacology, Asan Memorial Dental College and Hospital, Chengalpattu, Tamil Nadu, India
| | - Ganapathy Bhavani
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India; Department of Pharmacology, Meenakshi Ammal Dental College and Hospital, Meenakshi Academy of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Muthukrishnan Jansi Rani
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| |
Collapse
|
5
|
Pinanga YD, Pyo KH, Shin EA, Lee H, Lee EH, Kim W, Kim S, Kim JE, Kim S, Lee JW. Association between hepatocyte TM4SF5 expression and gut microbiome dysbiosis during non-alcoholic fatty liver disease development. Life Sci 2024; 358:123164. [PMID: 39454995 DOI: 10.1016/j.lfs.2024.123164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/24/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
Gut microbiome dysbiosis is involved in non-alcoholic fatty liver disease (NAFLD) development. Hepatic transmembrane 4 L six family member 5 (TM4SF5) overexpression promotes NAFLD. However, how gut microbiota are associated with TM4SF5-mediated NAFLD remains unexplored. We analyzed the gut microbiome using feces from hepatocyte-specific TM4SF5-overexpressing transgenic (Alb-TGTm4sf5-Flag, TG) or Tm4sf5-/- knock-out (KO) mice fed a normal chow diet (NCD), high-fat diet (HFD) for 2 weeks (HFD2W), or methionine-choline-deficient diet (MCD) for 4 weeks to investigate associations among Tm4sf5 expression, diet, and the gut microbiome. TG-NCD mice showed a higher Firmicutes-to-Bacteroidetes (F/B) ratio, with less enrichment of Akkermansia muciniphila and Lactobacillus reuteri. NASH-related microbiomes in feces were more abundant in TG-HFD2w mice than in KO-HFD2w mice. Further, TG-MCD showed a higher F/B ratio than TG-NCD or KO mice, with decreases or increases in microbiomes beneficial or detrimental to the liver, respectively. Such effects in TG-MCD animals were correlated with functional pathways producing short-chain fatty acids (SCFAs). Furthermore, potential functional pathways of the gut microbiome were metabolically parallel to NAFLD features in TG-MCD mice. These results suggest that hepatocyte Tm4sf5 supports gut microbiome dysbiosis and metabolic activity, leading to SCFA production and hepatic inflammation during NAFLD development.
Collapse
Affiliation(s)
- Yangie Dwi Pinanga
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung-Hee Pyo
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun-Ae Shin
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Haesong Lee
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun Hae Lee
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Wonsik Kim
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Soyeon Kim
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji Eon Kim
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Semi Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejon 34141, Republic of Korea
| | - Jung Weon Lee
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
6
|
Taberner-Cortés A, Aguilar-Ballester M, Jiménez-Martí E, Hurtado-Genovés G, Martín-Rodríguez RM, Herrero-Cervera A, Vinué Á, Martín-Vañó S, Martínez-Hervás S, González-Navarro H. Treatment with 1.25% cholesterol enriched diet produces severe fatty liver disease characterized by advanced fibrosis and inflammation and impaired autophagy in mice. J Nutr Biochem 2024; 134:109711. [PMID: 39111707 DOI: 10.1016/j.jnutbio.2024.109711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 07/15/2024] [Accepted: 07/26/2024] [Indexed: 09/06/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is reaching pandemic proportions due to overnutrition. The understanding of advanced stages that recapitulate the human pathology is of great importance to get a better mechanistic insight. We hypothesized that feeding of WT (C57BL) mice with a diet containing a high content of fat (21%), sugar (41.5%) and 1.25% of cholesterol (called from now on high fat, sucrose and cholesterol diet, HFSCD) will reproduce the characteristics of disease severity. Analysis of 16 weeks HFSCD-fed mice demonstrated increased liver weight and plasmatic liver damage markers compared with control diet (CD)-fed mice. HFSCD-fed mice developed greater hepatic triglyceride, cholesterol and NEFA content, inflammation and NAFLD activity score (NAS) indicating an advanced disease. HFSCD-fed mice displayed augmented hepatic total CD3+ T and Th9 lymphocytes, as well as reduced Th2 lymphocytes and CD206 anti-inflammatory macrophages. Moreover, T cells and anti-inflammatory macrophages correlated positively and inversely, respectively, with intrahepatic cholesterol content. Consistently, circulating cytotoxic CD8+ T lymphocytes, Th1, and B cell levels were elevated in HFSCD-fed WT mice. Hepatic and adipose tissue expression analysis demonstrated changes in fibrotic and metabolic genes related with cholesterol, triglycerides, and fatty acid synthesis in HFSCD-fed WT. These mice also exhibited reduced antioxidant capacity and autophagy and elevated ERK signaling pathway activation and CHOP levels. Our results indicate that the feeding with a cholesterol-enriched diet in WT mice produces an advanced NAFLD stage with fibrosis, characterized by deficient autophagy and ER stress along with inflammasome activation partially via ERK pathway activation.
Collapse
Affiliation(s)
| | | | - Elena Jiménez-Martí
- Metabolic Diseases Group, INCLIVA Biomedical Research Institute, Valencia, Spain; Biochemistry and Molecular Biology Department, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Gema Hurtado-Genovés
- Metabolic Diseases Group, INCLIVA Biomedical Research Institute, Valencia, Spain
| | | | | | - Ángela Vinué
- Metabolic Diseases Group, INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Susana Martín-Vañó
- Metabolic Diseases Group, INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Sergio Martínez-Hervás
- Metabolic Diseases Group, INCLIVA Biomedical Research Institute, Valencia, Spain; Endocrinology and Nutrition Department Clinic Hospital and Department of Medicine, University of Valencia, Valencia, Spain; Metabolic Diseases Group, CIBER de Diabetes y Enfermedades Metabólicas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Herminia González-Navarro
- Metabolic Diseases Group, INCLIVA Biomedical Research Institute, Valencia, Spain; Biochemistry and Molecular Biology Department, Faculty of Medicine, University of Valencia, Valencia, Spain; Metabolic Diseases Group, CIBER de Diabetes y Enfermedades Metabólicas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
7
|
Luo AJ, Chang FC, Lin SL. Exploring Angiopoietin-2: Clinical Insights and Experimental Perspectives in Kidney Diseases. Kidney Int Rep 2024; 9:3375-3385. [PMID: 39698365 PMCID: PMC11652073 DOI: 10.1016/j.ekir.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 12/20/2024] Open
Abstract
Angiopoietin-2, an important contributor to angiogenesis and vascular remodeling, is increasingly recognized in kidney research. This review explores clinical insights and experimental perspectives on angiopoietin-2 in kidney diseases. Traditionally seen as an antagonist of the Tie-2, which is a receptor tyrosine kinase of endothelial cells and some hematopoietic stem cells, angiopoietin-2 exerts both proangiogenic and antiangiogenic effects, making it a versatile and context-dependent player in kidney pathophysiology. Elevated circulating angiopoietin-2 levels in clinical scenarios are associated with sepsis and acute kidney injury (AKI), emphasizing its role as a biomarker of disease severity. In diabetic kidney disease, circulating angiopoietin-2 correlates with albuminuria, a crucial indicator of disease progression, and may serve as a treatment target in protecting the endothelium. Angiopoietin-2 is implicated in chronic kidney diseases (CKDs), where its elevated circulating levels correlate with kidney outcomes and cardiovascular complications, suggesting its potential impact on kidney function and overall health. In experimental settings, angiopoietin-2 plays a pivotal role in angiogenesis and lymphangiogenesis, influencing vascular stability and endothelial integrity. The context-dependent agonist and antagonist role of angiopoietin-2 is regulated by a Tie-2 phosphatase, vascular endothelial protein tyrosine phosphatase (VEPTP), further underscoring its complexity. Angiopoietin-2 is also involved in regulating cellular integrity, inflammation, and endothelial permeability, making it a promising therapeutic target for conditions characterized by disrupted endothelial junctions and vascular dysfunction. This review provides a comprehensive overview of the diverse roles of angiopoietin-2 in kidney research, offering insights into potential therapeutic targets and advancements in managing kidney diseases.
Collapse
Affiliation(s)
- An-Jie Luo
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Fan-Chi Chang
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shuei-Liong Lin
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
8
|
Hur S, Jeong H, Kim K, Kim KH, Kim SH, Lee Y, Nam KT. MIST1 regulates endoplasmic reticulum stress-induced hepatic apoptosis as a candidate marker of fatty liver disease progression. Cell Death Dis 2024; 15:805. [PMID: 39516480 PMCID: PMC11549289 DOI: 10.1038/s41419-024-07217-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
The liver regenerates after injury; however, prolonged injury can lead to chronic inflammation, fatty liver disease, fibrosis, and cancer. The mechanism involving the complex pathogenesis of the progression of liver injury to chronic liver disease remains unclear. In this study, we investigated the dynamics of gene expression associated with the progression of liver disease. We analyzed changes in gene expression over time in a mouse model of carbon tetrachloride (CCl4)-induced fibrosis using high-throughput RNA sequencing. Prolonged CCl4-induced liver injury increased the expression levels of genes associated with the unfolded protein response (UPR), which correlated with the duration of injury, with substantial, progressive upregulation of muscle, intestine, and stomach expression 1 (Mist1, bhlha15) in the mouse fibrosis model and other liver-damaged tissues. Knockdown of MIST1 in HepG2 cells decreased tribbles pseudokinase 3 (TRIB3) levels and increased apoptosis, consistent with the patterns detected in Mist1-knockout mice. MIST1 expression was confirmed in liver tissues from patients with metabolic dysfunction-associated steatohepatitis and alcoholic steatohepatitis (MASH) and correlated with disease progression. In conclusion, MIST1 is expressed in hepatocytes in response to damage, suggesting a new indicator of liver disease progression. Our results suggest that MIST1 plays a key role in the regulation of apoptosis and TRIB3 expression contributing to progressive liver disease after injury.
Collapse
Affiliation(s)
- Sumin Hur
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Haengdueng Jeong
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Keunyoung Kim
- Department of Pharmacy, Kangwon National University College of Pharmacy, Chuncheon, Korea
| | - Kwang H Kim
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Hee Kim
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Yura Lee
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Ki Taek Nam
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
9
|
Homanics GE. Exploratory studies of ethanol drinking in the white-tufted marmoset (Callithrix jacchus). Alcohol 2024; 120:99-107. [PMID: 38971210 DOI: 10.1016/j.alcohol.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
The white-tufted marmoset is a small, nonhuman primate that is rapidly gaining popularity as a model organism, especially for neuroscience research. To date, little work in the alcohol research field has utilized the marmoset. As a step toward establishing the marmoset as a research model for alcohol experimentation, a series of exploratory studies were undertaken to characterize ethanol drinking behavior. A voluntary drinking paradigm was established whereby the common marmoset would consume pharmacologically relevant amounts of ethanol. To facilitate ethanol consumption, ethanol was mixed with a marshmallow flavored solution (hereafter called marshmallow juice) to mask the presumed adverse taste of ethanol. Using marshmallow juice flavored solutions, marmosets readily consumed ethanol up to 1 g/kg during 10 min binge-like drinking sessions or up to 5 g/kg during ∼4 h drinking sessions. Consumption of 1.0-1.5 g/kg during a 30 min session resulted in blood ethanol concentrations of 49-73 mg/dl, which are predicted to be pharmacologically relevant. In animals that were stably consuming ethanol in marshmallow juice, gradually reducing the concentration of the marshmallow juice flavoring resulted in markedly reduced ethanol consumption. Lastly, when offered a choice between ethanol in marshmallow juice and marshmallow juice alone, marmosets displayed a very strong preference for the marshmallow juice solution without ethanol. From these studies, it is concluded that marmosets will voluntarily consume ethanol if the taste is masked with a sweet solution such as marshmallow juice. These studies represent the first report of alcohol consumption and preference in the white-tufted marmoset.
Collapse
Affiliation(s)
- Gregg E Homanics
- Departments of Anesthesiology & Perioperative Medicine, Neurobiology, and Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, 3501 Fifth Ave, Pittsburgh, PA 15261, USA.
| |
Collapse
|
10
|
Na S, Fan Y, Chen H, Li L, Li G, Zhang F, Wang R, Yang Y, Shen Z, Peng Z, Wu Y, Zhu Y, Yang Z, Dong G, Ye Q, Yue J. PPAR α affects hepatic lipid homeostasis by perturbing necroptosis signals in the intestinal epithelium. Acta Pharm Sin B 2024; 14:4858-4873. [PMID: 39664413 PMCID: PMC11628832 DOI: 10.1016/j.apsb.2024.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/06/2024] [Accepted: 07/10/2024] [Indexed: 12/13/2024] Open
Abstract
Rapid turnover of the intestinal epithelium is a critical strategy to balance the uptake of nutrients and defend against environmental insults, whereas inappropriate death promotes the spread of inflammation. PPARα is highly expressed in the small intestine and regulates the absorption of dietary lipids. However, as a key mediator of inflammation, the impact of intestinal PPARα signaling on cell death pathways is unknown. Here, we show that Pparα deficiency of intestinal epithelium up-regulates necroptosis signals, disrupts the gut vascular barrier, and promotes LPS translocation into the liver. Intestinal Pparα deficiency drives age-related hepatic steatosis and aggravates hepatic fibrosis induced by a high-fat plus high-sucrose diet (HFHS). PPARα levels correlate with TRIM38 and MLKL in the human ileum. Inhibition of PPARα up-regulates necroptosis signals in the intestinal organoids triggered by TNF-α and LPS stimuli via TRIM38/TRIF and CREB3L3/MLKL pathways. Butyric acid ameliorates hepatic steatosis induced by intestinal Pparα deficiency through the inhibition of necroptosis. Our data suggest that intestinal PPARα is essential for the maintenance of microenvironmental homeostasis and the spread of inflammation via the gut-liver axis.
Collapse
Affiliation(s)
- Shufang Na
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Provincial Clinical Research Center for Natural Polymer Biological Liver, Wuhan 430071, China
| | - Yanjie Fan
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - HongLei Chen
- Department of Pathology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Ling Li
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Provincial Clinical Research Center for Natural Polymer Biological Liver, Wuhan 430071, China
| | - Guolin Li
- Center for Biomedical Aging, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Furong Zhang
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Rongyan Wang
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Yafei Yang
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Zixia Shen
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Zhuang Peng
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Yafei Wu
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Yong Zhu
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Zheqiong Yang
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
| | - Guicheng Dong
- College of Life Science, Inner Mongolia Agricultural University, Hohhot 010011, China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Provincial Clinical Research Center for Natural Polymer Biological Liver, Wuhan 430071, China
| | - Jiang Yue
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan 430060, China
| |
Collapse
|
11
|
Svobodová G, Horní M, Velecká E, Boušová I. Metabolic dysfunction-associated steatotic liver disease-induced changes in the antioxidant system: a review. Arch Toxicol 2024:10.1007/s00204-024-03889-x. [PMID: 39443317 DOI: 10.1007/s00204-024-03889-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a heterogeneous condition characterized by liver steatosis, inflammation, consequent fibrosis, and cirrhosis. Chronic impairment of lipid metabolism is closely related to oxidative stress, leading to cellular lipotoxicity, mitochondrial dysfunction, and endoplasmic reticulum stress. The detrimental effect of oxidative stress is usually accompanied by changes in antioxidant defense mechanisms, with the alterations in antioxidant enzymes expression/activities during MASLD development and progression reported in many clinical and experimental studies. This review will provide a comprehensive overview of the present research on MASLD-induced changes in the catalytic activity and expression of the main antioxidant enzymes (superoxide dismutases, catalase, glutathione peroxidases, glutathione S-transferases, glutathione reductase, NAD(P)H:quinone oxidoreductase) and in the level of non-enzymatic antioxidant glutathione. Furthermore, an overview of the therapeutic effects of vitamin E on antioxidant enzymes during the progression of MASLD will be presented. Generally, at the beginning of MASLD development, the expression/activity of antioxidant enzymes usually increases to protect organisms against the increased production of reactive oxygen species. However, in advanced stage of MASLD, the expression/activity of several antioxidants generally decreases due to damage to hepatic and extrahepatic cells, which further exacerbates the damage. Although the results obtained in patients, in various experimental animal or cell models have been inconsistent, taken together the importance of antioxidant enzymes in MASLD development and progression has been clearly shown.
Collapse
Affiliation(s)
- Gabriela Svobodová
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, 500 05, Hradec Králové, Czech Republic
| | - Martin Horní
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, 500 05, Hradec Králové, Czech Republic
| | - Eva Velecká
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, 500 05, Hradec Králové, Czech Republic
| | - Iva Boušová
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, 500 05, Hradec Králové, Czech Republic.
| |
Collapse
|
12
|
Belka M, Gostyńska-Stawna A, Stawny M, Krajka-Kuźniak V. Activation of Nrf2 and FXR via Natural Compounds in Liver Inflammatory Disease. Int J Mol Sci 2024; 25:11213. [PMID: 39456994 PMCID: PMC11508530 DOI: 10.3390/ijms252011213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Liver inflammation is frequently linked to oxidative stress and dysregulation of bile acid and fatty acid metabolism. This review focuses on the farnesoid X receptor (FXR), a critical regulator of bile acid homeostasis, and its interaction with the nuclear factor erythroid 2-related factor 2 (Nrf2), a key modulator of cellular defense against oxidative stress. The review explores the interplay between FXR and Nrf2 in liver inflammatory diseases, highlighting the potential therapeutic effects of natural FXR agonists. Specifically, compounds such as auraptene, cafestol, curcumin, fargesone A, hesperidin, lycopene, oleanolic acid, resveratrol, rutin, ursolic acid, and withaferin A are reviewed for their ability to modulate both the FXR and Nrf2 pathways. This article discusses their potential to alleviate liver inflammation, oxidative stress, and damage in diseases such as metabolic-associated fatty liver disease (MAFLD), cholestatic liver injury, and viral hepatitis. In addition, we address the molecular mechanisms driving liver inflammation, including oxidative stress, immune responses, and bile acid accumulation, while also summarizing relevant experimental models. This review emphasizes the promising therapeutic potential of targeting both the Nrf2 and FXR pathways using natural compounds, paving the way for future treatments for liver diseases. Finally, the limitations of the clinical application were indicated, and further research directions were proposed.
Collapse
Affiliation(s)
- Marta Belka
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland
| | - Aleksandra Gostyńska-Stawna
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.-S.); (M.S.)
| | - Maciej Stawny
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.-S.); (M.S.)
| | - Violetta Krajka-Kuźniak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
| |
Collapse
|
13
|
Zhao Y, Chen Z, Dong R, Liu Y, Zhang Y, Guo Y, Yu M, Li X, Wang J. Multiomics analysis reveals the potential mechanism of high-fat diet in dextran sulfate sodium-induced colitis mice model. Food Sci Nutr 2024; 12:8309-8323. [PMID: 39479684 PMCID: PMC11521715 DOI: 10.1002/fsn3.4426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 11/02/2024] Open
Abstract
A high-fat diet (HFD) is recognized as an important contributor to inflammatory bowel disease (IBD). However, the precise underlying mechanism of HFD on IBD remains elusive. This study aimed to investigate the potential mechanism by which HFD affects IBD using 16S rRNA-sequencing and RNA-seq technology. Results indicated that HFD-treated mice exhibited notable alternations in the structure and composition of the gut microbiota, with some of these alternations being associated with the pathogenesis of IBD. Analysis of the colon transcriptome revealed 11 hub genes and 7 hub pathways among control, DSS-induced colitis, and HFD + DSS-treated groups. Further analysis explores the relationship between the hub pathways and genes, as well as the hub genes and gut microbiota. Overall, the findings indicate that the impact of HFD on DSS-induced colitis may be linked to intestinal dysbiosis and specific genes such as Abca8b, Ace2, Apoa1, Apoa4, Apoc3, Aspa, Dpp4, Maob, Slc34a2, Slc7a9, and Trpm6. These results provide valuable insights for determining potential therapeutic targets for addressing HFD-induced IBD.
Collapse
Affiliation(s)
- Yuyang Zhao
- Department of GastroenterologyChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| | - Zhimin Chen
- Department of PharmacologyCollege of Basic Medical Sciences, Jilin UniversityChangchunJilinChina
| | - Ruiyi Dong
- College of Physical Education, Hunan Normal UniversityChangshaChina
| | - Yufan Liu
- Department of PharmacologyCollege of Basic Medical Sciences, Jilin UniversityChangchunJilinChina
| | - Yixin Zhang
- Department of PharmacologyCollege of Basic Medical Sciences, Jilin UniversityChangchunJilinChina
| | - Yan Guo
- Department of PharmacologyCollege of Basic Medical Sciences, Jilin UniversityChangchunJilinChina
| | - Meiyi Yu
- Department of PharmacologyCollege of Basic Medical Sciences, Jilin UniversityChangchunJilinChina
| | - Xiang Li
- Department of PharmacologyCollege of Basic Medical Sciences, Jilin UniversityChangchunJilinChina
| | - Jiangbin Wang
- Department of GastroenterologyChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| |
Collapse
|
14
|
Liu T, Wu G, Gudd CLC, Trovato FM, Barbera T, Liu Y, Triantafyllou E, McPhail MJW, Thursz MR, Khamri W. Cis-interaction between CD52 and T cell receptor complex interferes with CD4 + T cell activation in acute decompensation of cirrhosis. EBioMedicine 2024; 108:105336. [PMID: 39276679 PMCID: PMC11418137 DOI: 10.1016/j.ebiom.2024.105336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Immune dysfunction contributes to a high rate of infection in patients with acute decompensation of cirrhosis. CD52 is a glycoprotein prominently expressed in lymphocytes. Immune regulation by CD52 may be involved in adaptive immune dysfunction in cirrhosis. This study aimed to investigate the function of CD52 on CD4+ T cells on the blood of patients with acute decompensation of cirrhosis. METHODS The expression of CD52 in the peripheral blood lymphocytes of 49 patients with cirrhosis was investigated using flow cytometry and transcriptomics. Potential cis-membrane ligands of CD52 were discovered via proximity labelling followed by proteomics. The function of CD52 on antigen-specific activation of CD4+ T cells was examined using flow cytometry in CD52 CRISPR-Cas9 knockout primary T cells. FINDINGS CD52 expression was elevated in CD4+ T cells in acute decompensation of cirrhosis, and this elevation was correlated with increased disease severity and mortality. Components of the T cell receptor complex including TCRβ, CD3γ and CD3ε were identified and validated as cis-membrane ligands of CD52. Knockout of CD52 promoted antigen-specific activation, proliferation, and pro-inflammatory cytokine secretion. INTERPRETATION Membrane bound CD52 demonstrated cis-interaction with the T cell receptor and served as a dynamic regulator of antigen-specific activation of CD4+ T cells. The upregulation of CD52 in the periphery of acute decompensation of cirrhosis hinders the recognition of the T cell receptor by MHC, contributing to impaired T cell function. The development of an alternative anti-CD52 antibody is required to restore T cell function and prevent infections in cirrhosis. FUNDING This study was supported by the NIHR Imperial Biomedical Research Centre, Institute for Translational Medicine and Therapeutics (P74713), Wellcome Trust (218304/Z/19/Z), and Medical Research Council (MR/X009904/1 and MR/R014019/1).
Collapse
Affiliation(s)
- Tong Liu
- Section of Hepatology & Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion & Reproduction, Imperial College London, London, United Kingdom
| | - Gang Wu
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Cathrin L C Gudd
- Section of Hepatology & Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion & Reproduction, Imperial College London, London, United Kingdom
| | - Francesca M Trovato
- Department of Inflammation Biology, Institute of Liver Studies, King's College London, London, United Kingdom
| | - Thomas Barbera
- Section of Hepatology & Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion & Reproduction, Imperial College London, London, United Kingdom
| | - Yan Liu
- Glycosciences Laboratory, Department of Metabolism, Digestion & Reproduction, Imperial College London, London, United Kingdom
| | - Evangelos Triantafyllou
- Section of Hepatology & Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion & Reproduction, Imperial College London, London, United Kingdom
| | - Mark J W McPhail
- Department of Inflammation Biology, Institute of Liver Studies, King's College London, London, United Kingdom
| | - Mark R Thursz
- Section of Hepatology & Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion & Reproduction, Imperial College London, London, United Kingdom
| | - Wafa Khamri
- Section of Hepatology & Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion & Reproduction, Imperial College London, London, United Kingdom.
| |
Collapse
|
15
|
Willett RA, Tryndyak VP, Hughes Hanks JM, Elkins L, Nagumalli SK, Avigan MI, Ross SA, da Costa GG, Beland FA, Rusyn I, Pogribny IP. A preclinical model of severe NASH-like liver injury by chronic administration of a high-fat and high-sucrose diet in mice. Toxicol Appl Pharmacol 2024; 491:117046. [PMID: 39084266 DOI: 10.1016/j.taap.2024.117046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a progressive liver disease, affecting 38% of adults globally. If left untreated, NAFLD may progress to more advanced forms of the disease, including non-alcoholic steatohepatitis (NASH), liver cirrhosis, and fibrosis. Early NAFLD detection is critical to prevent disease progression. Using an obesogenic high-fat and high-sucrose (HF/HS) diet, we characterized the progression of NAFLD in male and female Collaborative Cross CC042 mice after 20-, 40-, and 60-week intervals of chronic HF/HS diet feeding. The incidence and severity of liver steatosis, inflammation, and fibrosis increased in both sexes over time, with male mice progressing to a NASH-like disease state faster than female mice, as indicated by earlier and more pronounced changes in liver steatosis. Histopathological indication of macrovesicular steatosis and gene expression changes of key lipid metabolism genes were found to be elevated in both sexes after 20 weeks of HF/HS diet. Measurement of circulating markers of inflammation (CXCL10 and TNF-α), histopathological analysis of immune cell infiltrates, and gene expression changes in inflammation-related genes indicated significant liver inflammation after 40 and 60 weeks of HF/HS diet exposure in both sexes. Liver fibrosis, as assessed by Picosirius red and Masson's trichrome staining and changes in expression of key fibrosis related genes indicated significant changes after 40 and 60 weeks of HF/HS diet exposure. In conclusion, we present a preclinical animal model of dietary NAFLD progression, which recapitulates human pathophysiological and pathomorphological changes, that could be used to better understand the progression of NAFLD and support development of new therapeutics.
Collapse
Affiliation(s)
- Rose A Willett
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, AR, USA
| | - Volodymyr P Tryndyak
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, AR, USA
| | | | - Lana Elkins
- Toxicologic Pathology Associates, Jefferson, AR, USA
| | - Suresh K Nagumalli
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, AR, USA
| | - Mark I Avigan
- Office of Pharmacovigilance and Epidemiology, FDA-Center for Drug Evaluation and Research, Silver Spring, MD, USA
| | - Sharon A Ross
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | | | - Frederick A Beland
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, AR, USA
| | - Ivan Rusyn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Igor P Pogribny
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, AR, USA.
| |
Collapse
|
16
|
Rangra S, Rana D, Prajapati A, Benival D, Dwivedi P, Mandoli A. Nutritional and microbiota-based therapeutic interventions for alcohol-associated liver disease: From pathogenesis to therapeutic insights. Life Sci 2024; 352:122852. [PMID: 38909682 DOI: 10.1016/j.lfs.2024.122852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 06/06/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024]
Abstract
Alcohol-associated liver disease (ALD) manifests as a consequence of prolonged and excessive alcohol consumption. This disease is closely associated with the interplay between gut health and liver function, which can lead to complex pathophysiological changes in the body. This review offers a comprehensive exploration of ALD's multifaceted nature, with a keen focus on its pathogenesis and the potential of nutritional and microbiota-based therapies. Insights derived from diverse case studies are utilized to shed light on how interventions can rebalance the gut microbiome and enhance liver function in ALD patients. Furthermore, the feasibility of liver transplantation and stem cell therapy as ultimate measures for ALD has been discussed, with acknowledgment of the inherent risks and challenges accompanying them. ALD's complexity underscores the necessity for a thorough understanding of its etiology and progression to devise effective treatments that mitigate its profound impact on an individual's health.
Collapse
Affiliation(s)
- Shagun Rangra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad (NIPER-A), 382355, India
| | - Dhwani Rana
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad (NIPER-A), 382355, India
| | - Arvee Prajapati
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad (NIPER-A), 382355, India
| | - Derajram Benival
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad (NIPER-A), 382355, India
| | - Pradeep Dwivedi
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS) - Jodhpur, 342005, India
| | - Amit Mandoli
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research- Ahmedabad (NIPER-A), 382355, India.
| |
Collapse
|
17
|
Jung JC, Park SY, Kim KD, Shin WY, Lee KY. Evolution of splenomegaly in liver cirrhosis: Simulation using an electronic circuit. Adv Med Sci 2024; 69:377-384. [PMID: 39117272 DOI: 10.1016/j.advms.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 03/19/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
PURPOSE The evolution of splenomegaly in patients with liver cirrhosis remains largely unknown. In this study, we followed the changes in splenic volume and established the natural course of splenomegaly. We developed an electronic circuit that simulated splenoportal circulation and identified the underlying hemodynamic mechanisms. MATERIALS AND METHODS This retrospective observational study included 93 patients with cirrhosis. Splenic volumes were measured in imaging studies at 6-month intervals and normalized by the ratio of each patient's maximum volume during follow-up (%Vmax). An electronic simulation model was constructed using software and realized on a breadboard. RESULTS Overall, the %Vmax increased from 0.77 ± 0.21 to a maximum of 1.00 ± 0.00 (p < 0.001) during a median follow-up of 23 (3-162) months and then decreased to 0.84 ± 0.18 (p < 0.001) during the next 9 (3-132) months. No interventional radiology procedure was performed to improve hepatic fibrosis and portal hypertension. The evolution of %Vmax showed single-peaked symmetry. An electronic simulation model showed that the upslope of the evolution curve was dependent on the increased intrahepatic vascular resistance and portal hypertension, whereas the downslope was dependent on the decreased portosystemic shunt (PSS) resistance. CONCLUSIONS Splenomegaly in cirrhotic patients aggravated over a period of 23 months and then regressed spontaneously to its initial volume. Electronic simulation of splenoportal circulation showed that splenic enlargement was due to the advancement of liver cirrhosis and portal hypertension, whereas its regression was due to the development of a PSS.
Collapse
Affiliation(s)
- Jae Cheol Jung
- Department of Surgery, Inha University School of Medicine, Incheon, Republic of Korea
| | - Shin-Young Park
- Department of Surgery, Inha University School of Medicine, Incheon, Republic of Korea
| | - Kyeong Deok Kim
- Department of Surgery, Inha University School of Medicine, Incheon, Republic of Korea
| | - Woo Young Shin
- Department of Surgery, Inha University School of Medicine, Incheon, Republic of Korea
| | - Keon-Young Lee
- Department of Surgery, Inha University School of Medicine, Incheon, Republic of Korea.
| |
Collapse
|
18
|
Bentanachs R, Miró L, Sánchez RM, Ramírez-Carrasco P, Amat C, Alegret M, Pérez A, Roglans N, Laguna JC. Pemafibrate abrogates SLD in a rat experimental dietary model, inducing a shift in fecal bile acids and microbiota composition. Biomed Pharmacother 2024; 177:117067. [PMID: 38943989 DOI: 10.1016/j.biopha.2024.117067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND AND AIMS Drugs resolving steatotic liver disease (SLD) could prevent the evolution of metabolic dysfunction associated SLD (MASLD) to more aggressive forms but must show not only efficacy, but also a high safety profile. Repurposing of drugs in clinical use, such as pemafibrate and mirabegron, could facilitate the finding of an effective and safe drug-treatment for SLD. APPROACH AND RESULTS The SLD High Fat High Fructose (HFHFr) rat model develops steatosis without the influence of other metabolic disturbances, such as obesity, inflammation, or type 2 diabetes. Further, liver fatty acids are provided, as in human pathology, both from dietary origin and de novo lipid synthesis. We used the HFHFr model to evaluate the efficacy of pemafibrate and mirabegron, alone or in combination, in the resolution of SLD, analyzing zoometric, biochemical, histological, transcriptomic, fecal metabolomic and microbiome data. We provide evidence showing that pemafibrate, but not mirabegron, completely reverted liver steatosis, due to a direct effect on liver PPARα-driven fatty acid catabolism, without changes in total energy consumption, subcutaneous, perigonadal and brown fat, blood lipids and body weight. Moreover, pemafibrate treatment showed a neutral effect on whole-body glucose metabolism, but deeply modified fecal bile acid composition and microbiota. CONCLUSIONS Pemafibrate administration reverts liver steatosis in the HFHFr dietary rat SLD model without altering parameters related to metabolic or organ toxicity. Our results strongly support further clinical research to reposition pemafibrate for the treatment of SLD/MASLD.
Collapse
Affiliation(s)
- Roger Bentanachs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, Barcelona 08028, Spain; Institute of Biomedicine IBUB, University of Barcelona, Barcelona 08028, Spain.
| | - Lluïsa Miró
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, Barcelona 08028, Spain; Institute for Nutrition and Food Safety Research INSA-UB, University of Barcelona, Barcelona 08028, Spain.
| | - Rosa M Sánchez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, Barcelona 08028, Spain; Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid 28029, Spain; Institute of Biomedicine IBUB, University of Barcelona, Barcelona 08028, Spain.
| | - Patricia Ramírez-Carrasco
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, Barcelona 08028, Spain.
| | - Concepció Amat
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, Barcelona 08028, Spain; Institute for Nutrition and Food Safety Research INSA-UB, University of Barcelona, Barcelona 08028, Spain.
| | - Marta Alegret
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, Barcelona 08028, Spain; Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid 28029, Spain; Institute of Biomedicine IBUB, University of Barcelona, Barcelona 08028, Spain.
| | - Anna Pérez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, Barcelona 08028, Spain; Institute for Nutrition and Food Safety Research INSA-UB, University of Barcelona, Barcelona 08028, Spain.
| | - Núria Roglans
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, Barcelona 08028, Spain; Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid 28029, Spain; Institute of Biomedicine IBUB, University of Barcelona, Barcelona 08028, Spain.
| | - Juan C Laguna
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27-31, Barcelona 08028, Spain; Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid 28029, Spain; Institute of Biomedicine IBUB, University of Barcelona, Barcelona 08028, Spain.
| |
Collapse
|
19
|
Guixé-Muntet S, Quesada-Vázquez S, Gracia-Sancho J. Pathophysiology and therapeutic options for cirrhotic portal hypertension. Lancet Gastroenterol Hepatol 2024; 9:646-663. [PMID: 38642564 DOI: 10.1016/s2468-1253(23)00438-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 04/22/2024]
Abstract
Portal hypertension represents the primary non-neoplastic complication of liver cirrhosis and has life-threatening consequences, such as oesophageal variceal bleeding, ascites, and hepatic encephalopathy. Portal hypertension occurs due to increased resistance of the cirrhotic liver vasculature to portal blood flow and is further aggravated by the hyperdynamic circulatory syndrome. Existing knowledge indicates that the profibrogenic phenotype acquired by sinusoidal cells is the initial factor leading to increased hepatic vascular tone and fibrosis, which cause increased vascular resistance and portal hypertension. Data also suggest that the phenotype of hepatic cells could be further impaired due to the altered mechanical properties of the cirrhotic liver itself, creating a deleterious cycle that worsens portal hypertension in the advanced stages of liver disease. In this Review, we discuss recent discoveries in the pathophysiology and treatment of cirrhotic portal hypertension, a condition with few pharmacological treatment options.
Collapse
Affiliation(s)
- Sergi Guixé-Muntet
- Liver Vascular Biology Research Group, IDIBAPS Biomedical Research Institute, CIBEREHD, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Sergio Quesada-Vázquez
- Liver Vascular Biology Research Group, IDIBAPS Biomedical Research Institute, CIBEREHD, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, IDIBAPS Biomedical Research Institute, CIBEREHD, Hospital Clínic de Barcelona, Barcelona, Spain; Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
20
|
Jung W, Asaduddin M, Keum H, Son Y, Yoo D, Kim D, Lee S, Lee DY, Roh J, Park SH, Jon S. Longitudinal Magnetic Resonance Imaging with ROS-Responsive Bilirubin Nanoparticles Enables Monitoring of Nonalcoholic Steatohepatitis Progression to Cirrhosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305830. [PMID: 38459924 DOI: 10.1002/adma.202305830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Despite the vital importance of monitoring the progression of nonalcoholic fatty liver disease (NAFLD) and its progressive form, nonalcoholic steatohepatitis (NASH), an efficient imaging modality that is readily available at hospitals is currently lacking. Here, a new magnetic-resonance-imaging (MRI)-based imaging modality is presented that allows for efficient and longitudinal monitoring of NAFLD and NASH progression. The imaging modality uses manganese-ion (Mn2+)-chelated bilirubin nanoparticles (Mn@BRNPs) as a reactive-oxygen-species (ROS)-responsive MRI imaging probe. Longitudinal T1-weighted MR imaging of NASH model mice is performed after injecting Mn@BRNPs intravenously. The MR signal enhancement in the liver relative to muscle gradually increases up to 8 weeks of NASH progression, but decreases significantly as NASH progresses to the cirrhosis-like stage at weeks 10 and 12. A new dual input pseudo-three-compartment model is developed to provide information on NASH stage with a single MRI scan. It is also demonstrated that the ROS-responsive Mn@BRNPs can be used to monitor the efficacy of potential anti-NASH drugs with conventional MRI. The findings suggest that the ROS-responsive Mn@BRNPs have the potential to serve as an efficient MRI contrast for monitoring NASH progression and its transition to the cirrhosis-like stage.
Collapse
Affiliation(s)
- Wonsik Jung
- Department of Biological Sciences, Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, South Korea
| | - Muhammad Asaduddin
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, South Korea
| | - Hyeongseop Keum
- Department of Biological Sciences, Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, South Korea
| | - Youngju Son
- Department of Biological Sciences, Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, South Korea
| | - Dohyun Yoo
- Department of Biological Sciences, Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, South Korea
| | - Dohyeon Kim
- Department of Biological Sciences, Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, South Korea
| | - Seojung Lee
- Department of Biological Sciences, Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, South Korea
| | - Dong Yun Lee
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Seoul, 05505, South Korea
| | - Jin Roh
- Department of Pathology, Ajou University School of Medicine, 164 Worldcup-ro, Suwon, 16499, South Korea
| | - Sung-Hong Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, South Korea
| | - Sangyong Jon
- Department of Biological Sciences, Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, South Korea
| |
Collapse
|
21
|
Fuster-Martínez I, Català-Senent JF, Hidalgo MR, Roig FJ, Esplugues JV, Apostolova N, García-García F, Blas-García A. Integrated transcriptomic landscape of the effect of anti-steatotic treatments in high-fat diet mouse models of non-alcoholic fatty liver disease. J Pathol 2024; 262:377-389. [PMID: 38180387 DOI: 10.1002/path.6242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 10/20/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
High-fat diet (HFD) mouse models are widely used in research to develop medications to treat non-alcoholic fatty liver disease (NAFLD), as they mimic the steatosis, inflammation, and hepatic fibrosis typically found in this complex human disease. The aims of this study were to identify a complete transcriptomic signature of these mouse models and to characterize the transcriptional impact exerted by different experimental anti-steatotic treatments. For this reason, we conducted a systematic review and meta-analysis of liver transcriptomic studies performed in HFD-fed C57BL/6J mice, comparing them with control mice and HFD-fed mice receiving potential anti-steatotic treatments. Analyzing 21 studies broaching 24 different treatments, we obtained a robust HFD transcriptomic signature that included 2,670 differentially expressed genes and 2,567 modified gene ontology biological processes. Treated HFD mice generally showed a reversion of this HFD signature, although the extent varied depending on the treatment. The biological processes most frequently reversed were those related to lipid metabolism, response to stress, and immune system, whereas processes related to nitrogen compound metabolism were generally not reversed. When comparing this HFD signature with a signature of human NAFLD progression, we identified 62 genes that were common to both; 10 belonged to the group that were reversed by treatments. Altered expression of most of these 10 genes was confirmed in vitro in hepatocytes and hepatic stellate cells exposed to a lipotoxic or a profibrogenic stimulus, respectively. In conclusion, this study provides a vast amount of information about transcriptomic changes induced during the progression and regression of NAFLD and identifies some relevant targets. Our results may help in the assessment of treatment efficacy, the discovery of unmet therapeutic targets, and the search for novel biomarkers. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Isabel Fuster-Martínez
- Departamento de Farmacología, Universitat de València, Valencia, Spain
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia, Spain
| | - José F Català-Senent
- Computational Biomedicine Laboratory, Principe Felipe Research Center, Valencia, Spain
| | - Marta R Hidalgo
- Computational Biomedicine Laboratory, Principe Felipe Research Center, Valencia, Spain
| | - Francisco J Roig
- Computational Biomedicine Laboratory, Principe Felipe Research Center, Valencia, Spain
- Facultad de Ciencias de la Salud, Universidad San Jorge, Campus Universitario Villanueva de Gállego, Zaragoza, Spain
| | - Juan V Esplugues
- Departamento de Farmacología, Universitat de València, Valencia, Spain
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia, Spain
- CIBEREHD (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Madrid, Spain
| | - Nadezda Apostolova
- Departamento de Farmacología, Universitat de València, Valencia, Spain
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia, Spain
- CIBEREHD (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Madrid, Spain
| | | | - Ana Blas-García
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia, Spain
- CIBEREHD (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Madrid, Spain
- Departamento de Fisiología, Universitat de València, Valencia, Spain
| |
Collapse
|
22
|
Montero-Vallejo R, Maya-Miles D, Ampuero J, Martín F, Romero-Gómez M, Gallego-Durán R. Novel insights into metabolic-associated steatotic liver disease preclinical models. Liver Int 2024; 44:644-662. [PMID: 38291855 DOI: 10.1111/liv.15830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/02/2023] [Accepted: 12/18/2023] [Indexed: 02/01/2024]
Abstract
Metabolic-associated steatotic liver disease (MASLD) encompasses a wide spectrum of metabolic conditions associated with an excess of fat accumulation in the liver, ranging from simple hepatic steatosis to cirrhosis and hepatocellular carcinoma. Finding appropriate tools to study its development and progression is essential to address essential unmet therapeutic and staging needs. This review discusses advantages and shortcomings of different dietary, chemical and genetic factors that can be used to mimic this disease and its progression in mice from a hepatic and metabolic point of view. Also, this review will highlight some additional factors and considerations that could have a strong impact on the outcomes of our model to end up providing recommendations and a checklist to facilitate the selection of the appropriate MASLD preclinical model based on clinical aims.
Collapse
Affiliation(s)
- Rocío Montero-Vallejo
- SeLiver Group, Instituto de Biomedicina de Sevilla/CSIC/Hospital Virgen del Rocío, Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Sevilla, Spain
| | - Douglas Maya-Miles
- SeLiver Group, Instituto de Biomedicina de Sevilla/CSIC/Hospital Virgen del Rocío, Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Sevilla, Spain
| | - Javier Ampuero
- SeLiver Group, Instituto de Biomedicina de Sevilla/CSIC/Hospital Virgen del Rocío, Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Sevilla, Spain
- Digestive Diseases Unit, Hospital Universitario Virgen Del Rocío, Sevilla, Spain
| | - Franz Martín
- Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, University Pablo Olavide-University of Seville-CSIC, Seville, Spain
- Biomedical Research Network on Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Romero-Gómez
- SeLiver Group, Instituto de Biomedicina de Sevilla/CSIC/Hospital Virgen del Rocío, Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Sevilla, Spain
- Digestive Diseases Unit, Hospital Universitario Virgen Del Rocío, Sevilla, Spain
| | - Rocío Gallego-Durán
- SeLiver Group, Instituto de Biomedicina de Sevilla/CSIC/Hospital Virgen del Rocío, Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Sevilla, Spain
| |
Collapse
|
23
|
Rodríguez-Agudo R, González-Recio I, Serrano-Maciá M, Bravo M, Petrov P, Blaya D, Herranz JM, Mercado-Gómez M, Rejano-Gordillo CM, Lachiondo-Ortega S, Gil-Pitarch C, Azkargorta M, Van Liempd SM, Martinez-Cruz LA, Simão A, Elortza F, Martín C, Nevzorova YA, Cubero FJ, Delgado TC, Argemi J, Bataller R, Schoonjans K, Banales JM, Castro RE, Sancho-Bru P, Avila MA, Julve J, Jover R, Mabe J, Simon J, Goikoetxea-Usandizaga N, Martínez-Chantar ML. Anti-miR-873-5p improves alcohol-related liver disease by enhancing hepatic deacetylation via SIRT1. JHEP Rep 2024; 6:100918. [PMID: 38192540 PMCID: PMC10772393 DOI: 10.1016/j.jhepr.2023.100918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 01/10/2024] Open
Abstract
Background & Aims Current therapies for the treatment of alcohol-related liver disease (ALD) have proven largely ineffective. Patients relapse and the disease progresses even after liver transplantation. Altered epigenetic mechanisms are characteristic of alcohol metabolism given excessive acetate and NAD depletion and play an important role in liver injury. In this regard, novel therapeutic approaches based on epigenetic modulators are increasingly proposed. MicroRNAs, epigenetic modulators acting at the post-transcriptional level, appear to be promising new targets for the treatment of ALD. Methods MiR-873-5p levels were measured in 23 liver tissue from Patients with ALD, and GNMT levels during ALD were confirmed using expression databases (transcriptome n = 62, proteome n = 68). High-resolution proteomics and metabolomics in mice following the Gao-binge model were used to investigate miR-873-5p expression in ALD. Hepatocytes exposed to 50 mM alcohol for 12 h were used to study toxicity. The effect of anti-miR-873-5p in the treatment outcomes of ALD was investigated. Results The analysis of human and preclinical ALD samples revealed increased expression of miR-873-5p in the liver. Interestingly, there was an inverse correlation with NNMT, suggesting a novel mechanism for NAD depletion and aberrant acetylation during ALD progression. High-resolution proteomics and metabolomics identified miR-873-5p as a key regulator of NAD metabolism and SIRT1 deacetylase activity. Anti-miR-873-5p reduced NNMT activity, fuelled the NAD salvage pathway, restored the acetylome, and modulated the levels of NF-κB and FXR, two known SIRT1 substrates, thereby protecting the liver from apoptotic and inflammatory processes, and improving bile acid homeostasis. Conclusions These data indicate that targeting miR-873-5p, a repressor of GNMT previously associated with NAFLD and acetaminophen-induced liver failure. is a novel and attractive approach to treating alcohol-induced hepatoxicity. Impact and implications The role of miR-873-5p has not been explicitly examined in the progression of ALD, a pathology with no therapeutic options. In this study, inhibiting miR-873-5p exerted hepatoprotective effects against ALD through rescued SIRT1 activity and consequently restored bile acid homeostasis and attenuated the inflammatory response. Targeting hepatic miR-873-5p may represent a novel therapeutic approach for the treatment of ALD.
Collapse
Affiliation(s)
- Rubén Rodríguez-Agudo
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Irene González-Recio
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Marina Serrano-Maciá
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Miren Bravo
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Petar Petrov
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Experimental Hepatology Joint Research Unit, IIS Hospital La Fe and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain
| | - Delia Blaya
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Liver Cell Plasticity and Tissue Repair Lab, Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Jose María Herranz
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Instituto de Investigaciones Sanitarias de Navarra-IdiSNA, Pamplona, Spain
| | - María Mercado-Gómez
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Claudia María Rejano-Gordillo
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Sofía Lachiondo-Ortega
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Clàudia Gil-Pitarch
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Mikel Azkargorta
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Proteomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
| | - Sebastiaan Martijn Van Liempd
- Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Luis Alfonso Martinez-Cruz
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - A.L. Simão
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Félix Elortza
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Proteomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
| | - César Martín
- Biofisika Institute (UPV/EHU, CSIC) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Yulia A. Nevzorova
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT Complutense University School of Medicine Madrid Spain, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Department of Internal Medicine III, University Hospital RWTH Aachen, Germany
| | - Francisco Javier Cubero
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT Complutense University School of Medicine Madrid Spain, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Teresa C. Delgado
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Josepmaria Argemi
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Instituto de Investigaciones Sanitarias de Navarra-IdiSNA, Pamplona, Spain
| | - Ramón Bataller
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Liver Research Center, Pittsburgh, Pennsylvania, USA
| | - Kristina Schoonjans
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jesús M. Banales
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Ikerbasque, San Sebastian, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Rui E. Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Pau Sancho-Bru
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Liver Cell Plasticity and Tissue Repair Lab, Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Matías A. Avila
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
- Hepatology Program, Cima-University of Navarra, Pamplona, Spain
| | - Josep Julve
- Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain
| | - Ramiro Jover
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Experimental Hepatology Joint Research Unit, IIS Hospital La Fe and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain
| | - Jon Mabe
- Business Department, IK4-Tekniker, Eibar, Spain
| | - Jorge Simon
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Naroa Goikoetxea-Usandizaga
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - María L. Martínez-Chantar
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| |
Collapse
|
24
|
Babu S, Ranajit SK, Pattnaik G, Ghosh G, Rath G, Kar B. An Insight into Different Experimental Models used for Hepatoprotective Studies: A Review. Curr Drug Discov Technol 2024; 21:e191223224660. [PMID: 39206705 DOI: 10.2174/0115701638278844231214115102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/13/2023] [Accepted: 11/22/2023] [Indexed: 09/04/2024]
Abstract
Numerous factors, including exposure to harmful substances, drinking too much alcohol, contracting certain hepatitis serotypes, and using specific medicines, contribute to the development of liver illnesses. Lipid peroxidation and other forms of oxidative stress are the main mechanisms by which hepatotoxic substances harm liver cells. Pathological changes in the liver include a rise in the levels of blood serum, a decrease in antioxidant enzymes, as well as the formation of free radical radicals. It is necessary to find pharmaceutical alternatives to treat liver diseases to increase their efficacy and decrease their toxicity. For the development of new therapeutic medications, a greater knowledge of primary mechanisms is required. In order to mimic human liver diseases, animal models are developed. Animal models have been used for several decades to study the pathogenesis of liver disorders and related toxicities. For many years, animal models have been utilized to investigate the pathophysiology of liver illness and associated toxicity. The animal models are created to imitate human hepatic disorders. This review enlisted numerous hepatic damage in vitro and in vivo models using various toxicants, their probable biochemical pathways and numerous metabolic pathways via oxidative stressors, different serum biomarkers enzymes are discussed, which will help to identify the most accurate and suitable model to test any plant preparations to check and evaluate their hepatoprotective properties.
Collapse
Affiliation(s)
- Sucharita Babu
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, 751050, India
| | - Santosh K Ranajit
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, 751050, India
| | - Gurudutta Pattnaik
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, 751050, India
| | - Goutam Ghosh
- School of Pharmaceutical Sciences, Siksha O Anusandhan Deemed to be University, Bhubaneswar, 751030, India
| | - Goutam Rath
- School of Pharmaceutical Sciences, Siksha O Anusandhan Deemed to be University, Bhubaneswar, 751030, India
| | - Biswakanth Kar
- School of Pharmaceutical Sciences, Siksha O Anusandhan Deemed to be University, Bhubaneswar, 751030, India
| |
Collapse
|
25
|
Rastovic U, Bozzano SF, Riva A, Simoni-Nieves A, Harris N, Miquel R, Lackner C, Zen Y, Zamalloa A, Menon K, Heaton N, Chokshi S, Palma E. Human Precision-Cut Liver Slices: A Potential Platform to Study Alcohol-Related Liver Disease. Int J Mol Sci 2023; 25:150. [PMID: 38203321 PMCID: PMC10778645 DOI: 10.3390/ijms25010150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Alcohol-related liver disease (ALD) encompasses a range of pathological conditions that are complex to study at the clinical and preclinical levels. Despite the global burden of ALD, there is a lack of effective treatments, and mortality is high. One of the reasons for the unsuccessful development of novel therapies is that experimental studies are hindered by the challenge of recapitulating this multifactorial disorder in vitro, including the contributions of hepatotoxicity, impaired lipid metabolism, fibrosis and inflammatory cytokine storm, which are critical drivers in the pathogenesis of ALD in patients and primary targets for drug development. Here, we present the unique characteristics of the culture of human precision-cut liver slices (PCLS) to replicate key disease processes in ALD. PCLS were prepared from human liver specimens and treated with ethanol alone or in combination with fatty acids and lipopolysaccharide (FA + LPS) for up to 5 days to induce hepatotoxic, inflammatory and fibrotic events associated with ALD. Alcohol insult induced hepatocyte death which was more pronounced with the addition of FA + LPS. This mixture showed a significant increase in the cytokines conventionally associated with the prototypical inflammatory response observed in severe ALD, and interestingly, alcohol alone exhibited a different effect. Profibrogenic activation was also observed in the slices and investigated in the context of slice preparation. These results support the versatility of this organotypic model to study different pathways involved in alcohol-induced liver damage and ALD progression and highlight the applicability of the PCLS for drug discovery, confirming their relevance as a bridge between preclinical and clinical studies.
Collapse
Affiliation(s)
- Una Rastovic
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK
- Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Sergio Francesco Bozzano
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK
- Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Antonio Riva
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK
- Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Arturo Simoni-Nieves
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK
- Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Nicola Harris
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK
- Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Rosa Miquel
- Institute of Liver Studies, King’s College London, London WC2R 2LS, UK
| | - Carolin Lackner
- Institute of Pathology, Medical University of Graz, 8010 Graz, Austria
| | - Yoh Zen
- Institute of Liver Studies, King’s College London, London WC2R 2LS, UK
| | - Ane Zamalloa
- Institute of Liver Studies, King’s College London, London WC2R 2LS, UK
| | - Krishna Menon
- Institute of Liver Studies, King’s College London, London WC2R 2LS, UK
| | - Nigel Heaton
- Institute of Liver Studies, King’s College London, London WC2R 2LS, UK
| | - Shilpa Chokshi
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK
- Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| | - Elena Palma
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK
- Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| |
Collapse
|
26
|
Rodrigues Albuquerque E, Ratti da Silva G, de Abreu Braga F, Pelegrini Silva E, Sposito Negrini K, Rodrigues Fracasso JA, Pires Guarnier L, Jacomassi E, Ribeiro-Paes JT, da Silva Gomes R, Gasparotto Junior A, Lívero FADR. Bridging the Gap: Exploring the Preclinical Potential of Pereskia grandifolia in Metabolic-Associated Fatty Liver Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2023; 2023:8840427. [PMID: 38026733 PMCID: PMC10653969 DOI: 10.1155/2023/8840427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/02/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a complex condition characterized by steatosis and metabolic disturbances. Risk factors such as diabetes, cigarette smoking, and dyslipidaemia contribute to its development and progression. Effective and safe therapies for MAFLD are urgently needed. Pereskia grandifolia has shown potential as an alternative treatment, but its effectiveness against liver disease remains unexplored. This research aims to determine the hepatoprotective properties of P. grandifolia using a model of MAFLD. The study was carried out through various phases to assess the safety and efficacy of the ethanol-soluble fraction of P. grandifolia. Initially, an in vitro assay was performed to assess cell viability. This was followed by an acute toxicity test conducted in rats to determine the safety profile of the extract. Subsequently, the anti-inflammatory properties of P. grandifolia were examined in macrophages. For the MAFLD study, diabetic Wistar rats were made diabetic and exposed to a high fat diet and cigarette smoke, for 4 weeks. During the last 2 weeks, the rats were orally given either the vehicle (negative control group; C-), P. grandifolia (30, 100, and 300 mg/kg), or insulin in addition to simvastatin. A basal group of rats not exposed to these risk factors was also assessed. Blood samples were collected to measure cholesterol, triglycerides, glucose, ALT, and AST levels. Liver was assessed for lipid and oxidative markers, and liver histopathology was examined. P. grandifolia showed no signs of toxicity. It demonstrated anti-inflammatory effects by inhibiting phagocytosis and macrophage spreading. The MAFLD model induced liver abnormalities, including increased AST, ALT, disrupted lipid profile, oxidative stress, and significant hepatic damage. However, P. grandifolia effectively reversed these changes, highlighting its potential as a therapeutic agent. These findings emphasize the significance of P. grandifolia in mitigating hepatic consequences associated with various risk factors.
Collapse
Affiliation(s)
- Edilson Rodrigues Albuquerque
- Laboratory of Preclinical Research of Natural Products, Post Graduate Program in Animal Science with Emphasis on Bioactive Products, Universidade Paranaense, Umuarama, Brazil
| | - Gustavo Ratti da Silva
- Laboratory of Preclinical Research of Natural Products, Post Graduate Program in Animal Science with Emphasis on Bioactive Products, Universidade Paranaense, Umuarama, Brazil
| | - Fernanda de Abreu Braga
- Laboratory of Preclinical Research of Natural Products, Paranaense University, Umuarama, Brazil
| | - Ester Pelegrini Silva
- Laboratory of Preclinical Research of Natural Products, Paranaense University, Umuarama, Brazil
| | - Karina Sposito Negrini
- Laboratory of Preclinical Research of Natural Products, Paranaense University, Umuarama, Brazil
| | | | - Lucas Pires Guarnier
- Department of Genetic, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ezilda Jacomassi
- Laboratory of Preclinical Research of Natural Products, Post Graduate Program in Medicinal Plants and Phytotherapeutics in Basic Attention, Paranaense University, Umuarama, Brazil
| | | | - Roberto da Silva Gomes
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, USA
| | - Arquimedes Gasparotto Junior
- Laboratory of Cardiovascular Pharmacology, Faculty of Health Sciences, Federal University of Grande Dourados, Dourados, Brazil
| | - Francislaine Aparecida dos Reis Lívero
- Laboratory of Preclinical Research of Natural Products, Paranaense University, Umuarama, Brazil
- Laboratory of Cardiometabolic Pharmacology, Federal University of Paraná, Curitiba, Brazil
| |
Collapse
|
27
|
Hu Y, Hu X, Luo J, Huang J, Sun Y, Li H, Qiao Y, Wu H, Li J, Zhou L, Zheng S. Liver organoid culture methods. Cell Biosci 2023; 13:197. [PMID: 37915043 PMCID: PMC10619312 DOI: 10.1186/s13578-023-01136-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/20/2023] [Indexed: 11/03/2023] Open
Abstract
Organoids, three-dimensional structures cultured in vitro, can recapitulate the microenvironment, complex architecture, and cellular functions of in vivo organs or tissues. In recent decades, liver organoids have been developed rapidly, and their applications in biomedicine, such as drug screening, disease modeling, and regenerative medicine, have been widely recognized. However, the lack of repeatability and consistency, including the lack of standardized culture conditions, has been a major obstacle to the development and clinical application of liver organoids. It is time-consuming for researchers to identify an appropriate medium component scheme, and the usage of some ingredients remains controversial. In this review, we summarized and compared different methods for liver organoid cultivation that have been published in recent years, focusing on controversial medium components and discussing their advantages and drawbacks. We aimed to provide an effective reference for the development and standardization of liver organoid cultivation.
Collapse
Affiliation(s)
- Yiqing Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Xiaoyi Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jia Luo
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jiacheng Huang
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Yaohan Sun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Haoyu Li
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Yinbiao Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Hao Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jianhui Li
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, 310015, China
- The Organ Repair and Regeneration Medicine Institute of Hangzhou, Hangzhou, 310003, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, China.
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, 310015, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, China.
| |
Collapse
|
28
|
Kuang J, Wang J, Li Y, Li M, Zhao M, Ge K, Zheng D, Cheung KCP, Liao B, Wang S, Chen T, Zhang Y, Wang C, Ji G, Chen P, Zhou H, Xie C, Zhao A, Jia W, Zheng X, Jia W. Hyodeoxycholic acid alleviates non-alcoholic fatty liver disease through modulating the gut-liver axis. Cell Metab 2023; 35:1752-1766.e8. [PMID: 37591244 DOI: 10.1016/j.cmet.2023.07.011] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/19/2023] [Accepted: 07/24/2023] [Indexed: 08/19/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is regarded as a pandemic that affects about a quarter of the global population. Recently, host-gut microbiota metabolic interactions have emerged as distinct mechanistic pathways implicated in the development of NAFLD. Here, we report that a group of gut microbiota-modified bile acids (BAs), hyodeoxycholic acid (HDCA) species, are negatively correlated with the presence and severity of NAFLD. HDCA treatment has been shown to alleviate NAFLD in multiple mouse models by inhibiting intestinal farnesoid X receptor (FXR) and upregulating hepatic CYP7B1. Additionally, HDCA significantly increased abundances of probiotic species such as Parabacteroides distasonis, which enhances lipid catabolism through fatty acid-hepatic peroxisome proliferator-activated receptor alpha (PPARα) signaling, which in turn upregulates hepatic FXR. These findings suggest that HDCA has therapeutic potential for treating NAFLD, with a unique mechanism of simultaneously activating hepatic CYP7B1 and PPARα.
Collapse
Affiliation(s)
- Junliang Kuang
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jieyi Wang
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yitao Li
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Mengci Li
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Mingliang Zhao
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Kun Ge
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Dan Zheng
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Kenneth C P Cheung
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Boya Liao
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Shouli Wang
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Tianlu Chen
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yinan Zhang
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Congrong Wang
- Department of Endocrinology & Metabolism, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Peng Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510655, China
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Aihua Zhao
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Weiping Jia
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Xiaojiao Zheng
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Wei Jia
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China.
| |
Collapse
|
29
|
Abstract
Chronic liver diseases such as nonalcoholic fatty liver disease (NAFLD) or viral hepatitis are characterized by persistent inflammation and subsequent liver fibrosis. Liver fibrosis critically determines long-term morbidity (for example, cirrhosis or liver cancer) and mortality in NAFLD and nonalcoholic steatohepatitis (NASH). Inflammation represents the concerted response of various hepatic cell types to hepatocellular death and inflammatory signals, which are related to intrahepatic injury pathways or extrahepatic mediators from the gut-liver axis and the circulation. Single-cell technologies have revealed the heterogeneity of immune cell activation concerning disease states and the spatial organization within the liver, including resident and recruited macrophages, neutrophils as mediators of tissue repair, auto-aggressive features of T cells as well as various innate lymphoid cell and unconventional T cell populations. Inflammatory responses drive the activation of hepatic stellate cells (HSCs), and HSC subsets, in turn, modulate immune mechanisms via chemokines and cytokines or transdifferentiate into matrix-producing myofibroblasts. Current advances in understanding the pathogenesis of inflammation and fibrosis in the liver, mainly focused on NAFLD or NASH owing to the high unmet medical need, have led to the identification of several therapeutic targets. In this Review, we summarize the inflammatory mediators and cells in the diseased liver, fibrogenic pathways and their therapeutic implications.
Collapse
Affiliation(s)
- Linda Hammerich
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
30
|
Mohammed OA. Alogliptin exhibits multifaceted effects in thioacetamide-insulted rats: A novel approach to combating hepatic inflammation and fibrogenesis. Pathol Res Pract 2023; 250:154833. [PMID: 37769397 DOI: 10.1016/j.prp.2023.154833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 09/30/2023]
Abstract
Hepatic fibrosis arising from chronic liver injury is characterized by dysregulated healing, including hepatic stellate cell activation and excessive deposition of extracellular matrix proteins. Administration of the hepatotoxin thioacetamide (TAA) induces liver injury coupled to fibrogenesis in rodents, mimicking aspects of human disease. Alogliptin is a highly selective inhibitor of dipeptidyl peptidase-4 with purported antifibrotic actions. We investigated the protective effects of alogliptin against TAA-mediated hepatic fibrosis in rats. Adult male Sprague-Dawley rats received intraperitoneal injections of TAA (150 mg/kg) twice weekly for 6 weeks to induce liver fibrosis. A subset of rats also received daily oral alogliptin (20 mg/kg). At 6 weeks, liver injury and fibrosis were assessed by histology, hydroxyproline content, serum liver enzymes, inflammatory cytokines, oxidative stress markers, and genes related to inflammation, apoptosis, and fibrosis. TAA elicited necroinflammation, oxidative stress, upregulation of pro-fibrogenic mediators, increased hydroxyproline content, and excessive collagen deposition, indicating hepatic fibrosis. The administration of Alogliptin led to notable enhancements in liver histology, an extension in survival time, a decrease in hydroxyproline levels and the expression of fibrogenic genes, a reduction in inflammatory cytokines and oxidative stress, and mitigation of hepatocellular apoptosis in rats subjected to TAA treatment. Alogliptin displayed potent antifibrotic, antioxidant, and hepatoprotective properties in this model of toxic liver damage, likely by impeding NFκB while enhanced Nrf2 DNA binding activity which together modulate oxidative stress, inflammation, myofibroblast activation, and apoptosis. These results highlight the potential therapeutic value of alogliptin offering hope for improved treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Osama A Mohammed
- Department of Clinical Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| |
Collapse
|
31
|
Huang CJ, Hsu SJ, Hsu YC, Chen LK, Li C, Huang HC, Lee YH. Synthesis, characterization, and biological verification of asialoglycoprotein receptor-targeted lipopolysaccharide-encapsulated PLGA nanoparticles for the establishment of a liver fibrosis animal model. Biomater Sci 2023; 11:6650-6662. [PMID: 37609825 DOI: 10.1039/d3bm01058a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Liver fibrosis is generally preceded by various liver injuries and often leads to chronic liver diseases and even cirrhosis. Therefore, a liver fibrosis animal model is the cornerstone for the development of therapeutic strategies for hepatic diseases. Although administration of hepatotoxic substances and/or bile duct ligation have been widely performed to construct the in vivo model over the last decades, they are seriously hindered by time-consuming protocols, high mortality, and instability, indicating that an effective and safe approach for the induction of liver fibrosis is still urgently needed nowadays. In this study, we have developed asialoglycoprotein receptor (ASGPR)-targeted lipopolysaccharide (LPS)-loaded poly (lactic-co-glycolic acid) (PLGA) nanoparticles named ALPNPs for establishing an animal model of liver fibrosis. The ALPNPs are characterized as having a spherical nanostructure with size of 182.9 ± 8.89 nm and surface charge of -8.3 ± 1.48 mV. An anti-ASGPR antibody bound to the surface of the nanoparticles with a crosslinking efficiency of 95.03% allows ALPNPs to have hepatocyte-binding specificity. In comparison to free LPSs, the ALPNPs can induce higher aspartate aminotransferase and total bilirubin concentrations in plasma, reduce the blood flow rate in the portal system and the kidneys, and increase vascular resistance in the liver, kidneys, and collateral shunting vasculature. Based on histological and RNA-seq analyses, the ALPNPs can provide similar capability on inducing hepatic inflammation and fibrosis compared to free LPS but possess higher liver targetability than the naked drug. In addition, the ALPNPs are less toxic in organs other than the liver in comparison to free LPS, demonstrating that the ALPNPs do not elicit off-target effects in vivo. Given the aforementioned efficacies with other merits such as biocompatibility and drug release controllability provided by PLGA, we anticipate that the developed ALPNPs are highly applicable in establishing animal models of liver fibrosis in pre-clinical studies.
Collapse
Affiliation(s)
- Ching-Ju Huang
- Department of Biomedical Sciences and Engineering, National Central, University, Taoyuan 32001, Taiwan R.O.C.
| | - Shao-Jung Hsu
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan R.O.C.
- School of Medicine, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan R.O.C
| | - Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central, University, Taoyuan 32001, Taiwan R.O.C.
| | - Liang-Kun Chen
- Department of Biomedical Sciences and Engineering, National Central, University, Taoyuan 32001, Taiwan R.O.C.
| | - Chuan Li
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan R.O.C
| | - Hui-Chun Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan R.O.C.
- School of Medicine, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan R.O.C
| | - Yu-Hsiang Lee
- Department of Biomedical Sciences and Engineering, National Central, University, Taoyuan 32001, Taiwan R.O.C.
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan R.O.C
| |
Collapse
|
32
|
Zhang M, Bai X, Du Q, Xu J, Wang D, Chen L, Dong K, Chen Z, Yang J. The Different Mechanisms of Lipid Accumulation in Hepatocytes Induced by Oleic Acid/Palmitic Acid and High-Fat Diet. Molecules 2023; 28:6714. [PMID: 37764494 PMCID: PMC10536454 DOI: 10.3390/molecules28186714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the primary chronic liver disease worldwide, mainly manifested by hepatic steatosis. Hepatic lipids may be derived from dietary intake, plasma free fatty acid (FFA) uptake, or hepatic de novo lipogenesis (DNL). Currently, cellular and animal models of hepatocellular steatosis are widely used to study the pathogenesis of NAFLD and to investigate therapeutic agents. However, whether there are differences between the in vivo and in vitro models of the mechanisms that cause lipid accumulation has not been reported. We used OA/PA-induced NCTC 1469 cells and high-fat-diet-fed C57BL/6J mice to simulate a hepatocyte steatosis model of NAFLD and to detect indicators related to FFA uptake and DNL. In addition, when serological indicators were analysed in the mouse model, it was found that serum FASN levels decreased. The results revealed that, in the cellular model, indicators related to DNL were decreased, FASN enzyme activity was unchanged, and indicators related to FFA uptake were increased, including the high expression of CD36; while, in the animal model, indicators related to both FFA uptake and de novo synthesis were increased, including the high expression of CD36 and the increased protein levels of FASN with enhanced enzyme activity. In addition, after an analysis of the serological indicators in the mouse model, it was found that the serum levels of FASN were reduced. In conclusion, the OA/PA-induced cellular model can be used to study the mechanism of FFA uptake, whereas the high-fat-diet-induced mouse model can be used to study the mechanism of FFA uptake and DNL. Combined treatment with CD36 and FASN may be more effective against NAFLD. FASN in the serum can be used as one of the indicators for the clinical diagnosis of NAFLD.
Collapse
Affiliation(s)
- Miao Zhang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Xue Bai
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Qian Du
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Jiaojiao Xu
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Danqing Wang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Lei Chen
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Keting Dong
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Ziyue Chen
- School of Nursing, Capital Medical University, Beijing 100069, China;
| | - Jianhong Yang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| |
Collapse
|
33
|
Huang M, Wu Z, Jia L, Wang Y, Gao S, Liu Y, Zhang Y, Li J. Bioinformatics and network pharmacology identify promotional effects and potential mechanisms of ethanol on esophageal squamous cell carcinoma and experimental validation. Toxicol Appl Pharmacol 2023; 474:116615. [PMID: 37406968 DOI: 10.1016/j.taap.2023.116615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/28/2023] [Accepted: 07/01/2023] [Indexed: 07/07/2023]
Abstract
Ethanol is an important risk factor for esophageal squamous cell carcinoma (ESCC); however, the molecular mechanisms behind how ethanol promotes ESCC development remain poorly understood. In this study, ethanol-ESCC-associated target genes were constructed and screened using network pharmacology and subjected to Kyoto Encyclopedia of Genes and Genomes (KEGG) and bioinformatics analysis. A mouse ethanol-exposed esophageal cancer model was constructed with 4-nitroquinoline-1-oxide (4-NQO) to assess its survival and tumor lesion status, and the mechanism of ethanol-promoted ESCC lesions was verified by qRT-PCR and Western blotting. The results showed that 126 ethanol-ESCC crossover genes were obtained, which were significantly enriched in the PI3K/AKT signaling pathway. Bioinformatics results showed that the target genes TNF, IL6, IL1β and JUN were highly expressed in esophageal tumor samples and positively correlated with tumor proliferation and apoptosis genes, and the genetic information of these genes was mutated to different degrees. Animal model experiments showed that ethanol decreased the survival rate and aggravated the occurrence of esophageal cancer in mice. qRT-PCR showed that ethanol promoted the expression of TNF, IL6, IL1β and JUN mRNA in mouse esophageal tumor tissues, and Western blotting showed that ethanol promoted p-PI3K and p-AKT protein expression in mouse esophageal tumor tissues. In conclusion, ethanol promotes esophageal carcinogenesis by increasing the expression of TNF, IL6, IL1β and JUN and activating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Ming Huang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Zhongbing Wu
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Lei Jia
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Yu Wang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Shuang Gao
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Ying Liu
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Yushuang Zhang
- The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China.
| | - Jing Li
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China; The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China.
| |
Collapse
|
34
|
Fernández-Varo G, Jiménez W, Cable E, Ginès P, Harris G, Bukofzer S. Partial vasopressin 1a receptor agonism reduces portal hypertension and hyperaldosteronism and induces a powerful diuretic and natriuretic effect in rats with cirrhosis and ascites. Biomed Pharmacother 2023; 165:115116. [PMID: 37418980 DOI: 10.1016/j.biopha.2023.115116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/09/2023] Open
Abstract
The vasopressin system has emerged as a therapeutic focus for lowering portal hypertension and reducing splanchnic vasodilation in patients with refractory ascites. Clinically available vasopressin agonists are limited by preferential selectivity for V1 receptors that also have steep concentration-response curves with potential risks of excess vasoconstriction and/or complete antidiuretic effects. OCE-205 is a novel, selective, partial V1a receptor agonist with mixed agonist/antagonist activity and no V2 receptor activation at therapeutic doses. We carried out two studies assessing the in vivo effects of OCE-205 in different rat models of cirrhosis and ascites. In a carbon tetrachloride rat cirrhosis model, OCE-205 administration produced a marked reduction in portal hypertension and hyperaldosteronism, along with robust diuretic and natriuretic effects. These effects were accompanied by marked decreases in ascites volume, with three of five animals experiencing total mobilization of ascites. There was no evidence of fluid overload or sodium or water retention, confirming OCE-205's lack of V2 receptor activity. In a second, corroborative study using a bile duct ligation rat model of ascites, OCE-205 produced significant decreases in ascites volume and body weight and a significant increase in urine volume versus vehicle. Urine sodium excretion increased significantly after the first administration of OCE-205 relative to vehicle; however, repeat administration over 5 days did not lead to hyponatremia. Thus, in separate in vivo models, the mixed agonist/antagonist OCE-205 demonstrated relevant and expected endpoint findings consistent with its known mechanism of action and in vitro pharmacology without apparent unwanted effects or nonspecific toxicities.
Collapse
Affiliation(s)
- Guillermo Fernández-Varo
- Hospital Clinic Universitari, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Wladimiro Jiménez
- Hospital Clinic Universitari, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Edward Cable
- Ferring Research Institute Inc., 4244 Sorrento Valley Boulevard, San Diego, CA 92121, USA
| | - Pere Ginès
- Hospital Clinic Universitari, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Geoff Harris
- Ocelot Bio, Inc., 12670 High Bluff Drive, San Diego, CA 92130, USA
| | - Stan Bukofzer
- Ocelot Bio, Inc., 12670 High Bluff Drive, San Diego, CA 92130, USA.
| |
Collapse
|
35
|
Goikoetxea-Usandizaga N, Bravo M, Egia-Mendikute L, Abecia L, Serrano-Maciá M, Urdinguio RG, Clos-García M, Rodríguez-Agudo R, Araujo-Legido R, López-Bermudo L, Delgado TC, Lachiondo-Ortega S, González-Recio I, Gil-Pitarch C, Peña-Cearra A, Simón J, Benedé-Ubieto R, Ariño S, Herranz JM, Azkargorta M, Salazar-Bermeo J, Martí N, Varela-Rey M, Falcón-Pérez JM, Lorenzo Ó, Nogueiras R, Elortza F, Nevzorova YA, Cubero FJ, Saura D, Martínez-Cruz LA, Sabio G, Palazón A, Sancho-Bru P, Elguezabal N, Fraga MF, Ávila MA, Bataller R, Marín JJ, Martín F, Martínez-Chantar ML. The outcome of boosting mitochondrial activity in alcohol-associated liver disease is organ-dependent. Hepatology 2023; 78:878-895. [PMID: 36745935 PMCID: PMC10442112 DOI: 10.1097/hep.0000000000000303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 12/23/2022] [Accepted: 12/23/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Alcohol-associated liver disease (ALD) accounts for 70% of liver-related deaths in Europe, with no effective approved therapies. Although mitochondrial dysfunction is one of the earliest manifestations of alcohol-induced injury, restoring mitochondrial activity remains a problematic strategy due to oxidative stress. Here, we identify methylation-controlled J protein (MCJ) as a mediator for ALD progression and hypothesize that targeting MCJ may help in recovering mitochondrial fitness without collateral oxidative damage. APPROACH AND RESULTS C57BL/6 mice [wild-type (Wt)] Mcj knockout and Mcj liver-specific silencing (MCJ-LSS) underwent the NIAAA dietary protocol (Lieber-DeCarli diet containing 5% (vol/vol) ethanol for 10 days, plus a single binge ethanol feeding at day 11). To evaluate the impact of a restored mitochondrial activity in ALD, the liver, gut, and pancreas were characterized, focusing on lipid metabolism, glucose homeostasis, intestinal permeability, and microbiota composition. MCJ, a protein acting as an endogenous negative regulator of mitochondrial respiration, is downregulated in the early stages of ALD and increases with the severity of the disease. Whole-body deficiency of MCJ is detrimental during ALD because it exacerbates the systemic effects of alcohol abuse through altered intestinal permeability, increased endotoxemia, and dysregulation of pancreatic function, which overall worsens liver injury. On the other hand, liver-specific Mcj silencing prevents main ALD hallmarks, that is, mitochondrial dysfunction, steatosis, inflammation, and oxidative stress, as it restores the NAD + /NADH ratio and SIRT1 function, hence preventing de novo lipogenesis and improving lipid oxidation. CONCLUSIONS Improving mitochondrial respiration by liver-specific Mcj silencing might become a novel therapeutic approach for treating ALD.
Collapse
Affiliation(s)
- Naroa Goikoetxea-Usandizaga
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Miren Bravo
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Leire Egia-Mendikute
- Cancer Immunology and Immunotherapy Lab, Centre for Cooperative Research in Biosciences CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Leticia Abecia
- Inflammation and Macrophage Plasticity Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Immunology, Microbiology and Parasitology Department, Medicine and Nursing Faculty, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Marina Serrano-Maciá
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Rocío G. Urdinguio
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), El Entrego, Spain
- Health Research Institute of Asturias (ISPA), Oviedo, Spain
- University Institute of Oncology (IUOPA), University of Oviedo, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERer), Madrid, Spain
| | - Marc Clos-García
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Rubén Rodríguez-Agudo
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Raquel Araujo-Legido
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Sevilla-CSIC, Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERdem), Spain
| | - Lucía López-Bermudo
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Sevilla-CSIC, Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERdem), Spain
| | - Teresa C. Delgado
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Sofía Lachiondo-Ortega
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Irene González-Recio
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Clàudia Gil-Pitarch
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Ainize Peña-Cearra
- Inflammation and Macrophage Plasticity Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Jorge Simón
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Raquel Benedé-Ubieto
- Department of Immunology, Ophthalmology and ENT Complutense University School of Medicine Madrid Spain
- Gregorio Maraóón Health Research Institute, Madrid, Spain
- Department of Genetics, Physiology and Microbiology. Faculty of Biology. Complutense University of Madrid, Madrid, Spain
| | - Silvia Ariño
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Jose M. Herranz
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERdem), Spain
- Instituto de Investigaciones Sanitarias de Navarra-IdiSNA, Pamplona, Spain
- Hepatology Program, Cima-University of Navarra, Navarra, Spain
| | - Mikel Azkargorta
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Proteomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Julio Salazar-Bermeo
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE). Edificio Torregaitán, Universidad Miguel Hernández de Elche (UMH), Elche, Spain
| | - Nuria Martí
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE). Edificio Torregaitán, Universidad Miguel Hernández de Elche (UMH), Elche, Spain
| | - Marta Varela-Rey
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| | - Juan M. Falcón-Pérez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Óscar Lorenzo
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERdem), Spain
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Rubén Nogueiras
- Department of Physiology, Research Centre of Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain
| | - Félix Elortza
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Proteomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Yulia A. Nevzorova
- Department of Immunology, Ophthalmology and ENT Complutense University School of Medicine Madrid Spain
- Gregorio Maraóón Health Research Institute, Madrid, Spain
- Department of Internal Medicine III, University Hospital RWTH Aachen, Germany
| | - Francisco J. Cubero
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT Complutense University School of Medicine Madrid Spain
- Gregorio Maraóón Health Research Institute, Madrid, Spain
| | - Domingo Saura
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE). Edificio Torregaitán, Universidad Miguel Hernández de Elche (UMH), Elche, Spain
| | - Luis Alfonso Martínez-Cruz
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Stress Kinases in Diabetes, Cancer and Biochemistry, Madrid, Spain
| | - Asís Palazón
- Cancer Immunology and Immunotherapy Lab, Centre for Cooperative Research in Biosciences CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Pau Sancho-Bru
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Natalia Elguezabal
- Animal Health Department, NEIKER-BRTA-Instituto Vasco de Investigación y Desarrollo Agrario, Derio, Bizkaia, Spain
| | - Mario F. Fraga
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), El Entrego, Spain
- Health Research Institute of Asturias (ISPA), Oviedo, Spain
- University Institute of Oncology (IUOPA), University of Oviedo, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERer), Madrid, Spain
| | - Matías A. Ávila
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Instituto de Investigaciones Sanitarias de Navarra-IdiSNA, Pamplona, Spain
- Hepatology Program, Cima-University of Navarra, Navarra, Spain
| | - Ramón Bataller
- Division of Gastroenterology and Hepatology, Departments of Medicine and Nutrition, and Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, North Carolina, USA
- Department of Gastroenterology and Hepatology, Division of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - José J.G. Marín
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
- Experimental Hepatology and Drug Targeting (HEVEPHARM), IBSAL, University of Salamanca, Salamanca, Spain
| | - Franz Martín
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Sevilla-CSIC, Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERdem), Spain
| | - María Luz Martínez-Chantar
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, Madrid, Spain
| |
Collapse
|
36
|
Yang S, Xu B, Han Y, Jiang M, Luo T, Wu N, Cao J, Zheng Y, Shen L, Qin W, Shi H, Dong L. TAF15 exacerbates nonalcoholic steatohepatitis progression by regulating lipid metabolism and inflammation via FASN and p65 NF-κB. Liver Int 2023; 43:1920-1936. [PMID: 37183512 DOI: 10.1111/liv.15607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/11/2023] [Accepted: 05/02/2023] [Indexed: 05/16/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic fatty liver disease (NAFLD) consists of a broad spectrum of conditions, and nonalcoholic steatohepatitis (NASH) is the advanced form of NAFLD. TAF15 is a DNA and RNA binding protein and is involved in crucial inflammatory signalling pathways. We aimed to investigate the role of TAF15 in the progression of NASH and the underlying molecular mechanism. METHODS We generated mice with hepatocyte-specific knockdown and overexpression of TAF15 using a specific adeno-associated virus (AAV). NASH models were established by feeding mice high-fat and high-cholesterol diets and methionine- and choline-deficient diets. Cleavage under targets and tagmentation and dual-luciferase reporter assays were performed to investigate the effect of TAF15 on FASN transcription. Coimmunoprecipitation and immunofluorescence assays were conducted to explore the interaction of TAF15 and p65. In vitro coculture systems were established to study the interactions of hepatocytes, macrophages and HSCs. RESULTS TAF15 was significantly increased in the livers of mouse NASH models and primary hepatocyte NASH model. Knockdown of TAF15 inhibited steatosis, inflammation and fibrosis, while overexpression of TAF15 promoted NASH phenotypes. Mechanistically, TAF15 bound directly to the promoter region of FASN to facilitate its expression, thereby promoting steatosis. Moreover, TAF15 interacted with p65 and activated the NF-κB signalling pathway, increasing the secretion of proinflammatory cytokines and triggering M1 macrophage polarization. Treatment with the FASN inhibitor orlistat partially reversed the phenotypes. CONCLUSIONS These results suggested that TAF15 exacerbated NASH progression by regulating lipid metabolism and inflammation via transcriptional activation of FASN and interacting with p65 to activate the NF-κB signalling pathway.
Collapse
Affiliation(s)
- Suzhen Yang
- Department of Digestive Disease and Gastrointestinal Motility Research Room, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Bing Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuying Han
- School of Medicine, Northwest University, Xi'an, China
| | - MingZuo Jiang
- Department of Gastroenterology and Hepatology, the Affiliated Jinling Hospital of Nanjing University Medical School, Nanjing, China
| | - Tingting Luo
- School of Medicine, Northwest University, Xi'an, China
| | - Nan Wu
- School of Medicine, Northwest University, Xi'an, China
| | - Jiayi Cao
- School of Medicine, Northwest University, Xi'an, China
| | - Ying Zheng
- Department of Digestive Disease and Gastrointestinal Motility Research Room, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lin Shen
- Department of Digestive Disease and Gastrointestinal Motility Research Room, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wen Qin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Haitao Shi
- Department of Digestive Disease and Gastrointestinal Motility Research Room, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lei Dong
- Department of Digestive Disease and Gastrointestinal Motility Research Room, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
37
|
Lin CY, Omoscharka E, Liu Y, Cheng K. Establishment of a Rat Model of Alcoholic Liver Fibrosis with Simulated Human Drinking Patterns and Low-Dose Chemical Stimulation. Biomolecules 2023; 13:1293. [PMID: 37759693 PMCID: PMC10526499 DOI: 10.3390/biom13091293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Although alcohol is a well-known causal factor associated with liver diseases, challenges remain in inducing liver fibrosis in experimental rodent models. These challenges include rodents' natural aversion to high concentrations of alcohol, rapid alcohol metabolism, the need for a prolonged duration of alcohol administration, and technical difficulties. Therefore, it is crucial to establish an experimental model that can replicate the features of alcoholic liver fibrosis. The objective of this study was to develop a feasible rat model of alcoholic liver fibrosis that emulates human drinking patterns and combines low-dose chemicals within a relatively short time frame. We successfully developed an 8-week rat model of alcoholic liver fibrosis that mimics chronic and heavy drinking patterns. Rats were fed with a control liquid diet, an alcohol liquid diet, or alcohol liquid diet combined with multiple binges via oral gavage. To accelerate the progression of alcoholic liver fibrosis, we introduced low-dose carbon tetrachloride (CCl4) through intraperitoneal injection. This model allows researchers to efficiently evaluate potential therapeutics in preclinical studies of alcoholic liver fibrosis within a reasonable time frame.
Collapse
Affiliation(s)
- Chien-Yu Lin
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Evanthia Omoscharka
- Department of Pathology, University Health/Truman Medical Center, School of Medicine, University of Missouri-Kansas City, 2301 Holmes Street, Kansas City, MO 64108, USA
| | - Yanli Liu
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| |
Collapse
|
38
|
Semenovich DS, Andrianova NV, Zorova LD, Pevzner IB, Abramicheva PA, Elchaninov AV, Markova OV, Petrukhina AS, Zorov DB, Plotnikov EY. Fibrosis Development Linked to Alterations in Glucose and Energy Metabolism and Prooxidant-Antioxidant Balance in Experimental Models of Liver Injury. Antioxidants (Basel) 2023; 12:1604. [PMID: 37627599 PMCID: PMC10451385 DOI: 10.3390/antiox12081604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/03/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
The development of liver fibrosis is one of the most severe and life-threatening outcomes of chronic liver disease (CLD). For targeted therapy of CLD, it is highly needed to reveal molecular targets for normalizing metabolic processes impaired in damaged liver and associated with fibrosis. In this study, we investigated the morphological and biochemical changes in rat liver models of fibrosis induced by chronic administration of thioacetamide, carbon tetrachloride, bile duct ligation (BDL), and ischemia/reperfusion (I/R), with a specific focus on carbohydrate and energy metabolism. Changes in the levels of substrates and products, as well as enzyme activities of the major glucose metabolic pathways (glycolysis, glucuronidation, and pentose phosphate pathway) were examined in rat liver tissue after injury. We examined key markers of oxidative energy metabolism, such as the activity of the Krebs cycle enzymes, and assessed mitochondrial respiratory activity. In addition, pro- and anti-oxidative status was assessed in fibrotic liver tissue. We found that 6 weeks of exposure to thioacetamide, carbon tetrachloride, BDL or I/R resulted in a decrease in the activity of glycolytic enzymes, retardation of mitochondrial respiration, elevation of glucuronidation, and activation of pentose phosphate pathways, accompanied by a decrease in antioxidant activity and the onset of oxidative stress in rat liver. Resemblance and differences in the changes in the fibrosis models used are described, including energy metabolism alterations and antioxidant status in the used fibrosis models. The least pronounced changes in glucose metabolism and mitochondrial functions in the I/R and thioacetamide models were associated with the least advanced fibrosis. Ultimately, liver fibrosis significantly altered the metabolic profile in liver tissue and the flux of glucose metabolic pathways, which could be the basis for targeted therapy of liver fibrosis in CLD caused by toxic, cholestatic, or I/R liver injury.
Collapse
Affiliation(s)
- Dmitry S. Semenovich
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia
| | - Nadezda V. Andrianova
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia
| | - Ljubava D. Zorova
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia
| | - Irina B. Pevzner
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia
| | - Polina A. Abramicheva
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia
| | - Andrey V. Elchaninov
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
| | - Olga V. Markova
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia
| | - Aleksandra S. Petrukhina
- K.I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, 109472 Moscow, Russia
| | - Dmitry B. Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia
| | - Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia
| |
Collapse
|
39
|
Zheng K, Hao F, Medrano-Garcia S, Chen C, Guo F, Morán-Blanco L, Rodríguez-Perales S, Torres-Ruiz R, Peligros MI, Vaquero J, Bañares R, Gómez Del Moral M, Regueiro JR, Martínez-Naves E, Mohamed MR, Gallego-Durán R, Maya D, Ampuero J, Romero-Gómez M, Gilbert-Ramos A, Guixé-Muntet S, Fernández-Iglesias A, Gracia-Sancho J, Coll M, Graupera I, Ginès P, Ciudin A, Rivera-Esteban J, Pericàs JM, Frutos MD, Ramos Molina B, Herranz JM, Ávila MA, Nevzorova YA, Fernández-Malavé E, Cubero FJ. Neuroblastoma RAS viral oncogene homolog (N-RAS) deficiency aggravates liver injury and fibrosis. Cell Death Dis 2023; 14:514. [PMID: 37563155 PMCID: PMC10415403 DOI: 10.1038/s41419-023-06029-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/18/2023] [Accepted: 07/31/2023] [Indexed: 08/12/2023]
Abstract
Progressive hepatic damage and fibrosis are major features of chronic liver diseases of different etiology, yet the underlying molecular mechanisms remain to be fully defined. N-RAS, a member of the RAS family of small guanine nucleotide-binding proteins also encompassing the highly homologous H-RAS and K-RAS isoforms, was previously reported to modulate cell death and renal fibrosis; however, its role in liver damage and fibrogenesis remains unknown. Here, we approached this question by using N-RAS deficient (N-RAS-/-) mice and two experimental models of liver injury and fibrosis, namely carbon tetrachloride (CCl4) intoxication and bile duct ligation (BDL). In wild-type (N-RAS+/+) mice both hepatotoxic procedures augmented N-RAS expression in the liver. Compared to N-RAS+/+ counterparts, N-RAS-/- mice subjected to either CCl4 or BDL showed exacerbated liver injury and fibrosis, which was associated with enhanced hepatic stellate cell (HSC) activation and leukocyte infiltration in the damaged liver. At the molecular level, after CCl4 or BDL, N-RAS-/- livers exhibited augmented expression of necroptotic death markers along with JNK1/2 hyperactivation. In line with this, N-RAS ablation in a human hepatocytic cell line resulted in enhanced activation of JNK and necroptosis mediators in response to cell death stimuli. Of note, loss of hepatic N-RAS expression was characteristic of chronic liver disease patients with fibrosis. Collectively, our study unveils a novel role for N-RAS as a negative controller of the progression of liver injury and fibrogenesis, by critically downregulating signaling pathways leading to hepatocyte necroptosis. Furthermore, it suggests that N-RAS may be of potential clinical value as prognostic biomarker of progressive fibrotic liver damage, or as a novel therapeutic target for the treatment of chronic liver disease.
Collapse
Affiliation(s)
- Kang Zheng
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of Anesthesiology, Nanjing Pukou District Hospital of Chinese Medicine Central Laboratory affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Fengjie Hao
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Sandra Medrano-Garcia
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Chaobo Chen
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- Department of General Surgery, Wuxi Xishan People's Hospital, Wuxi, China
- Department of General Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Feifei Guo
- Department of Obstetrics and Gynaecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Laura Morán-Blanco
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
| | - Sandra Rodríguez-Perales
- Molecular Cytogenetics and Genome Editing Unit, Human Cancer Genetics Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Raúl Torres-Ruiz
- Molecular Cytogenetics and Genome Editing Unit, Human Cancer Genetics Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - María Isabel Peligros
- Servicio de Anatomía Patológica Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Javier Vaquero
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Rafael Bañares
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Manuel Gómez Del Moral
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of Cell Biology, Complutense University School of Medicine, Madrid, Spain
| | - José R Regueiro
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Eduardo Martínez-Naves
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | | | - Rocío Gallego-Durán
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Douglas Maya
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Javier Ampuero
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Manuel Romero-Gómez
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Albert Gilbert-Ramos
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute, Barcelona, Spain
| | - Sergi Guixé-Muntet
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute, Barcelona, Spain
| | - Anabel Fernández-Iglesias
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute, Barcelona, Spain
| | - Jordi Gracia-Sancho
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute, Barcelona, Spain
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Mar Coll
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Laboratorio de Plasticidad de Células Hepáticas y Reparación de Tejidos, Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Isabel Graupera
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Laboratorio de Plasticidad de Células Hepáticas y Reparación de Tejidos, Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Liver Unit, Hospital Clinic, Barcelona, Spain
| | - Pere Ginès
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Liver Unit, Hospital Clinic, Barcelona, Spain
| | - Andreea Ciudin
- Endocrinology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute for Research (VHIR), Barcelona, Spain
| | - Jesús Rivera-Esteban
- Liver Unit, Internal Medicine Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute for Research (VHIR), Barcelona, Spain
| | - Juan M Pericàs
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Unit, Internal Medicine Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute for Research (VHIR), Barcelona, Spain
| | - María Dolores Frutos
- Department of General and Digestive System Surgery, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - Bruno Ramos Molina
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Laboratorio de Obesidad y Metabolismo, Instituto de Investigación Biomédica de Murcia (IMIB-Arrixaca), Murcia, Spain
| | - José María Herranz
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Hepatology Programme, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Matías A Ávila
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Hepatology Programme, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Yulia A Nevzorova
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Edgar Fernández-Malavé
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain.
| |
Collapse
|
40
|
Cheng Y, Lin S, Ren T, Zhang J, Shi Y, Chen Y, Chen Y. New murine model of alcoholic hepatitis in obesity-induced metabolic-associated fatty liver disease. Exp Anim 2023; 72:389-401. [PMID: 37019681 PMCID: PMC10435355 DOI: 10.1538/expanim.22-0160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) and alcoholic hepatitis (AH) are among the most prevalent liver diseases worldwide, and their coexistence is common in clinical practice. However, currently established models of MAFLD-AH coexistence do not fully replicate their pathological characteristics and require sophisticated experimental techniques. Therefore, we aimed to develop an easily replicable model that mimics obesity-induced MAFLD-AH in patients. Our goal was to establish a murine model that replicates MAFLD and AH coexistence, resulting in significant liver injury and inflammation. To this end, we administered a single ethanol gavage dose to ob/ob mice on a chow diet. The administration of a single dose of ethanol led to elevated serum transaminase levels, increased liver steatosis, and apoptosis in ob/ob mice. Furthermore, ethanol binge caused a significant increase in oxidative stress in ob/ob mice, as measured via 4-hydroxynonenal. Importantly, the single dose of ethanol also markedly exacerbated liver neutrophil infiltration and upregulated the hepatic mRNA expression of several chemokines and neutrophil-related proteins, including Cxcl1, Cxcl2, and Lcn2. Whole-liver transcriptomic analysis revealed that ethanol-induced changes in gene expression profile shared similar features with AH and MAFLD. In ob/ob mice, a single dose of ethanol binge caused significant liver injury and neutrophil infiltration. This easy-to-replicate murine model successfully mimics the pathological and clinical features of patients with coexisting MAFLD and AH and closely resembles the transcriptional regulation seen in human disease.
Collapse
Affiliation(s)
- Yuqing Cheng
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, P.R. China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, 1665 Kongjiang Road, Shanghai, 200092, P.R. China
| | - Shuangzhe Lin
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, P.R. China
| | - Tianyi Ren
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, P.R. China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, 1665 Kongjiang Road, Shanghai, 200092, P.R. China
| | - Jianbin Zhang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, P.R. China
| | - Yingying Shi
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, P.R. China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, 1665 Kongjiang Road, Shanghai, 200092, P.R. China
| | - Yingwei Chen
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, P.R. China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, 1665 Kongjiang Road, Shanghai, 200092, P.R. China
| | - Yuanwen Chen
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, 221 West Yan’an Road, Shanghai, 200040, P.R. China
- Department of Geriatrics, Huadong Hospital Affiliated to Fudan University, 221 West Yan’an Road, Shanghai, 200040, P.R. China
| |
Collapse
|
41
|
Zhang Y, Zhang Y, Chen T, Lin Y, Gong J, Xu Q, Wang J, Li J, Meng Y, Li Y, Li X. Caveolin-1 depletion attenuates hepatic fibrosis via promoting SQSTM1-mediated PFKL degradation in HSCs. Free Radic Biol Med 2023; 204:95-107. [PMID: 37116593 DOI: 10.1016/j.freeradbiomed.2023.04.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 04/30/2023]
Abstract
The key glycolytic enzyme phosphofructokinase (PFK) is responsible for maintaining glycolytic stability and an important energy source for activating hepatic stellate cells (HSCs). However, its regulation in activated HSCs remains unclear. Caveolin-1 (Cav1), a major constituent of caveolae, has emerged as a key target for triggering glycolysis. However, the relationship between Cav1 and glycolysis during HSC activation is not well established. In this study, Cav1 was upregulated in mouse and human fibrotic liver tissues. We concluded that HSC-specific Cav1 knockdown markedly alleviates liver injury and fibrosis. Mechanistically, Cav1 was elevated during primary mouse HSC activation, competing with SQSTM1 for the regulatory subunit of PFK liver type and inhibiting the SQSTM1-mediated autophagy-independent lysosomal degradation pathway to sustain HSC activation. We also identified the heptapeptide alamandine as a promising therapeutic agent that downregulates Cav1 protein levels via proteasomal degradation and may impair glycolysis. Our study provides evidence of the crucial role and mechanism of Cav1 in the glucose metabolic network in HSCs and highlights Cav1 as a critical therapeutic target for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Yijie Zhang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Tingting Chen
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Ying Lin
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Jiacheng Gong
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Qihan Xu
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Jun Wang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Jierui Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Ying Meng
- Department of Respiratory Diseases, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| | - Yang Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| |
Collapse
|
42
|
Krylov D, Rodimova S, Karabut M, Kuznetsova D. Experimental Models for Studying Structural and Functional State of the Pathological Liver (Review). Sovrem Tekhnologii Med 2023; 15:65-82. [PMID: 38434194 PMCID: PMC10902899 DOI: 10.17691/stm2023.15.4.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Indexed: 03/05/2024] Open
Abstract
Liver pathologies remain one of the leading causes of mortality worldwide. Despite a high prevalence of liver diseases, the possibilities of diagnosing, prognosing, and treating non-alcoholic and alcoholic liver diseases still have a number of limitations and require the development of new methods and approaches. In laboratory studies, various models are used to reconstitute the pathological conditions of the liver, including cell cultures, spheroids, organoids, microfluidic systems, tissue slices. We reviewed the most commonly used in vivo, in vitro, and ex vivo models for studying non-alcoholic fatty liver disease and alcoholic liver disease, toxic liver injury, and fibrosis, described their advantages, limitations, and prospects for use. Great emphasis was placed on the mechanisms of development of pathological conditions in each model, as well as the assessment of the possibility of reconstructing various key aspects of pathogenesis for all these pathologies. There is currently no consensus on the choice of the most adequate model for studying liver pathology. The choice of a certain effective research model is determined by the specific purpose and objectives of the experiment.
Collapse
Affiliation(s)
- D.P. Krylov
- Laboratory Assistant, Scientific Laboratory of Molecular Biotechnologies, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Student, Institute of Biology and Biomedicine; National Research Lobachevsky State University of Nizhny Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603022, Russia
| | - S.A. Rodimova
- Junior Researcher, Laboratory of Regenerative Medicine, Scientific Laboratory of Molecular Biotechnologies, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - M.M. Karabut
- Researcher, Laboratory of Genomics of Adaptive Antitumor Immunity, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - D.S. Kuznetsova
- Head of Laboratory of Molecular Biotechnologies, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Head of the Research Laboratory for Molecular Genetic Researches, Institute of Clinical Medicine; National Research Lobachevsky State University of Nizhny Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603022, Russia
| |
Collapse
|
43
|
Gil M, Azkargorta M, Fuster C, Martínez-Gómez M, Raurell I, Barberá A, Pericàs JM, Hide D, Elortza F, Genescà J, Martell M. Proteomic Analysis of Dysfunctional Liver Sinusoidal Endothelial Cells Reveals Substantial Differences in Most Common Experimental Models of Chronic Liver Diseases. Int J Mol Sci 2023; 24:11904. [PMID: 37569282 PMCID: PMC10418749 DOI: 10.3390/ijms241511904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
Molecular markers of dedifferentiation of dysfunctional liver sinusoidal endothelial cells (LSEC) have not been fully elucidated. We aimed at deciphering the molecular profile of dysfunctional LSEC in different pathological scenarios. Flow cytometry was used to sort CD11b-/CD32b+ and CD11b-/CD32b- LSEC from three rat models of liver disease (bile duct ligation-BDL; inhaled carbon tetrachloride-CCl4; and high fat glucose/fructose diet-HFGFD). A full proteomic profile was performed applying nano-scale liquid chromatography tandem mass spectrometry (nLC-MS) and analyzed with PEAKS software. The percentage of CD32b- LSEC varied across groups, suggesting different capillarization processes. Both CD32+ and CD32b- LSEC from models are different from control LSEC, but differently expressed proteins in CD32b- LSEC are significantly higher. Heatmaps evidenced specific protein expression patterns for each model. Analysis of biological significance comparing dysfunctional CD32b- LSEC with specialized CD32b+ LSEC from controls showed central similarities represented by 45 common down-regulated proteins involved in the suppression of the endocytic machinery and 63 common up-regulated proteins associated with the actin-dependent cytoskeleton reorganization. In summary; substantial differences but also similarities in dysfunctional LSEC from the three most common models of liver disease were found, supporting the idea that LSEC may harbor different protein expression profiles according to the etiology or disease stage.
Collapse
Affiliation(s)
- Mar Gil
- Liver Diseases, Vall d’Hebron Institut de Recerca (VHIR), Liver Unit, Hospital Universitari Vall d’Hebron (HUVH), Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (M.G.); (C.F.); (M.M.-G.); (I.R.); (A.B.); (J.M.P.); (D.H.)
| | - Mikel Azkargorta
- Proteomics Platform, CIC bioGUNE, BRTA (Basque Research & Technology Alliance), Bizkaia Science and Technology Park, 48160 Derio, Spain; (M.A.); (F.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Carla Fuster
- Liver Diseases, Vall d’Hebron Institut de Recerca (VHIR), Liver Unit, Hospital Universitari Vall d’Hebron (HUVH), Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (M.G.); (C.F.); (M.M.-G.); (I.R.); (A.B.); (J.M.P.); (D.H.)
| | - María Martínez-Gómez
- Liver Diseases, Vall d’Hebron Institut de Recerca (VHIR), Liver Unit, Hospital Universitari Vall d’Hebron (HUVH), Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (M.G.); (C.F.); (M.M.-G.); (I.R.); (A.B.); (J.M.P.); (D.H.)
| | - Imma Raurell
- Liver Diseases, Vall d’Hebron Institut de Recerca (VHIR), Liver Unit, Hospital Universitari Vall d’Hebron (HUVH), Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (M.G.); (C.F.); (M.M.-G.); (I.R.); (A.B.); (J.M.P.); (D.H.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Aurora Barberá
- Liver Diseases, Vall d’Hebron Institut de Recerca (VHIR), Liver Unit, Hospital Universitari Vall d’Hebron (HUVH), Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (M.G.); (C.F.); (M.M.-G.); (I.R.); (A.B.); (J.M.P.); (D.H.)
| | - Juan Manuel Pericàs
- Liver Diseases, Vall d’Hebron Institut de Recerca (VHIR), Liver Unit, Hospital Universitari Vall d’Hebron (HUVH), Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (M.G.); (C.F.); (M.M.-G.); (I.R.); (A.B.); (J.M.P.); (D.H.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Diana Hide
- Liver Diseases, Vall d’Hebron Institut de Recerca (VHIR), Liver Unit, Hospital Universitari Vall d’Hebron (HUVH), Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (M.G.); (C.F.); (M.M.-G.); (I.R.); (A.B.); (J.M.P.); (D.H.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Felix Elortza
- Proteomics Platform, CIC bioGUNE, BRTA (Basque Research & Technology Alliance), Bizkaia Science and Technology Park, 48160 Derio, Spain; (M.A.); (F.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Joan Genescà
- Liver Diseases, Vall d’Hebron Institut de Recerca (VHIR), Liver Unit, Hospital Universitari Vall d’Hebron (HUVH), Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (M.G.); (C.F.); (M.M.-G.); (I.R.); (A.B.); (J.M.P.); (D.H.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - María Martell
- Liver Diseases, Vall d’Hebron Institut de Recerca (VHIR), Liver Unit, Hospital Universitari Vall d’Hebron (HUVH), Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (M.G.); (C.F.); (M.M.-G.); (I.R.); (A.B.); (J.M.P.); (D.H.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| |
Collapse
|
44
|
Li L, Sun H, Chen J, Ding C, Yang X, Han H, Sun Q. Mitigation of non-alcoholic steatohepatitis via recombinant Orosomucoid 2, an acute phase protein modulating the Erk1/2-PPARγ-Cd36 pathway. Cell Rep 2023; 42:112697. [PMID: 37355990 DOI: 10.1016/j.celrep.2023.112697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/27/2023] [Accepted: 06/09/2023] [Indexed: 06/27/2023] Open
Abstract
The therapeutic administration of recombinant proteins is utilized in a multitude of research studies for treating various diseases. In this study, we investigate the therapeutic potential of Orosomucoid 2 (Orm2), an acute phase protein predominantly secreted by hepatocytes, for treating non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Our results show that high Orm2 expression prevents high-fat-diet (HFD)-induced obesity in mice. Pharmacological administration of recombinant ORM2 protein ameliorates hepatic steatosis, inflammation, hepatocyte injury, and fibrosis in mouse livers afflicted by NAFLD and NASH under dietary stress. Orm2 knockout mice develop spontaneous obesity under a regular diet and exacerbate HFD-induced steatosis, steatohepatitis, and fibrosis. Mechanistically, Orm2 deletion activates the Erk1/2-PPARγ-Cd36 signaling pathway, increasing fatty acid uptake and absorption in hepatocytes and mice. Overall, our findings underscore the critical role of Orm2 in preventing NASH and associated NAFLD in the context of obesity.
Collapse
Affiliation(s)
- Li Li
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Haoming Sun
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jionghao Chen
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Cong Ding
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaojun Yang
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Hua Han
- Department of Biomedicine, Future Agriculture Institute, Northwest A&F University, Yangling, Shaanxi, China
| | - Qingzhu Sun
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
45
|
Martinez-Lopez S, Angel-Gomis E, Sanchez-Ardid E, Pastor-Campos A, Picó J, Gomez-Hurtado I. The 3Rs in Experimental Liver Disease. Animals (Basel) 2023; 13:2357. [PMID: 37508134 PMCID: PMC10376896 DOI: 10.3390/ani13142357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Patients with cirrhosis present multiple physiological and immunological alterations that play a very important role in the development of clinically relevant secondary complications to the disease. Experimentation in animal models is essential to understand the pathogenesis of human diseases and, considering the high prevalence of liver disease worldwide, to understand the pathophysiology of disease progression and the molecular pathways involved, due to the complexity of the liver as an organ and its relationship with the rest of the organism. However, today there is a growing awareness about the sensitivity and suffering of animals, causing opposition to animal research among a minority in society and some scientists, but also about the attention to the welfare of laboratory animals since this has been built into regulations in most nations that conduct animal research. In 1959, Russell and Burch published the book "The Principles of Humane Experimental Technique", proposing that in those experiments where animals were necessary, everything possible should be done to try to replace them with non-sentient alternatives, to reduce to a minimum their number, and to refine experiments that are essential so that they caused the least amount of pain and distress. In this review, a comprehensive summary of the most widely used techniques to replace, reduce, and refine in experimental liver research is offered, to assess the advantages and weaknesses of available experimental liver disease models for researchers who are planning to perform animal studies in the near future.
Collapse
Affiliation(s)
- Sebastian Martinez-Lopez
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
- Departamento de Medicina Clínica, Universidad Miguel Hernández, 03550 Sant Joan, Spain
| | - Enrique Angel-Gomis
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
- Departamento de Medicina Clínica, Universidad Miguel Hernández, 03550 Sant Joan, Spain
| | - Elisabet Sanchez-Ardid
- CIBERehd, Instituto de Salud Carlos III, 28220 Madrid, Spain
- Servicio de Patología Digestiva, Institut de Recerca IIB-Sant Pau, Hospital de Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Alberto Pastor-Campos
- Oficina de Investigación Responsable, Universidad Miguel Hernández, 03202 Elche, Spain
| | - Joanna Picó
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
| | - Isabel Gomez-Hurtado
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
- Departamento de Medicina Clínica, Universidad Miguel Hernández, 03550 Sant Joan, Spain
- CIBERehd, Instituto de Salud Carlos III, 28220 Madrid, Spain
| |
Collapse
|
46
|
Yamada K, Tanaka T, Kai K, Matsufuji S, Ito K, Kitajima Y, Manabe T, Noshiro H. Suppression of NASH-Related HCC by Farnesyltransferase Inhibitor through Inhibition of Inflammation and Hypoxia-Inducible Factor-1α Expression. Int J Mol Sci 2023; 24:11546. [PMID: 37511305 PMCID: PMC10380354 DOI: 10.3390/ijms241411546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/06/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Inflammatory processes play major roles in carcinogenesis and the progression of hepatocellular carcinoma (HCC) derived from non-alcoholic steatohepatitis (NASH). But, there are no therapies for NASH-related HCC, especially focusing on these critical steps. Previous studies have reported that farnesyltransferase inhibitors (FTIs) have anti-inflammatory and anti-tumor effects. However, the influence of FTIs on NASH-related HCC has not been elucidated. In hepatoblastoma and HCC cell lines, HepG2, Hep3B, and Huh-7, we confirmed the expression of hypoxia-inducible factor (HIF)-1α, an accelerator of tumor aggressiveness and the inflammatory response. We established NASH-related HCC models under inflammation and free fatty acid burden and confirmed that HIF-1α expression was increased under both conditions. Tipifarnib, which is an FTI, strongly suppressed increased HIF-1α, inhibited cell proliferation, and induced apoptosis. Simultaneously, intracellular interleukin-6 as an inflammation marker was increased under both conditions and significantly suppressed by tipifarnib. Additionally, tipifarnib suppressed the expression of phosphorylated nuclear factor-κB and transforming growth factor-β. Finally, in a NASH-related HCC mouse model burdened with diethylnitrosamine and a high-fat diet, tipifarnib significantly reduced tumor nodule formation in association with decreased serum interleukin-6. In conclusion, tipifarnib has anti-tumor and anti-inflammatory effects in a NASH-related HCC model and may be a promising new agent to treat this disease.
Collapse
Affiliation(s)
- Kohei Yamada
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Tomokazu Tanaka
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Keita Kai
- Department of Pathology, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Shohei Matsufuji
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Kotaro Ito
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Yoshihiko Kitajima
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
- Department of Surgery, National Hospital Organization Higashisaga Hospital, Saga 849-0101, Japan
| | - Tatsuya Manabe
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Hirokazu Noshiro
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| |
Collapse
|
47
|
Mechelinck M, Hein M, Kupp C, Braunschweig T, Helmedag MJ, Klinkenberg A, Habigt MA, Klinge U, Tolba RH, Uhlig M. Experimental Liver Cirrhosis Inhibits Restenosis after Balloon Angioplasty. Int J Mol Sci 2023; 24:11351. [PMID: 37511114 PMCID: PMC10379020 DOI: 10.3390/ijms241411351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/09/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The effect of liver cirrhosis on vascular remodeling in vivo remains unknown. Therefore, this study investigates the influence of cholestatic liver cirrhosis on carotid arterial remodeling. A total of 79 male Sprague Dawley rats underwent bile duct ligation (cirrhotic group) or sham surgery (control group) and 28 days later left carotid artery balloon dilatation; 3, 7, 14 and 28 days after balloon dilatation, the rats were euthanized and carotid arteries were harvested. Histological sections were planimetrized, cell counts determined, and systemic inflammatory parameters measured. Up to day 14 after balloon dilatation, both groups showed a comparable increase in neointima area and degree of stenosis. By day 28, however, both values were significantly lower in the cirrhotic group (% stenosis: 20 ± 8 vs. 42 ± 10, p = 0.010; neointimal area [mm2]: 0.064 ± 0.025 vs. 0.138 ± 0.025, p = 0.024). Simultaneously, cell density in the neointima (p = 0.034) and inflammatory parameters were significantly higher in cirrhotic rats. This study demonstrates that cholestatic liver cirrhosis in rats substantially increases neointimal cell consolidation between days 14 and 28. Thereby, consolidation proved important for the degree of stenosis. This may suggest that patients with cholestatic cirrhosis are at lower risk for restenosis after coronary intervention.
Collapse
Affiliation(s)
- Mare Mechelinck
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Marc Hein
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Carolin Kupp
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Till Braunschweig
- Department of Pathology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Marius J Helmedag
- Department of General, Visceral and Transplantation Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Axel Klinkenberg
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Moriz A Habigt
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Uwe Klinge
- Department of General, Visceral and Transplantation Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - René H Tolba
- Institute for Laboratory Animal Science and Experimental Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Moritz Uhlig
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
48
|
Wang H, Shen H, Seo W, Hwang S. Experimental models of fatty liver diseases: Status and appraisal. Hepatol Commun 2023; 7:e00200. [PMID: 37378635 DOI: 10.1097/hc9.0000000000000200] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
Fatty liver diseases, including alcohol-associated liver disease (ALD) and nonalcoholic fatty liver disease nonalcoholic fatty liver disease (NAFLD), affect a large number of people worldwide and become one of the major causes of end-stage liver disease, such as liver cirrhosis and hepatocellular carcinoma (HCC). Unfortunately, there are currently no approved pharmacological treatments for ALD or NAFLD. This situation highlights the urgent need to explore new intervention targets and discover effective therapeutics for ALD and NAFLD. The lack of properly validated preclinical disease models is a major obstacle to the development of clinical therapies. ALD and NAFLD models have been in the development for decades, but there are still no models that recapitulate the full spectrum of ALD and NAFLD. Throughout this review, we summarize the current in vitro and in vivo models used for research on fatty liver diseases and discuss the advantages and limitations of these models.
Collapse
Affiliation(s)
- Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Haiyuan Shen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Wonhyo Seo
- Laboratory of Hepatotoxicity, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
49
|
Li S, Li C, Khan MI, Liu J, Shi Z, Gao D, Qiu B, Ding W. Microneedle array facilitates hepatic sinusoid construction in a large-scale liver-acinus-chip microsystem. MICROSYSTEMS & NANOENGINEERING 2023; 9:75. [PMID: 37303831 PMCID: PMC10247758 DOI: 10.1038/s41378-023-00544-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/03/2023] [Accepted: 02/24/2023] [Indexed: 06/13/2023]
Abstract
Hepatic sinusoids play a key role in maintaining high activities of liver cells in the hepatic acinus. However, the construction of hepatic sinusoids has always been a challenge for liver chips, especially for large-scale liver microsystems. Herein, we report an approach for the construction of hepatic sinusoids. In this approach, hepatic sinusoids are formed by demolding a self-developed microneedle array from a photocurable cell-loaded matrix in a large-scale liver-acinus-chip microsystem with a designed dual blood supply. Primary sinusoids formed by demolded microneedles and spontaneously self-organized secondary sinusoids can be clearly observed. Benefiting from significantly enhanced interstitial flows by formed hepatic sinusoids, cell viability is witnessed to be considerably high, liver microstructure formation occurs, and hepatocyte metabolism is enhanced. In addition, this study preliminarily demonstrates the effects of the resulting oxygen and glucose gradients on hepatocyte functions and the application of the chip in drug testing. This work paves the way for the biofabrication of fully functionalized large-scale liver bioreactors.
Collapse
Affiliation(s)
- Shibo Li
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui 230027 China
- Department of Oncology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| | - Chengpan Li
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Muhammad Imran Khan
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Jing Liu
- School of Biology, Food and Environment, Hefei University, Hefei, Anhui 230601 China
| | - Zhengdi Shi
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195 USA
| | - Bensheng Qiu
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Weiping Ding
- Department of Oncology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| |
Collapse
|
50
|
Wang XX, Jin R, Li XH, Yang Q, Teng X, Liu FF, Wu N, Rao HY, Liu F. Collagen co-localized with macrovesicular steatosis better differentiates fibrosis progression in non-alcoholic fatty liver disease mouse models. Front Med (Lausanne) 2023; 10:1172058. [PMID: 37332758 PMCID: PMC10272541 DOI: 10.3389/fmed.2023.1172058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a global commonly occurring liver disease. However, its exact pathogenesis is not fully understood. The purpose of this study was to quantitatively evaluate the progression of steatosis and fibrosis by examining their distribution, morphology, and co-localization in NAFLD animal models. Methods Six mouse NAFLD groups were established: (1) western diet (WD) group; (2) WD with fructose in drinking water (WDF) group; (3) WDF + carbon tetrachloride (CCl4) group, WDF plus intraperitoneal injection of CCl4; (4) high-fat diet (HFD) group, (5) HFD with fructose (HFDF) group; and (6) HFDF + CCl4 group, HFDF plus intraperitoneal injection of CCl4. Liver tissue specimens from NAFLD model mice were collected at different time points. All the tissues were serially sectioned for histological staining and second-harmonic generation (SHG)/two-photon excitation fluorescence imaging (TPEF) imaging. The progression of steatosis and fibrosis was analyzed using SHG/TPEF quantitative parameters with respect to the non-alcoholic steatohepatitis Clinical Research Network scoring system. Results qSteatosis showed a good correlation with steatosis grade (R: 0.823-0.953, p < 0.05) and demonstrated high performance (area under the curve [AUC]: 0.617-1) in six mouse models. Based on their high correlation with histological scoring, qFibrosis containing four shared parameters (#LongStrPS, #ThinStrPS, #ThinStrPSAgg, and #LongStrPSDis) were selected to create a linear model that could accurately identify differences among fibrosis stages (AUC: 0.725-1). qFibrosis co-localized with macrosteatosis generally correlated better with histological scoring and had a higher AUC in six animal models (AUC: 0.846-1). Conclusion Quantitative assessment using SHG/TPEF technology can be used to monitor different types of steatosis and fibrosis progression in NAFLD models. The collagen co-localized with macrosteatosis could better differentiate fibrosis progression and might aid in developing a more reliable and translatable fibrosis evaluation tool for animal models of NAFLD.
Collapse
Affiliation(s)
- Xiao-Xiao Wang
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Rui Jin
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Xiao-He Li
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Qiang Yang
- Hangzhou Choutu Technology Co., Ltd., Hangzhou, China
| | - Xiao Teng
- HistoIndex Pte Ltd, Singapore, Singapore
| | - Fang-Fang Liu
- Department of Pathology, Peking University People's Hospital, Beijing, China
| | - Nan Wu
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Hui-Ying Rao
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Feng Liu
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| |
Collapse
|