1
|
Kuchay MS, Choudhary NS, Ramos-Molina B. Pathophysiological underpinnings of metabolic dysfunction-associated steatotic liver disease. Am J Physiol Cell Physiol 2025; 328:C1637-C1666. [PMID: 40244183 DOI: 10.1152/ajpcell.00951.2024] [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: 01/22/2025] [Revised: 01/31/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is emerging as the leading cause of chronic liver disease worldwide, reflecting the global epidemics of obesity, metabolic syndrome, and type 2 diabetes. Beyond its strong association with excess adiposity, MASLD encompasses a heterogeneous population that includes individuals with normal body weight ("lean MASLD") highlighting the complexity of its pathogenesis. This disease results from a complex interplay between genetic susceptibility, epigenetic modifications, and environmental factors, which converge to disrupt metabolic homeostasis. Adipose tissue dysfunction and insulin resistance trigger an overflow of lipids to the liver, leading to mitochondrial dysfunction, oxidative stress, and hepatocellular injury. These processes promote hepatic inflammation and fibrogenesis, driven by cross talk among hepatocytes, immune cells, and hepatic stellate cells, with key contributions from gut-liver axis perturbations. Recent advances have unraveled pivotal molecular pathways, such as transforming growth factor-β signaling, Notch-induced osteopontin, and sphingosine kinase 1-mediated responses, that orchestrate fibrogenic activation. Understanding these interconnected mechanisms is crucial for developing targeted therapies. This review integrates current knowledge on the pathophysiology of MASLD, emphasizing emerging concepts such as lean metabolic dysfunction-associated steatohepatitis (MASH), epigenetic alterations, hepatic extracellular vesicles, and the relevance of extrahepatic signals. It also discusses novel therapeutic strategies under investigation, aiming to provide a comprehensive and structured overview of the evolving MASLD landscape for both basic scientists and clinicians.
Collapse
Affiliation(s)
| | - Narendra Singh Choudhary
- Institute of Digestive and Hepatobiliary Sciences, Medanta-The Medicity Hospital, Gurugram, India
| | - Bruno Ramos-Molina
- Group of Obesity, Diabetes & Metabolism, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| |
Collapse
|
2
|
Agarwal H, Wang Y, Tinsley B, Wang X, Ozcan L. RAP1A suppresses hepatic steatosis by regulating amino acid-mediated mTORC1 activation. JHEP Rep 2025; 7:101303. [PMID: 40124164 PMCID: PMC11929108 DOI: 10.1016/j.jhepr.2024.101303] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 03/25/2025] Open
Abstract
Background & Aims Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by triglyceride (TG) build-up in hepatocytes; however, our understanding of the underlying molecular mechanisms is limited. Here, we investigated the role of hepatic GTPase RAP1A in MASLD and its more progressive form, metabolic dysfunction-associated steatohepatitis (MASH). Methods RAP1A was silenced or activated by AAV8-TBG-mediated gene expression or treating mice with a small molecule RAP1 activator (n = 4-12 per group). Primary hepatocytes were used to further probe the newly elucidated pathway. Liver samples from patients with MASH and control livers were analyzed for active RAP1A levels (n = 4 per group). Results Activation of hepatic RAP1A is suppressed in obese mice with MASLD and restoring its activity decreases liver steatosis. RAP1A activation lowers hepatic TG accumulation through decreasing sterol regulatory element-binding protein 1 (SREBP1) cleavage by inhibiting the mechanistic target of rapamycin complex 1 (mTORC1). The mechanism linking RAP1A activation to suppression of mTORC1 involves the lowering of membrane-bound amino acid transporters, which leads to reduced hepatocyte amino acid uptake, decreased intracellular amino acid levels, and inhibition of amino acid-mediated mTORC1 activation. Furthermore, we observed that active-RAP1A levels were decreased in mice fed a MASH-provoking diet (98% lower, p <0.01) and liver extracts from patients with MASH (86% lower, p <0.05). Accordingly, restoration of RAP1A activity in mice liver lowered liver fibrotic gene expression and prevented fibrosis formation, whereas RAP1A silencing promoted the progression of MASH. Conclusions Activation of hepatic RAP1A lowers MASLD and MASH formation by suppressing amino acid-mediated mTORC1 activation and decreasing cleaved SREBP1. These data provide mechanistic insight into amino acid-mediated mTORC1 regulation and raise the possibility that hepatic RAP1A may serve as a mechanistic node linking obesity with MASLD and MASH. Impact and implications Metabolic dysfunction-associated liver pathologies are inadequately treated with currently available therapy. Here we demonstrate that the small GTPase RAS-associated protein 1A (RAP1A) protects against liver steatosis and fibrosis development by decreasing hepatocyte amino acid levels, which results in lower mTORC1 activity and SREBP1 cleavage. The results may present new targets against metabolic dysfunction related liver diseases.
Collapse
Affiliation(s)
- Heena Agarwal
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Yating Wang
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Brea Tinsley
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Lale Ozcan
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| |
Collapse
|
3
|
Gaspar RC, Macêdo APA, Nakandakari SCBR, Muñoz VR, Abud GF, Vieira RFL, de Sousa Neto IV, Pavan ICB, da Silva LGS, Simabuco FM, da Silva ASR, Junior WS, Marchini JS, Nonino CB, Cintra DE, Ropelle ER, Pajvani UB, de Freitas EC, Pauli JR. Notch1 Signalling Is Downregulated by Aerobic Exercise, Leading to Improvement of Hepatic Metabolism in Obese Mice. Liver Int 2025; 45:e70068. [PMID: 40078075 DOI: 10.1111/liv.70068] [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: 11/01/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND AND AIMS Notch1 protein plays a significant role in hepatic metabolism, as evidenced by its correlation with insulin resistance in the livers of obese individuals, making it an intriguing research target. Therefore, this study aims to investigate the impact of aerobic exercise on Notch1 pathways in the hepatic tissue of obese mice and its role in controlling hepatic metabolism. METHODS Therefore, we conducted a cross-sectional study utilising liver biopsies from lean and obese humans, as well as an intervention study involving mice subjected to a high-fat diet. The obese-trained mice group underwent a treadmill-running protocol for 4 weeks. RESULTS Our findings revealed that obese individuals exhibited increased NOTCH1 mRNA levels compared to lean subjects. The detrimental effects of Notch1 signalling were confirmed by Notch1-overexpressed HepG2 cell lines. Obese mice with higher hepatic Notch1 signalling demonstrated a reduction in this pathway when subjected to a 4-week treadmill running. Another benefit noticed in this trained group was the amelioration of insulin resistance, as well as a reduction in pyruvate intolerance and gluconeogenic enzymes. Additionally, we observed that these protective findings were accompanied by a decrease in mTORC1 pathway activity and lipid accumulation in the liver. Pharmacological inhibition of Notch1 in obese mice led to an increase in mitochondrial respiration in the liver. CONCLUSIONS We conclude that Notch1 signalling may be a potentially useful therapeutic target in obesity, while aerobic exercise training suppresses the Notch1 pathway in the liver, contributing to the regulation of hepatic glucose and lipid metabolism in obese mice.
Collapse
Affiliation(s)
- Rafael Calais Gaspar
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
| | - Ana Paula Azevêdo Macêdo
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
| | | | - Vitor Rosetto Muñoz
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
| | - Gabriela Ferreira Abud
- School of Physical Education and Sport of Ribeirao Preto (EEFERP/USP), University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Renan Fudoli Lins Vieira
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
| | - Ivo Vieira de Sousa Neto
- School of Physical Education and Sport of Ribeirao Preto (EEFERP/USP), University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Isadora Carolina Betim Pavan
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
| | - Luiz Guilherme Salvino da Silva
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
- Applied Molecular Signaling Laboratory (LabSIMA), Department of Biochemistry, Federal University of São Paulo, Campinas, Sao Paulo, Brazil
| | - Adelino S R da Silva
- School of Physical Education and Sport of Ribeirao Preto (EEFERP/USP), University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Wilson Salgado Junior
- School of Medicine of Ribeirão Preto, University of Sao Paulo (USP), Ribeirao Preto, Sao Paulo, Brazil
| | - Julio Sergio Marchini
- Department of Internal Medicine, Division of Nutrology, Ribeirao Preto Medical School, University of Sao Paulo (USP), Ribeirao Preto, Sao Paulo, Brazil
| | - Carla Barbosa Nonino
- Division of Nutrition and Metabolism, Department of Health Sciences, Ribeirao Preto Medical School, University of São Paulo (USP), Ribeirao Preto, Sao Paulo, Brazil
| | - Dennys Esper Cintra
- Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Sao Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Sao Paulo, Brazil
| | - Utpal B Pajvani
- Department of Medicine, Columbia University, New York, New York, USA
| | - Ellen Cristini de Freitas
- School of Physical Education and Sport of Ribeirao Preto (EEFERP/USP), University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Sao Paulo, Brazil
| |
Collapse
|
4
|
Qin ZX, Zuo L, Zeng Z, Ma R, Xie W, Zhu X, Zhou X. GalNac-siRNA conjugate delivery technology promotes the treatment of typical chronic liver diseases. Expert Opin Drug Deliv 2025; 22:455-469. [PMID: 39939158 DOI: 10.1080/17425247.2025.2466767] [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: 11/18/2024] [Revised: 01/26/2025] [Accepted: 02/10/2025] [Indexed: 02/14/2025]
Abstract
INTRODUCTION Nucleic acid-based therapeutics have become a key pillar of the 'third wave' of modern medicine, following the eras of small molecule inhibitors and antibody drugs. Their rapid progress is heavily dependent on delivery technologies, with the development of N-acetylgalactosamine (GalNAc) conjugates marking a breakthrough in targeting liver diseases. This technology has gained significant attention for its role in addressing chronic conditions like chronic hepatitis B (CHB) and nonalcoholic steatohepatitis (NASH), which are challenging to treat with conventional methods. AREAS COVERED This review explores the origins, mechanisms, and advantages of GalNAc-siRNA delivery systems, highlighting their ability to target hepatocytes via the asialoglycoprotein receptor (ASGPR). The literature reviewed covers preclinical and clinical advancements, particularly in CHB and NASH. Key developments in stabilization chemistry and conjugation technologies are examined, emphasizing their impact on enhancing therapeutic efficacy and patient compliance. EXPERT OPINION GalNAc-siRNA technology represents a transformative advancement in RNA interference (RNAi) therapies, addressing unmet needs in liver-targeted diseases. While significant progress has been made, challenges remain, including restricted targeting scope and scalability concerns. Continued innovation is expected to expand applications, improve delivery efficiency, and overcome limitations, establishing GalNAc-siRNA as a cornerstone for future nucleic acid-based treatments.
Collapse
Affiliation(s)
- Zhen-Xin Qin
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Ling Zuo
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Ziran Zeng
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Rongguan Ma
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Wenyan Xie
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Xiao Zhu
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
5
|
Xu M, Xu H, Ling YW, Liu JJ, Song P, Fang ZQ, Yue ZS, Duan JL, He F, Wang L. Neutrophil extracellular traps-triggered hepatocellular senescence exacerbates lipotoxicity in non-alcoholic steatohepatitis. J Adv Res 2025:S2090-1232(25)00175-4. [PMID: 40068761 DOI: 10.1016/j.jare.2025.03.015] [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: 12/16/2024] [Revised: 02/19/2025] [Accepted: 03/08/2025] [Indexed: 03/17/2025] Open
Abstract
INTRODUCTION Neutrophils are initial responders in inflammation and contribute to non-alcoholic fatty liver disease (NAFLD) progression to steatohepatitis (NASH). Neutrophil extracellular traps (NETs) are implicated in liver injury, yet their precise mechanisms in NASH progression remains unclear. OBJECTIVES This study investigates how NETs drive NASH progression by exacerbating hepatocyte lipotoxicity and explore the regulatory mechanism of NETs formation and its downstream effects on liver pathology. METHODS Clinical samples from NASH patients and diet-induced NASH mice were analyzed for NET levels. NETs were pharmacologically inhibited, and senescent cells were selectively eliminated in mice. Myeloid-specific RBP-J knockout mice were generated to disrupt Notch signaling, with subsequent evaluation of NET formation, senescence markers, steatosis, fibrosis, and inflammation. RESULTS NETs were elevated in NASH patients and mice, correlating with hepatocyte senescence and lipotoxicity. Pharmacological NET disruption reduced hepatocyte senescence, accompanied by attenuated steatosis and fibrosis. Senescent cell clearance replicated these improvements, confirming liver senescence emerges is a vital step for NETs to promote the progression of NASH. Myeloid-specific Notch signaling ablation suppressed NET generation, concurrently decreasing lipid deposition and liver inflammation. CONCLUSION Our findings elucidate a novel mechanism by which neutrophil-derived Notch driven NETs exacerbate NASH by promoting cell senescence, thereby contributing to hepatic steatosis and fibrosis. This insight may provide potential intervention strategies and therapeutic targets for NASH treatment.
Collapse
Affiliation(s)
- Ming Xu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Hao Xu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yu-Wei Ling
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jing-Jing Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Ping Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Zhi-Qiang Fang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Zhen-Sheng Yue
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Juan-Li Duan
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Fei He
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
6
|
Wang T, Xia G, Li X, Gong M, Lv X. Endoplasmic reticulum stress in liver fibrosis: Mechanisms and therapeutic potential. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167695. [PMID: 39864668 DOI: 10.1016/j.bbadis.2025.167695] [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: 09/29/2024] [Revised: 01/07/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
This paper reviews the important role of endoplasmic reticulum stress in the patho mechanism of liver fibrosis and its potential as a potential target for the treatment of liver fibrosis. Liver fibrosis is the result of sustained inflammation and injury to the liver due to a variety of factors, triggering excessive deposition of extracellular matrix and fibrous scar formation, which in turn leads to loss of liver function and a variety of related complications. Endoplasmic reticulum stress is one of the characteristics of chronic liver disease and is closely related to the pathological process of chronic liver disease, including alcohol-related liver disease, viral hepatitis, and liver fibrosis. The unfolded protein response is one of the important response mechanisms to endoplasmic reticulum stress. It is associated with several pathological aspects of liver fibrosis and the maintenance of endoplasmic reticulum homeostasis. Interventions targeting endoplasmic reticulum stress for the treatment of liver fibrosis have potential research and application value. An in-depth understanding of the biological basis of endoplasmic reticulum stress is also needed in the treatment of liver fibrosis, as well as the development of more effective drugs and interventions to accurately regulate the endoplasmic reticulum signaling network, to achieve the restoration and maintenance of endoplasmic reticulum homeostasis at the cellular and organ levels, and to further promote the reversal of the pathological process of liver fibrosis.
Collapse
Affiliation(s)
- Tiantian Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Hefei, China; School of Pharmacy, Anhui Medical University, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Guoqing Xia
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Hefei, China; School of Pharmacy, Anhui Medical University, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xue Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Hefei, China; School of Pharmacy, Anhui Medical University, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Mingxu Gong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Hefei, China; School of Pharmacy, Anhui Medical University, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiongwen Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Hefei, China; School of Pharmacy, Anhui Medical University, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China.
| |
Collapse
|
7
|
Yan S, Zhao Y, Yang Y, Liu B, Xu W, Ma Z, Yang Q. Progress of ADAM17 in Fibrosis-Related Diseases. Mediators Inflamm 2025; 2025:9999723. [PMID: 40224489 PMCID: PMC11986189 DOI: 10.1155/mi/9999723] [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: 07/27/2024] [Accepted: 01/09/2025] [Indexed: 04/15/2025] Open
Abstract
Fibrosis leads to structural damage and functional decline and is characterized by an accumulation of fibrous connective tissue and a reduction in parenchymal cells. Because of its extremely poor prognosis, organ fibrosis poses a significant economic burden. In order to prevent and treat fibrosis more effectively, potential mechanisms need to be investigated. A disintegrin and metalloprotease 17 (ADAM17) is a membrane-bound protein. It regulates intracellular signaling and membrane protein degradation. Fibrosis mediated by ADAM17 has been identified as an important contributor, although the specific relationship between its multiple regulatory functions and the pathogenesis is unclear. This article describes ADAM17 activation, function, and regulation, as well as the role of ADAM17 mediated fibrosis injury in kidney, liver, heart, lung, skin, endometrium, and retina. To develop new therapeutic approaches based on ADAM17 related signal pathways.
Collapse
Affiliation(s)
- Suyan Yan
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Yaqi Zhao
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong, China
| | - Yuyu Yang
- UCL School of Pharmacy, University College London, London, UK
| | - Baocheng Liu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Wei Xu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong, China
| | - Zhenzhen Ma
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
- Shandong University of Traditional Chinese Medicine, Jinan 250021, Shandong, China
| | - Qingrui Yang
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong, China
| |
Collapse
|
8
|
Ma X, Huang T, Chen X, Li Q, Liao M, Fu L, Huang J, Yuan K, Wang Z, Zeng Y. Molecular mechanisms in liver repair and regeneration: from physiology to therapeutics. Signal Transduct Target Ther 2025; 10:63. [PMID: 39920130 PMCID: PMC11806117 DOI: 10.1038/s41392-024-02104-8] [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: 11/08/2023] [Revised: 09/02/2024] [Accepted: 12/12/2024] [Indexed: 02/09/2025] Open
Abstract
Liver repair and regeneration are crucial physiological responses to hepatic injury and are orchestrated through intricate cellular and molecular networks. This review systematically delineates advancements in the field, emphasizing the essential roles played by diverse liver cell types. Their coordinated actions, supported by complex crosstalk within the liver microenvironment, are pivotal to enhancing regenerative outcomes. Recent molecular investigations have elucidated key signaling pathways involved in liver injury and regeneration. Viewed through the lens of metabolic reprogramming, these pathways highlight how shifts in glucose, lipid, and amino acid metabolism support the cellular functions essential for liver repair and regeneration. An analysis of regenerative variability across pathological states reveals how disease conditions influence these dynamics, guiding the development of novel therapeutic strategies and advanced techniques to enhance liver repair and regeneration. Bridging laboratory findings with practical applications, recent clinical trials highlight the potential of optimizing liver regeneration strategies. These trials offer valuable insights into the effectiveness of novel therapies and underscore significant progress in translational research. In conclusion, this review intricately links molecular insights to therapeutic frontiers, systematically charting the trajectory from fundamental physiological mechanisms to innovative clinical applications in liver repair and regeneration.
Collapse
Affiliation(s)
- Xiao Ma
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiangzheng Chen
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qian Li
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mingheng Liao
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Fu
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiwei Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhen Wang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yong Zeng
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
9
|
Geng W, Liao W, Cao X, Yang Y. Therapeutic Targets and Approaches to Manage Inflammation of NAFLD. Biomedicines 2025; 13:393. [PMID: 40002806 PMCID: PMC11853636 DOI: 10.3390/biomedicines13020393] [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: 12/23/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and its advanced form, non-alcoholic steatohepatitis (NASH), are the leading causes of chronic liver disease globally. They are driven by complex mechanisms where inflammation plays a pivotal role in disease progression. Current therapies, including lifestyle changes and pharmacological agents, are limited in efficacy, particularly in addressing the advanced stages of the disease. Emerging approaches targeting inflammation, metabolic dysfunction, and fibrosis offer promising new directions, though challenges such as treatment complexity and heterogeneity persist. This review concludes the main therapeutic targets and approaches to manage inflammation currently and emphasizes the critical need for future drug development and combination therapy for NAFLD/NASH management.
Collapse
Affiliation(s)
- Wanying Geng
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China;
- Department of Gastroenterology, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (W.L.); (X.C.)
| | - Wanying Liao
- Department of Gastroenterology, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (W.L.); (X.C.)
| | - Xinyuan Cao
- Department of Gastroenterology, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (W.L.); (X.C.)
| | - Yingyun Yang
- Department of Gastroenterology, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (W.L.); (X.C.)
| |
Collapse
|
10
|
Li Y, Xing Y, Liu N, Liu B, Wang Z. SOX9: a key transcriptional regulator in organ fibrosis. Front Pharmacol 2025; 16:1507282. [PMID: 39974732 PMCID: PMC11835943 DOI: 10.3389/fphar.2025.1507282] [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: 10/07/2024] [Accepted: 01/17/2025] [Indexed: 02/21/2025] Open
Abstract
The SOX9 gene locus is not only extensive but also intricate, and it could promote fibrosis in different organs or tissues, including cardiac fibrosis, liver fibrosis, kidney fibrosis, pulmonary fibrosis, as well as other organ fibrosis. Many disorders are associated with the process of fibrosis; moreover, fibrosis is a common symptom of chronic inflammatory diseases, characterized by the accumulation of excessive components in the extracellular matrix through different signaling pathways. The advanced stage of the fibrotic process leads to organ dysfunction and, ultimately, death. In this review, we first give an overview of the original structure and functions of SOX9. Second, we will discuss the role of SOX9 in fibrosis in various organs or tissues. Third, we describe and reveal the possibility of SOX9 as an antifibrotic treatment target. Finally, we will focus on the application of novel technologies for SOX9 and the subsequent investigation of fibrosis.
Collapse
Affiliation(s)
| | | | | | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Zhihui Wang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Li D, Chen J, Ye C, Lin B, Zhang T, Chen Q, Yu C, Wan X. Celastrol ameliorates fibrosis in Western diet/tetrachloromethane-induced nonalcoholic steatohepatitis by suppressing Notch/osteopontin signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156369. [PMID: 39798343 DOI: 10.1016/j.phymed.2025.156369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Celastrol was recently identified as a potential treatment for obesity and hepatic steatosis. However, whether Celastrol effectively suppresses the nonalcoholic fatty liver disease (NAFLD) stage remains unknown. This study aimed to evaluate the role of Celastrol in the progression from simple steatosis to nonalcoholic steatohepatitis (NASH) and fibrosis. METHODS C57BL/6 mice were fed a Western diet combined with a weekly low-dose injection of CCl4 (WD/CCl4) for 16 weeks to establish NASH models. The effects of Celastrol on NASH were further explored through histopathological assessments, immunoblotting, and in vitro analyses. RESULTS Celastrol treatment effectively attenuated hepatic steatosis and fibrosis in WD/CCl4-induced NASH models, in which Notch2 was downregulated by Celastrol in a posttranscriptional manner. In vitro experiments revealed that Notch2 suppression in Celastrol-treated hepatocytes further decreased osteopontin (OPN) levels, inhibiting hepatic stellate cells (HSCs) activation. Moreover, the protective effects of Celastrol on NASH progression were abolished in Notch2-overexpressing mice. CONCLUSION This study demonstrated the protective effects of Celastrol on NASH-related liver fibrosis by modulating Notch/OPN signaling, providing fresh insights into the potential application of Celastrol in NASH treatment.
Collapse
Affiliation(s)
- Dingwu Li
- Department of Gastroenterology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jianing Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Chenhui Ye
- Department of Gastroenterology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Bingru Lin
- Department of Gastroenterology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Tiantian Zhang
- Department of Gastroenterology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Qingxia Chen
- Department of Gastroenterology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Chaohui Yu
- Department of Gastroenterology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| | - Xingyong Wan
- Department of Gastroenterology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
12
|
Lou L, Peng K, Ouyang S, Ding W, Mo J, Yan J, Gong X, Liu G, Lu J, Yue P, Zhang K, Zhang J, Wang YD, Zhang XL. Periostin-mediated NOTCH1 activation between tumor cells and HSCs crosstalk promotes liver metastasis of small cell lung cancer. J Exp Clin Cancer Res 2025; 44:6. [PMID: 39762921 PMCID: PMC11706058 DOI: 10.1186/s13046-024-03266-7] [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: 09/05/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Metastasis is the primary cause of mortality in small cell lung cancer (SCLC), with the liver being a predominant site for distal metastasis. Despite this clinical significance, mechanisms underlying the interaction between SCLC and liver microenvironment, fostering metastasis, remain unclear. METHODS SCLC patient tissue array, bioinformatics analysis were performed to demonstrate the role of periostin (POSTN) in SCLC progression, metastasis, and prognosis. Cell migration, invasion and sphere formation assay were performed to determine the oncogenic role of POSTN. RNA sequencing analysis was utilized to identify the key signaling pathway regulated by POSTN. Immunoprecipitation, immunofluorescence and co-culture system were used to clarify the mechanism of POSTN-NOTCH1 axis in tumor cells-hepatic stellate cells (HSCs) crosstalk. Subcutaneous xenograft model and liver metastasis model were established to examine the anti-tumor growth and metastases effect of targeting POSTN-NOTCH1 signaling axis. RESULTS Elevated expression of POSTN in SCLC is correlated with accelerated tumor progression and metastasis. Conditioned medium rich in POSTN derived from SCLC tumors demonstrates the ability to activate HSCs in the liver microenvironment. Mechanistically, POSTN emerges as a binding partner for the membrane receptor NOTCH1 and transducing the extracellular signals to intracellular fibroblasts. Furthermore, targeting the POSTN-NOTCH1 signaling axis proves effective in suppressing SCLC tumor growth and inhibiting liver metastasis. This study elucidates that the SCLC-derived secreted protein POSTN interacts with NOTCH1 on HSCs to promote the activation of HSCs, thereby providing a favorable microenvironment for liver metastasis. CONCLUSION These findings uncover the intricate communications between primary SCLC cells and HSCs in the tumor microenvironment mediated by the secreted protein POSTN in the context of liver metastasis. Consequently, targeting the POSTN-NOTCH1 signaling axis emerges as a promising therapeutic strategy for metastatic SCLC.
Collapse
Affiliation(s)
- Linlin Lou
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Keren Peng
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Shumin Ouyang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Wen Ding
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Jianshan Mo
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Jiayu Yan
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Xiaoxiao Gong
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Guopin Liu
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Jinjian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Peibin Yue
- Department of Medicine, Division of Hematology-Oncology, and Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Kai Zhang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Jian Zhang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| | - Yan-Dong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Xiao-Lei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| |
Collapse
|
13
|
Canalis E, Yu J, Schilling L, Jafar-nejad P, Carrer M. A NOTCH3 pathogenic variant influences osteogenesis and can be targeted by antisense oligonucleotides in induced pluripotent stem cells. PLoS One 2025; 20:e0316644. [PMID: 39752389 PMCID: PMC11698389 DOI: 10.1371/journal.pone.0316644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 12/15/2024] [Indexed: 01/06/2025] Open
Abstract
Lateral Meningocele Syndrome (LMS), a disorder associated with NOTCH3 pathogenic variants, presents with neurological, craniofacial and skeletal abnormalities. Mouse models of the disease exhibit osteopenia that is ameliorated by the administration of Notch3 antisense oligonucleotides (ASO) targeting either Notch3 or the Notch3 mutation. To determine the consequences of LMS pathogenic variants in human cells and whether they can be targeted by ASOs, induced pluripotent NCRM1 and NCRM5 stem (iPS) cells harboring a NOTCH36692-93insC insertion were created. Parental iPSCs, NOTCH36692-93insC and isogenic controls, free of chromosomal aberrations as determined by human CytoSNP850 array, were cultured under conditions of neural crest, mesenchymal and osteogenic cell differentiation. The expected cell phenotype was confirmed by surface markers and a decline in OCT3/4 and NANOG mRNA. NOTCH36692-93insC cells displayed enhanced expression of Notch target genes HES1, HEY1, 2 and L demonstrating a NOTCH3 gain-of-function. There was enhanced osteogenesis in NOTCH36692-93insC cells as evidenced by increased mineralized nodule formation and ALPL, BGLAP and BSP expression. ASOs targeting NOTCH3 decreased both NOTCH3 wild type and NOTCH36692-93insC mutant mRNA by 40% in mesenchymal and 90% in osteogenic cells. ASOs targeting the NOTCH3 insertion decreased NOTCH36692-93insC by 70-80% in mesenchymal cells and by 45-55% in osteogenic cells and NOTCH3 mRNA by 15-30% and 20-40%, respectively. In conclusion, a NOTCH3 pathogenic variant causes a modest increase in osteoblastogenesis in human iPS cells in vitro and NOTCH3 and NOTCH3 mutant specific ASOs downregulate NOTCH3 transcripts associated with LMS.
Collapse
Affiliation(s)
- Ernesto Canalis
- Departments of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, United States of America
- Departments of Medicine, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, United States of America
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT, United States of America
| | - Jungeun Yu
- Departments of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, United States of America
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT, United States of America
| | - Lauren Schilling
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT, United States of America
| | | | - Michele Carrer
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, United States of America
| |
Collapse
|
14
|
Di X, Li Y, Wei J, Li T, Liao B. Targeting Fibrosis: From Molecular Mechanisms to Advanced Therapies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410416. [PMID: 39665319 PMCID: PMC11744640 DOI: 10.1002/advs.202410416] [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: 08/28/2024] [Revised: 10/27/2024] [Indexed: 12/13/2024]
Abstract
As the final stage of disease-related tissue injury and repair, fibrosis is characterized by excessive accumulation of the extracellular matrix. Unrestricted accumulation of stromal cells and matrix during fibrosis impairs the structure and function of organs, ultimately leading to organ failure. The major etiology of fibrosis is an injury caused by genetic heterogeneity, trauma, virus infection, alcohol, mechanical stimuli, and drug. Persistent abnormal activation of "quiescent" fibroblasts that interact with or do not interact with the immune system via complicated signaling cascades, in which parenchymal cells are also triggered, is identified as the main mechanism involved in the initiation and progression of fibrosis. Although the mechanisms of fibrosis are still largely unknown, multiple therapeutic strategies targeting identified molecular mechanisms have greatly attenuated fibrotic lesions in clinical trials. In this review, the organ-specific molecular mechanisms of fibrosis is systematically summarized, including cardiac fibrosis, hepatic fibrosis, renal fibrosis, and pulmonary fibrosis. Some important signaling pathways associated with fibrosis are also introduced. Finally, the current antifibrotic strategies based on therapeutic targets and clinical trials are discussed. A comprehensive interpretation of the current mechanisms and therapeutic strategies targeting fibrosis will provide the fundamental theoretical basis not only for fibrosis but also for the development of antifibrotic therapies.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Ya Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jingwen Wei
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Tianyue Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Banghua Liao
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| |
Collapse
|
15
|
Liu Y, Yin W. CD36 in liver diseases. Hepatol Commun 2025; 9:e0623. [PMID: 39774047 PMCID: PMC11717518 DOI: 10.1097/hc9.0000000000000623] [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: 01/15/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
Cluster of differentiation 36 (CD36) is a transmembrane glycoprotein with the ability to bind to multiple ligands and perform diverse functions. Through the recognition of long-chain fatty acids, proteins containing thrombospondin structural homology repeat domains such as thrombospondin-1, and molecules with molecular structures consistent with danger- or pathogen-associated molecular patterns, CD36 participates in various physiological and pathological processes of the body. CD36 is widely expressed in various cell types, including hepatocytes and KCs in the liver, where it plays a pivotal role in lipid metabolism, inflammation, and oxidative stress. Accumulating evidence suggests that CD36 plays a complex role in the development of nonalcoholic simple fatty liver disease and NASH and contributes to the pathogenesis of inflammatory liver injury, hepatitis B/hepatitis C, liver fibrosis, and liver cancer. This review summarizes the current understanding of the structural properties, expression patterns, and functional mechanisms of CD36 in the context of liver pathophysiology. Furthermore, the potential of CD36 as a therapeutic target for the prevention and treatment of liver diseases is highlighted.
Collapse
|
16
|
Gilgenkrantz H, Sayegh RA, Lotersztajn S. Immunoregulation of Liver Fibrosis: New Opportunities for Antifibrotic Therapy. Annu Rev Pharmacol Toxicol 2025; 65:281-299. [PMID: 39259981 DOI: 10.1146/annurev-pharmtox-020524-012013] [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] [Indexed: 09/13/2024]
Abstract
Liver fibrosis develops in response to chronic liver injury and is characterized by a sustained inflammatory response that leads to excessive collagen deposition by myofibroblasts. The fibrogenic response is governed by the release of inflammatory mediators from innate, adaptive, and innate-like lymphoid cells and from nonprofessional immune cells (i.e., epithelial cells, hepatic myofibroblasts, and liver sinusoidal endothelial cells). Upon removal of the underlying cause, liver fibrosis can resolve via activation of specific immune cell subsets. Despite major advances in the understanding of fibrosis pathogenesis, there is still no approved antifibrotic therapy. This review summarizes our current knowledge of the immune cell landscape and the inflammatory mechanisms underlying liver fibrosis progression and regression. We discuss how reprogramming immune cell phenotype, in particular through targeting selective inflammatory pathways or modulating cell-intrinsic metabolism, may be translated into antifibrogenic therapies.
Collapse
Affiliation(s)
- Helene Gilgenkrantz
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France;
| | - Rola Al Sayegh
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France;
| | - Sophie Lotersztajn
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France;
| |
Collapse
|
17
|
Chen T, Yang W, Dong R, Yao H, Sun M, Wang J, Zhou Q, Xu J. The effect and application of adiponectin in hepatic fibrosis. Gastroenterol Rep (Oxf) 2024; 12:goae108. [PMID: 39737222 PMCID: PMC11683834 DOI: 10.1093/gastro/goae108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/04/2024] [Accepted: 09/24/2024] [Indexed: 01/01/2025] Open
Abstract
Hepatic fibrosis, a degenerative liver lesion, significantly contributes to the deterioration and mortality among patients with chronic liver diseases. The condition arises from various factors including toxins, such as alcohol, infections like different types of viral hepatitis, and metabolic diseases. Currently, there are no effective treatments available for liver fibrosis. Recent research has shown that adiponectin (ADPN) exhibits inhibitory effects on hepatic fibrosis. ADPN, an adipocytokine secreted by mature adipocytes, features receptors that are widely distributed across multiple tissues, especially the liver. In the liver, direct effects of ADPN on liver fibrosis include reducing inflammation and regulating hepatic stellate cell proliferation and migration. And its indirect effects include alleviating hepatic endoplasmic reticulum stress and reducing inflammation in hepatic lobules, thereby mitigating hepatic fibrosis. This review aims to elucidate the regulatory role of ADPN in liver fibrosis, explore how ADPN and its receptors alleviate endoplasmic reticulum stress, summarize ADPN detection methods, and discuss its potential as a novel marker and therapeutic agent in combating hepatic fibrosis.
Collapse
Affiliation(s)
- Taoran Chen
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Wenjing Yang
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Rongrong Dong
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Han Yao
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Miao Sun
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Jiaxin Wang
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Qi Zhou
- Department of Pediatrics, First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Jiancheng Xu
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun, Jilin, P. R. China
| |
Collapse
|
18
|
Yan L, Song YS, Zhou J, Zhu L, Shi TW, Yu HQ, Dong ZQ, Wang W, Long T, Liu HY, Shi ZY, Li JG. Expression of nicastrin, NICD1, and Hes1 in NCSTN knockout mice: implications for hidradenitis suppurativa, Alzheimer's, and liver cancer. Eur J Med Res 2024; 29:622. [PMID: 39719647 DOI: 10.1186/s40001-024-02225-4] [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: 10/19/2024] [Accepted: 12/15/2024] [Indexed: 12/26/2024] Open
Abstract
BACKGROUND Nicastrin, a subunit of the γ-secretase complex, is encoded by the NCSTN gene and regulates notch signaling, it is involved in the pathogenesis of hidradenitis suppurativa (HS), Alzheimer disease (AD), and liver cancer. However, the animal models for studying HS are relatively scarce. METHODS CRISPR/Cas-mediated genetic engineering was used to generate targeted knockout offspring mice (C57BL/6J). Different doses (10 mg/kg, 20 mg/kg, and 30 mg/kg) and injection methods (subcutaneous/intraperitoneal/gavage injection) of tamoxifen were used to induce the construction of NCSTN knockout mice (mice model). The expressions of nicastrin, NICD1, hes1 in skin, brain, and liver tissue in mice model and wild-type (WT) mice were measured by qRT-PCR and IHC. RESULTS The construction of mice model was successfully induced by tamoxifen, knockout efficiency was 93%, there was no difference in knockout efficiency among three doses, injection methods, genders (P > 0.05). HS-like lesions appeared on the skin of NCSTN knockout mice after 1 month of treatment with tamoxifen, male mice had a higher number of skin lesions compared to female mice (male vs female = 76.5% vs 41.7%, P = 0.027). Compared with WT mice, the expressions of nicastrin (skin P = 0.0009, brain P = 0.0194, liver P = 0.0066), NICD1 (skin P = 0.0115, brain P = 0.0307, liver P = 0.008), hes1 (skin P = 0.0476, brain P = 0.0143, liver P = 0.0003) in mice model all decreased. CONCLUSIONS The NCSTN knockout mouse might be employed as HS animal model; Reducing nicastrin may affect the expression of notch1-hes1 pathway molecules in skin, brain, and liver tissues; low dose (10 mg/kg/d) tamoxifen could be used to induce the deletion of the target gene in mice.
Collapse
Affiliation(s)
- Lu Yan
- Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, No. 7 Weiwu, Zhengzhou, 450003, Henan, China
- The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), The Affiliated Zhengzhou People's Hospital of Xinxiang Medical University, The Affiliated Zhengzhou People's Hospital of Southern Medical University, No. 33 Huanghe Road, Zhengzhou, China
| | - Yin-Sen Song
- The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), The Affiliated Zhengzhou People's Hospital of Xinxiang Medical University, The Affiliated Zhengzhou People's Hospital of Southern Medical University, No. 33 Huanghe Road, Zhengzhou, China
| | - Jian Zhou
- Henan Cancer Hospital (Affiliated Cancer Hospital of Zhengzhou University), 127 Dongming Rd, Zhengzhou, China
| | - Lin Zhu
- The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), The Affiliated Zhengzhou People's Hospital of Xinxiang Medical University, The Affiliated Zhengzhou People's Hospital of Southern Medical University, No. 33 Huanghe Road, Zhengzhou, China
| | - Tian-Wei Shi
- Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, No. 7 Weiwu, Zhengzhou, 450003, Henan, China.
- The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), The Affiliated Zhengzhou People's Hospital of Xinxiang Medical University, The Affiliated Zhengzhou People's Hospital of Southern Medical University, No. 33 Huanghe Road, Zhengzhou, China.
| | - Hui-Qian Yu
- Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, No. 7 Weiwu, Zhengzhou, 450003, Henan, China.
| | - Zi-Qing Dong
- Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Baiyun District, Guangzhou, China
| | - Wei Wang
- The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), The Affiliated Zhengzhou People's Hospital of Xinxiang Medical University, The Affiliated Zhengzhou People's Hospital of Southern Medical University, No. 33 Huanghe Road, Zhengzhou, China
| | - Ting Long
- The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), The Affiliated Zhengzhou People's Hospital of Xinxiang Medical University, The Affiliated Zhengzhou People's Hospital of Southern Medical University, No. 33 Huanghe Road, Zhengzhou, China
| | - Hao-Ying Liu
- The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), The Affiliated Zhengzhou People's Hospital of Xinxiang Medical University, The Affiliated Zhengzhou People's Hospital of Southern Medical University, No. 33 Huanghe Road, Zhengzhou, China
| | - Zhe-Ye Shi
- The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), The Affiliated Zhengzhou People's Hospital of Xinxiang Medical University, The Affiliated Zhengzhou People's Hospital of Southern Medical University, No. 33 Huanghe Road, Zhengzhou, China
| | - Jian-Guo Li
- Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, No. 7 Weiwu, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
19
|
Ma ZJ, Qiu YK, Yu ZW, Song TT, Hu YT, Peng AK, Qi R. Natural small molecule hinokitone mitigates NASH fibrosis by targeting regulation of FXR-mediated hepatocyte apoptosis. J Adv Res 2024:S2090-1232(24)00593-9. [PMID: 39675503 DOI: 10.1016/j.jare.2024.12.016] [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: 07/21/2024] [Revised: 09/23/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024] Open
Abstract
INTRODUCTION Liver fibrosis is the common fate of NASH and poses a major health threat with very limited pharmacological treatments. OBJECTIVES This study aims to investigate the preventive effect of hinokitone (HO), an isolated compound from Agathis dammara, on NASH fibrosis and its underlying mechanism. METHODS To investigate the effect of HO on NASH fibrosis, C57BL/6 mice were either fed a high-fat diet (HFD) in conjunction with intraperitoneal injection of CCl4 for 8 weeks or single CCl4 for 14 days to establish mouse liver fibrosis model, and HO was administered by gavage simultaneously. To elucidate the underlying mechanisms, HepG2 cells were stimulated by palmitic acid (PA) or tumor necrosis factor α plus actinomycin-D (Act-D + TNFα) to induce hepatocyte apoptosis model. Furthermore, hepatocyte Farnesoid-X-receptor (FXR) specifically knocked out mice were established by the albumin-Cre-loxP recombination enzyme system to ascertain the role of FXR in the anti-NASH fibrosis effects of HO. RESULTS The results showed that HO presented dose-dependent anti-liver fibrosis efficacy in NASH mice induced by HFD + CCl4 and CCl4-induced mouse liver fibrosis. Cellularly, HO significantly inhibited PA-induced lipotoxic apoptosis and Act-D + TNFα-induced exogenous apoptosis in hepatocytes, which in turn prevented HSC activation. Mechanistically, bioinformatics analysis and surface plasmon resonance assay had identified hepatocyte FXR as a target of HO. Specifically, HO directly bound to FXR and upregulated its protein level by inhibiting proteasomal degradation. In turn, HO attenuated hepatocyte lipid deposition through upregulating the FXR's downstream target genes SHP and CES1, and reduced cleaved-CASP8 level, thereby inhibiting hepatocyte apoptosis. Furthermore, HO lost its function in the inhibition of hepatocyte apoptosis and liver fibrosis when knockout hepatocyte FXR. CONCLUSION In conclusion, HO has an inhibitory effect on NASH fibrosis. This effect is mediated by targeting upregulation of hepatocyte FXR, which in turn attenuates hepatocyte apoptosis and thus indirectly inhibits the activation of HSCs.
Collapse
Affiliation(s)
- Ze-Jiang Ma
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, State Key Laboratory of Natural and Biomimetic Drugs, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University, Beijing 100191, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing 100191, China; Department of Physiology, School of Basic Medical Sciences, Xinjiang Second Medical College, Karamay, China
| | - Ying-Kun Qiu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Zhe-Wei Yu
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Tian-Tian Song
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, State Key Laboratory of Natural and Biomimetic Drugs, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University, Beijing 100191, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing 100191, China
| | - Yi-Tong Hu
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, State Key Laboratory of Natural and Biomimetic Drugs, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University, Beijing 100191, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing 100191, China
| | - An-Kang Peng
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, State Key Laboratory of Natural and Biomimetic Drugs, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University, Beijing 100191, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing 100191, China; Department of Cardiology, Pu'er People's Hospital, Yunnan, China
| | - Rong Qi
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, State Key Laboratory of Natural and Biomimetic Drugs, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University, Beijing 100191, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing 100191, China.
| |
Collapse
|
20
|
Yadav P, Quadri K, Kadian R, Waziri A, Agrawal P, Alam MS. New approaches to the treatment of metabolic dysfunction-associated steatotic liver with natural products. ILIVER 2024; 3:100131. [DOI: 10.1016/j.iliver.2024.100131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
21
|
Wang S, Gao J, Yang M, Zhang G, Yin L, Tong X. OPN-Mediated Crosstalk Between Hepatocyte E4BP4 and Hepatic Stellate Cells Promotes MASH-Associated Liver Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405678. [PMID: 39473081 PMCID: PMC11653607 DOI: 10.1002/advs.202405678] [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: 05/23/2024] [Revised: 08/06/2024] [Indexed: 12/19/2024]
Abstract
Stressed hepatocytes promote liver fibrosis through communications with hepatic stellate cells (HSCs) during chronic liver injury. However, intra-hepatocyte players that facilitate such cell-to-cell communications are largely undefined. It is previously reported that hepatocyte E4BP4 is potently induced by ER stress and hepatocyte deletion of E4bp4 protects mice from high-fat diet-induced liver steatosis. Here how hepatocyte E4bp4 deficiency impacts the activation of HSCs and the progression toward MASH-associated liver fibrosis is examined. Hepatic E4BP4 is increased in mouse models of NASH diet- or CCl4-induced liver fibrosis. Hepatocyte-specific E4bp4 deletion protected mice against NASH diet-induced liver injury, inflammation, and fibrosis without impacting liver steatosis. Hepatocyte E4BP4 overexpression activated HSCs in a medium transfer experiment, whereas hepatocyte E4bp4 depletion did the opposite. RNA-Seq analysis identified the pro-fibrogenic factor OPN as a critical target of E4BP4 within hepatocytes. Antibody neutralization or shRNA depletion of Opn abrogated hepatocyte E4BP4-induced HSC activation. E4BP4 interacted with and stabilized YAP, an established activator of OPN. Loss of hepatic Yap blocked OPN induction in the liver of Ad-E4bp4-injected mice. Hepatocyte E4BP4 induces OPN via YAP to activate HSCs and promote liver fibrosis during diet-induced MASH. Inhibition of the hepatocyte E4BP4-OPN pathway could offer a novel therapeutic avenue for treating MASLD/MASH.
Collapse
Affiliation(s)
- Sujuan Wang
- Department of Infectious DiseasesThe Second Xiangya HospitalCentral South University139 Renmin Middle Rd, Furong DistrictChangshaHunan410011P. R. China
| | - Jiashi Gao
- Department of Infectious DiseasesThe Second Xiangya HospitalCentral South University139 Renmin Middle Rd, Furong DistrictChangshaHunan410011P. R. China
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolNCRC 20–3843, 2800 Plymouth RoadAnn ArborMI48105USA
- Caswell Diabetes InstituteUniversity of Michigan Medical SchoolNCRC 20–3843, 2800 Plymouth RoadAnn ArborMI48105USA
| | - Meichan Yang
- Department of RadiologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical University106 Zhongshan 2nd RoadGuangzhouGuangdong51008P. R. China
- Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and ApplicationGuangzhouGuangdong51008P. R. China
| | - Gary Zhang
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolNCRC 20–3843, 2800 Plymouth RoadAnn ArborMI48105USA
- Caswell Diabetes InstituteUniversity of Michigan Medical SchoolNCRC 20–3843, 2800 Plymouth RoadAnn ArborMI48105USA
| | - Lei Yin
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolNCRC 20–3843, 2800 Plymouth RoadAnn ArborMI48105USA
- Caswell Diabetes InstituteUniversity of Michigan Medical SchoolNCRC 20–3843, 2800 Plymouth RoadAnn ArborMI48105USA
| | - Xin Tong
- Department of Molecular & Integrative PhysiologyUniversity of Michigan Medical SchoolNCRC 20–3843, 2800 Plymouth RoadAnn ArborMI48105USA
- Caswell Diabetes InstituteUniversity of Michigan Medical SchoolNCRC 20–3843, 2800 Plymouth RoadAnn ArborMI48105USA
| |
Collapse
|
22
|
Xin H, Lai Q, Liu Y, Liao N, Wang Y, Liao B, Zhou K, Zhou Y, Bai Y, Chen Z, Zhou Y. Integrative radiomics analyses identify universal signature for predicting prognosis and therapeutic vulnerabilities across primary and secondary liver cancers: A multi-cohort study. Pharmacol Res 2024; 210:107535. [PMID: 39626849 DOI: 10.1016/j.phrs.2024.107535] [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: 08/28/2024] [Revised: 10/22/2024] [Accepted: 11/29/2024] [Indexed: 12/06/2024]
Abstract
As the hallmark of cancer, genetic and phenotypic heterogeneity leads to biomarkers that are typically tailored to specific cancer type or subtype. This specificity introduces complexities in facilitating streamlined evaluations across diverse cancer types and optimizing therapeutic outcomes. In this study, we comprehensively characterized the radiological patterns underlying liver cancer (LC) by integrating radiomics profiles from computed tomography (CT) images of hepatocellular carcinoma (HCC), intrahepatic cholangiocarcinoma (ICC), and colorectal cancer liver metastases (CRLM) through unsupervised clustering analysis. We identified three distinct radiomics clusters, displaying heterogeneity in prognosis. Subsequently, we formulated a shared prognosticator, the liver cancer radiomics signature (LCRS), by discovering and manifesting connectivity among radiomics phenotypes using GGI strategy. We validated that the LCRS is independent prognostic factor after adjusting for clinic-pathologic variables (all P < 0.05), with the LCRS-High group consistently associated with worse survival outcomes across HCC, ICC, and CRLM. However, the LCRS-High group showed clinical benefit from adjuvant chemotherapy, leading to reduced disease recurrence risk and improved survival. By contrast, the LCRS-Low group, including a subset of gastric cancer liver metastases (GCLM), exhibited more favorable response to immune checkpoint inhibitors (ICIs)-based combinational therapy (P = 0.02, hazard ratio (HR): 0.34 [95 % confidence interval (CI): 0.13-0.88]). Further analysis revealed that Notch signaling pathway was enriched in LCRS-High tumors, while LCRS-Low tumors exhibited higher infiltration of natural killer cell. These findings highlight the promise of this universal scoring model to personalize management strategies for patients with LC.
Collapse
Affiliation(s)
- Hongjie Xin
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qianwei Lai
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanping Liu
- Department of Gastroenterology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Naying Liao
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Wang
- Department of Gastroenterology, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, China
| | - Bihong Liao
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Keyang Zhou
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yuchen Zhou
- Department of General Surgery, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yang Bai
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Zhihua Chen
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China.
| | - Yuanping Zhou
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
23
|
Patidar P, Hirani N, Bharti S, Baig MS. Key regulators of hepatic stellate cell activation in alcohol liver Disease: A comprehensive review. Int Immunopharmacol 2024; 141:112938. [PMID: 39163683 DOI: 10.1016/j.intimp.2024.112938] [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: 05/28/2024] [Revised: 07/27/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024]
Abstract
Alcoholic liver disease (ALD) is a broad category of disorders that begin with liver injury, lead to liver fibrosis, and ultimately conclude in alcohol-induced liver cirrhosis, the most chronic and irreversible liver damage. Liver fibrosis (LF) is a common pathological characteristic observed in most chronic liver inflammatory conditions that involve prolonged inflammation. In this review, we have summarized ethanol-mediated hepatic stellate cell (HSCs) activation and its role in liver fibrosis progression. We highlight important molecular mechanisms that are modulated by ethanol, play a role in the activation of HSCs and the progression of liver fibrosis and identifying potential targets to ameliorate liver fibrosis.
Collapse
Affiliation(s)
- Pramod Patidar
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Nik Hirani
- MRC Centre for Inflammation Research, Queen'sMedicalResearch Institute, University of Edinburgh, Edinburgh, EH164TJ, UK
| | - Shreya Bharti
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India.
| |
Collapse
|
24
|
Mašek J, Filipovic I, Van Hul N, Belicová L, Jiroušková M, Oliveira DV, Frontino AM, Hankeova S, He J, Turetti F, Iqbal A, Červenka I, Sarnová L, Verboven E, Brabec T, Björkström NK, Gregor M, Dobeš J, Andersson ER. Jag1 insufficiency alters liver fibrosis via T cell and hepatocyte differentiation defects. EMBO Mol Med 2024; 16:2946-2975. [PMID: 39358604 PMCID: PMC11554675 DOI: 10.1038/s44321-024-00145-8] [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: 05/15/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
Fibrosis contributes to tissue repair, but excessive fibrosis disrupts organ function. Alagille syndrome (ALGS, caused by mutations in JAGGED1) results in liver disease and characteristic fibrosis. Here, we show that Jag1Ndr/Ndr mice, a model for ALGS, recapitulate ALGS-like fibrosis. Single-cell RNA-seq and multi-color flow cytometry of the liver revealed immature hepatocytes and paradoxically low intrahepatic T cell infiltration despite cholestasis in Jag1Ndr/Ndr mice. Thymic and splenic regulatory T cells (Tregs) were enriched and Jag1Ndr/Ndr lymphocyte immune and fibrotic capacity was tested with adoptive transfer into Rag1-/- mice, challenged with dextran sulfate sodium (DSS) or bile duct ligation (BDL). Transplanted Jag1Ndr/Ndr lymphocytes were less inflammatory with fewer activated T cells than Jag1+/+ lymphocytes in response to DSS. Cholestasis induced by BDL in Rag1-/- mice with Jag1Ndr/Ndr lymphocytes resulted in periportal Treg accumulation and three-fold less periportal fibrosis than in Rag1-/- mice with Jag1+/+ lymphocytes. Finally, the Jag1Ndr/Ndr hepatocyte expression profile and Treg overrepresentation were corroborated in patients' liver samples. Jag1-dependent hepatic and immune defects thus interact to determine the fibrotic process in ALGS.
Collapse
Affiliation(s)
- Jan Mašek
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden.
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00, Prague 2, Czech Republic.
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, 14183, Sweden.
| | - Iva Filipovic
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Noémi Van Hul
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Lenka Belicová
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Markéta Jiroušková
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
| | - Daniel V Oliveira
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00, Prague 2, Czech Republic
| | - Anna Maria Frontino
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00, Prague 2, Czech Republic
| | - Simona Hankeova
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Jingyan He
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Fabio Turetti
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00, Prague 2, Czech Republic
| | - Afshan Iqbal
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Igor Červenka
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Lenka Sarnová
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
| | - Elisabeth Verboven
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Tomáš Brabec
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00, Prague 2, Czech Republic
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
| | - Jan Dobeš
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00, Prague 2, Czech Republic
| | - Emma R Andersson
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden.
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, 14183, Sweden.
| |
Collapse
|
25
|
Šisl D, Planinić P, Novak S, Filipović M, Flegar D, Šućur A, Turčić P, Kovačić N, Kalajzić I, Grčević D, Kelava T. Modulation of Notch signaling pathway in activated hepatic stellate cells does not ameliorate the outcome of liver fibrosis in carbon tetrachloride and DDC-feeding models. Front Pharmacol 2024; 15:1440236. [PMID: 39529885 PMCID: PMC11551037 DOI: 10.3389/fphar.2024.1440236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
Background Recent research suggests a possible role of Notch signaling pathway in development of liver fibrosis, but exact cellular and molecular mechanisms are still not well defined. Methods: We modulated Notch signaling in activated hepatic stellate cells/myofibroblasts using the model of inducible activation or inhibition of Notch signaling selective for αSMA positive cells in murine models of toxic fibrosis induced by CCl4 and cholestatic fibrosis induced by DDC supplemented feeding. Results Our results confirm that Notch signaling pathway is activated in both CCL4 and DDC model of liver fibrosis and that αSMA positive myofibroblasts are of activated hepatic stellate cells origin. However, neither the inhibition of canonical Notch signaling (in tamoxifen treated αSMACreER/RBP-Jfl/fl mice) nor its overactivation (in tamoxifen treated αSMACreER/NICD1 mice) changed the degree of liver fibrosis in comparison to the control groups in either of the investigated models. Furthermore, after the withdrawal of the fibrogenic treatment the degree of resolution of fibrosis was similar between the animals with Notch overactivation and controls. In addition to genetic manipulation, we investigated the effect of antibodies against NOTCH1 and NOTCH2 on the development of liver fibrosis. Treatment with antibodies had effects on thymus and spleen respectively, but failed to ameliorate liver fibrosis. In conclusion, our data demonstrate that modulation of Notch activity in activated HSC is not sufficient to change the outcome of liver fibrosis. The results obtained with inhibitory antibodies further demonstrate limitations of targeting Notch 1 and 2 receptors as antifibrotic therapy. Notch pathway remains a potential target for the treatment of liver fibrosis, but future studies should be directed to Notch 3 signaling and/or targeting different populations of cells.
Collapse
Affiliation(s)
- Dino Šisl
- Laboratory for Molecular Immunology, School of Medicine, Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
- Department of Physiology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Pavao Planinić
- Department of Physiology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Physiology, School of Medicine, University of Mostar, Mostar, Bosnia and Herzegovina
| | - Sanja Novak
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, United States
| | - Maša Filipović
- Laboratory for Molecular Immunology, School of Medicine, Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
- Department of Physiology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Darja Flegar
- Laboratory for Molecular Immunology, School of Medicine, Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
- Department of Physiology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Alan Šućur
- Laboratory for Molecular Immunology, School of Medicine, Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
- Department of Physiology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Petra Turčić
- Department of Pharmacology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Nataša Kovačić
- Laboratory for Molecular Immunology, School of Medicine, Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
- Department of Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ivo Kalajzić
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, United States
| | - Danka Grčević
- Laboratory for Molecular Immunology, School of Medicine, Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
- Department of Physiology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Tomislav Kelava
- Laboratory for Molecular Immunology, School of Medicine, Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
- Department of Physiology, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
26
|
Prasad K, Bhattacharya D, Shams SGE, Izarraras K, Hart T, Mayfield B, Blaszczyk MB, Zhou Z, Pajvani UB, Friedman SL, Bhattacharya M. Kisspeptin Alleviates Human Hepatic Fibrogenesis by Inhibiting TGFβ Signaling in Hepatic Stellate Cells. Cells 2024; 13:1651. [PMID: 39404414 PMCID: PMC11476267 DOI: 10.3390/cells13191651] [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: 09/11/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
The peptide hormone kisspeptin attenuates liver steatosis, metabolic dysfunction-associated steatohepatitis (MASH), and fibrosis in mouse models by signaling via the kisspeptin 1 receptor (KISS1R). However, whether kisspeptin impacts fibrogenesis in the human liver is not known. We investigated the impact of a potent kisspeptin analog (KPA) on fibrogenesis using human precision-cut liver slices (hPCLS) from fibrotic livers from male patients, in human hepatic stellate cells (HSCs), LX-2, and in primary mouse HSCs. In hPCLS, 48 h and 72 h of KPA (3 nM, 100 nM) treatment decreased collagen secretion and lowered the expression of fibrogenic and inflammatory markers. Immunohistochemical studies revealed that KISS1R is expressed and localized to HSCs in MASH/fibrotic livers. In HSCs, KPA treatment reduced transforming growth factor b (TGFβ)-the induced expression of fibrogenic and inflammatory markers, in addition to decreasing TGFβ-induced collagen secretion, cell migration, proliferation, and colony formation. Mechanistically, KISS1R signaling downregulated TGFβ signaling by decreasing SMAD2/3 phosphorylation via the activation of protein phosphatases, PP2A, which dephosphorylates SMAD 2/3. This study revealed for the first time that kisspeptin reverses human hepatic fibrogenesis, thus identifying it as a new therapeutic target to treat hepatic fibrosis.
Collapse
Affiliation(s)
- Kavita Prasad
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (K.P.); (S.G.E.S.); (K.I.); (T.H.)
| | - Dipankar Bhattacharya
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (D.B.); (S.L.F.)
| | - Shams Gamal Eldin Shams
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (K.P.); (S.G.E.S.); (K.I.); (T.H.)
| | - Kimberly Izarraras
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (K.P.); (S.G.E.S.); (K.I.); (T.H.)
| | - Tia Hart
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (K.P.); (S.G.E.S.); (K.I.); (T.H.)
| | - Brent Mayfield
- Department of Medicine, Columbia University, New York, NY 10032, USA; (B.M.); (U.B.P.)
| | - Maryjka B. Blaszczyk
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (M.B.B.); (Z.Z.)
| | - Zhongren Zhou
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (M.B.B.); (Z.Z.)
| | - Utpal B. Pajvani
- Department of Medicine, Columbia University, New York, NY 10032, USA; (B.M.); (U.B.P.)
| | - Scott L. Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (D.B.); (S.L.F.)
| | - Moshmi Bhattacharya
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (K.P.); (S.G.E.S.); (K.I.); (T.H.)
| |
Collapse
|
27
|
Ntambi JN, Kalyesubula M, Cootway D, Lewis SA, Phang YX, Liu Z, O'Neill LM, Lefers L, Huff H, Miller JR, Pegkou Christofi V, Anderson E, Aljohani A, Mutebi F, Dutta M, Patterson A, Ntambi JM. Hepatic stearoyl-CoA desaturase-1 deficiency induces fibrosis and hepatocellular carcinoma-related gene activation under a high carbohydrate low fat diet. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159538. [PMID: 39067685 PMCID: PMC11323073 DOI: 10.1016/j.bbalip.2024.159538] [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: 03/11/2024] [Revised: 07/09/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Stearoyl-CoA desaturase-1 (SCD1) is a pivotal enzyme in lipogenesis, which catalyzes the synthesis of monounsaturated fatty acids (MUFA) from saturated fatty acids, whose ablation downregulates lipid synthesis, preventing steatosis and obesity. Yet deletion of SCD1 promotes hepatic inflammation and endoplasmic reticulum stress, raising the question of whether hepatic SCD1 deficiency promotes further liver damage, including fibrosis. To delineate whether SCD1 deficiency predisposes the liver to fibrosis, cirrhosis, and hepatocellular carcinoma (HCC), we employed in vivo SCD1 deficient global and liver-specific mouse models fed a high carbohydrate low-fat diet and in vitro established AML12 mouse cells. The absence of liver SCD1 remarkably increased the saturation of liver lipid species, as indicated by lipidomic analysis, and led to hepatic fibrosis. Consistently, SCD1 deficiency promoted hepatic gene expression related to fibrosis, cirrhosis, and HCC. Deletion of SCD1 increased the circulating levels of Osteopontin, known to be increased in fibrosis, and alpha-fetoprotein, often used as an early marker and a prognostic marker for patients with HCC. De novo lipogenesis or dietary supplementation of oleate, an SCD1-generated MUFA, restored the gene expression related to fibrosis, cirrhosis, and HCC. Although SCD1 deficient mice are protected against obesity and fatty liver, our results show that MUFA deprivation results in liver injury, including fibrosis, thus providing novel insights between MUFA insufficiency and pathways leading to fibrosis, cirrhosis, and HCC under lean non-steatotic conditions.
Collapse
Affiliation(s)
- Jayne-Norah Ntambi
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA; Tufts Medical Center, Radiation Oncology, 800 Washington St., Box 359, Boston, MA 02111, USA
| | - Mugagga Kalyesubula
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Dylan Cootway
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Sarah A Lewis
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Yar Xin Phang
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Zhaojin Liu
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Lucas M O'Neill
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Lucas Lefers
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Hailey Huff
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Jacqueline Rose Miller
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Veronica Pegkou Christofi
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Ethan Anderson
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Ahmed Aljohani
- College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11564, Saudi Arabia; King Abdullah International Medical Research Center (KAIMRC), Riyadh 11564, Saudi Arabia
| | - Francis Mutebi
- School of Veterinary Medicine and Animal Resources, College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Mainak Dutta
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary & Biomedical Sciences, University Park, PA 16802, United States; Department of Biotechnology, Birla Institute of Technology and Science (BITS) Pilani Dubai Campus, Academic City, Dubai 345055, United Arab Emirates
| | - Andrew Patterson
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary & Biomedical Sciences, University Park, PA 16802, United States; The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, United States
| | - James M Ntambi
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA; Department of Nutritional Sciences, University of Wisconsin-Madison, 1415 Linden Drive, Madison, WI 53706, USA.
| |
Collapse
|
28
|
Diwan R, Gaytan SL, Bhatt HN, Pena-Zacarias J, Nurunnabi M. Liver fibrosis pathologies and potentials of RNA based therapeutics modalities. Drug Deliv Transl Res 2024; 14:2743-2770. [PMID: 38446352 DOI: 10.1007/s13346-024-01551-8] [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] [Accepted: 02/14/2024] [Indexed: 03/07/2024]
Abstract
Liver fibrosis (LF) occurs when the liver tissue responds to injury or inflammation by producing excessive amounts of scar tissue, known as the extracellular matrix. This buildup stiffens the liver tissue, hinders blood flow, and ultimately impairs liver function. Various factors can trigger this process, including bloodborne pathogens, genetic predisposition, alcohol abuse, non-steroidal anti-inflammatory drugs, non-alcoholic steatohepatitis, and non-alcoholic fatty liver disease. While some existing small-molecule therapies offer limited benefits, there is a pressing need for more effective treatments that can truly cure LF. RNA therapeutics have emerged as a promising approach, as they can potentially downregulate cytokine levels in cells responsible for liver fibrosis. Researchers are actively exploring various RNA-based therapeutics, such as mRNA, siRNA, miRNA, lncRNA, and oligonucleotides, to assess their efficacy in animal models. Furthermore, targeted drug delivery systems hold immense potential in this field. By utilizing lipid nanoparticles, exosomes, nanocomplexes, micelles, and polymeric nanoparticles, researchers aim to deliver therapeutic agents directly to specific biomarkers or cytokines within the fibrotic liver, increasing their effectiveness and reducing side effects. In conclusion, this review highlights the complex nature of liver fibrosis, its underlying causes, and the promising potential of RNA-based therapeutics and targeted delivery systems. Continued research in these areas could lead to the development of more effective and personalized treatment options for LF patients.
Collapse
Affiliation(s)
- Rimpy Diwan
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA
- Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX, 79968, USA
| | - Samantha Lynn Gaytan
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA
- Department of Interdisciplinary Health Sciences, College of Health Sciences, The University of Texas El Paso, El Paso, Texas, 79968, USA
| | - Himanshu Narendrakumar Bhatt
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA
- Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX, 79968, USA
| | - Jacqueline Pena-Zacarias
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA
- Department of Biological Sciences, College of Science, The University of Texas El Paso, El Paso, Texas, 79968, USA
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA.
- Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX, 79968, USA.
- Department of Interdisciplinary Health Sciences, College of Health Sciences, The University of Texas El Paso, El Paso, Texas, 79968, USA.
- Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX, 79968, USA.
| |
Collapse
|
29
|
Yang X, Liu Z, Zhou J, Guo J, Han T, Liu Y, Li Y, Bai Y, Xing Y, Wu J, Hu D. SPP1 promotes the polarization of M2 macrophages through the Jak2/Stat3 signaling pathway and accelerates the progression of idiopathic pulmonary fibrosis. Int J Mol Med 2024; 54:89. [PMID: 39129313 PMCID: PMC11335352 DOI: 10.3892/ijmm.2024.5413] [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: 04/12/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal pulmonary disease that requires further investigation to understand its pathogenesis. The present study demonstrated that secreted phosphoprotein 1 (SPP1) was aberrantly highly expressed in the lung tissue of patients with IPF and was significantly positively associated with macrophage and T‑cell activity. Cell localization studies revealed that SPP1 was primarily overexpressed in macrophages, rather than in T cells. Functionally, knocking down SPP1 expression in vitro inhibited the secretion of fibrosis‑related factors and M2 polarization in macrophages. Furthermore, knocking down SPP1 expression inhibited the macrophage‑induced epithelial‑to‑mesenchymal transition in both epithelial and fibroblastic cells. Treatment with SPP1 inhibitors in vivo enhanced lung function and ameliorated pulmonary fibrosis. Mechanistically, SPP1 appears to promote macrophage M2 polarization by regulating the JAK/STAT3 signaling pathway both in vitro and in vivo. In summary, the present study found that SPP1 promotes M2 polarization of macrophages through the JAK2/STAT3 signaling pathway, thereby accelerating the progression of IPF. Inhibition of SPP1 expression in vivo can effectively alleviate the development of IPF, indicating that SPP1 in macrophages may be a potential therapeutic target for IPF.
Collapse
Affiliation(s)
- Xuelian Yang
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui 232001, P.R. China
| | - Ziqin Liu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui 232001, P.R. China
| | - Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui 232001, P.R. China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui 232001, P.R. China
| | - Tao Han
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui 232001, P.R. China
| | - Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui 232001, P.R. China
| | - Yunyun Li
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui 232001, P.R. China
| | - Ying Bai
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui 232001, P.R. China
| | - Yingru Xing
- Department of Clinical Laboratory, Anhui Zhongke Gengjiu Hospital, Hefei, Anhui 230000, P.R. China
| | - Jing Wu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui 232001, P.R. China
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of The Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, Anhui 232001, P.R. China
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui 232001, P.R. China
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of The Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, Anhui 232001, P.R. China
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, P.R. China
| |
Collapse
|
30
|
Nataraj K, Schonfeld M, Rodriguez A, Sharma M, Weinman S, Tikhanovich I. Androgen Effects on Alcohol-induced Liver Fibrosis Are Controlled by a Notch-dependent Epigenetic Switch. Cell Mol Gastroenterol Hepatol 2024; 19:101414. [PMID: 39349250 PMCID: PMC11609386 DOI: 10.1016/j.jcmgh.2024.101414] [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: 04/01/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND & AIMS Alcohol-associated liver disease (ALD) is a major cause of alcohol-related mortality. Sex is an important variable; however, the mechanism behind sex differences is not yet established. METHODS Kdm5b flox/flox Kdm5c flox male mice were subjected to gonadectomy or sham surgery. Mice were fed a Western diet and 20% alcohol in the drinking water for 18 weeks. To induce knockout, mice received 2 × 1011 genome copies of AAV8-CMV-Cre or AAV8-control. To test the role of Notch, mice were treated with 10 mg/kg of avagacestat for 4 weeks. RESULTS We found that Kdm5b/Kdm5c knockout promoted alcohol-induced liver disease, whereas gonadectomy abolished this effect, suggesting that male sex hormones promote liver disease in the absence of KDM5 demethylases. In contrast, in the thioacetamide-induced fibrosis model, male sex hormones showed a protective effect regardless of genotype. In human liver disease samples, we found that androgen receptor expression positively correlated with fibrosis levels when KDM5B levels were low and negatively when KDM5B was high, suggesting that a KDM5B-dependent epigenetic state defines the androgen receptor role in liver fibrosis. Using isolated cells, we found that this difference was due to the differential effect of testosterone on hepatic stellate cell activation in the absence or presence of KDM5B/KDM5C. Moreover, this effect was mediated by KDM5-dependent suppression of Notch signaling. In KDM5-deficient mice, Notch3 and Jag1 gene expression was induced, facilitating testosterone-mediated induction of Notch signaling and stellate cell activation. Inhibiting Notch with avagacestat greatly reduced liver fibrosis and abolished the effect of Kdm5b/Kdm5c loss. CONCLUSIONS Male sex hormone signaling can promote or prevent alcohol-associated liver fibrosis depending on the KDM5-dependent epigenetic state.
Collapse
Affiliation(s)
- Kruti Nataraj
- Department of Internal Medicine, Kansas City VA Medical Center, Kansas City, Missouri
| | - Michael Schonfeld
- Department of Internal Medicine, Kansas City VA Medical Center, Kansas City, Missouri
| | - Adriana Rodriguez
- Department of Internal Medicine, Kansas City VA Medical Center, Kansas City, Missouri
| | - Madhulika Sharma
- Department of Internal Medicine, Kansas City VA Medical Center, Kansas City, Missouri
| | - Steven Weinman
- Department of Internal Medicine, Kansas City VA Medical Center, Kansas City, Missouri; Kansas City VA Medical Center, Kansas City, Missouri
| | - Irina Tikhanovich
- Department of Internal Medicine, Kansas City VA Medical Center, Kansas City, Missouri.
| |
Collapse
|
31
|
Matchett KP, Paris J, Teichmann SA, Henderson NC. Spatial genomics: mapping human steatotic liver disease. Nat Rev Gastroenterol Hepatol 2024; 21:646-660. [PMID: 38654090 DOI: 10.1038/s41575-024-00915-2] [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] [Accepted: 02/28/2024] [Indexed: 04/25/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD, formerly known as non-alcoholic fatty liver disease) is a leading cause of chronic liver disease worldwide. MASLD can progress to metabolic dysfunction-associated steatohepatitis (MASH, formerly known as non-alcoholic steatohepatitis) with subsequent liver cirrhosis and hepatocellular carcinoma formation. The advent of current technologies such as single-cell and single-nuclei RNA sequencing have transformed our understanding of the liver in homeostasis and disease. The next frontier is contextualizing this single-cell information in its native spatial orientation. This understanding will markedly accelerate discovery science in hepatology, resulting in a further step-change in our knowledge of liver biology and pathobiology. In this Review, we discuss up-to-date knowledge of MASLD development and progression and how the burgeoning field of spatial genomics is driving exciting new developments in our understanding of human liver disease pathogenesis and therapeutic target identification.
Collapse
Affiliation(s)
- Kylie P Matchett
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Jasmin Paris
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Cambridge, UK
- Department of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK
| | - Neil C Henderson
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK.
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
32
|
Banerjee A, Farci P. Fibrosis and Hepatocarcinogenesis: Role of Gene-Environment Interactions in Liver Disease Progression. Int J Mol Sci 2024; 25:8641. [PMID: 39201329 PMCID: PMC11354981 DOI: 10.3390/ijms25168641] [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: 06/26/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
The liver is a complex organ that performs vital functions in the body. Despite its extraordinary regenerative capacity compared to other organs, exposure to chemical, infectious, metabolic and immunologic insults and toxins renders the liver vulnerable to inflammation, degeneration and fibrosis. Abnormal wound healing response mediated by aberrant signaling pathways causes chronic activation of hepatic stellate cells (HSCs) and excessive accumulation of extracellular matrix (ECM), leading to hepatic fibrosis and cirrhosis. Fibrosis plays a key role in liver carcinogenesis. Once thought to be irreversible, recent clinical studies show that hepatic fibrosis can be reversed, even in the advanced stage. Experimental evidence shows that removal of the insult or injury can inactivate HSCs and reduce the inflammatory response, eventually leading to activation of fibrolysis and degradation of ECM. Thus, it is critical to understand the role of gene-environment interactions in the context of liver fibrosis progression and regression in order to identify specific therapeutic targets for optimized treatment to induce fibrosis regression, prevent HCC development and, ultimately, improve the clinical outcome.
Collapse
Affiliation(s)
- Anindita Banerjee
- Department of Transfusion Transmitted Diseases, ICMR-National Institute of Immunohaematology, Mumbai 400012, Maharashtra, India;
| | - Patrizia Farci
- Hepatic Pathogenesis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
33
|
Abel ED, Gloyn AL, Evans-Molina C, Joseph JJ, Misra S, Pajvani UB, Simcox J, Susztak K, Drucker DJ. Diabetes mellitus-Progress and opportunities in the evolving epidemic. Cell 2024; 187:3789-3820. [PMID: 39059357 PMCID: PMC11299851 DOI: 10.1016/j.cell.2024.06.029] [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: 03/07/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
Diabetes, a complex multisystem metabolic disorder characterized by hyperglycemia, leads to complications that reduce quality of life and increase mortality. Diabetes pathophysiology includes dysfunction of beta cells, adipose tissue, skeletal muscle, and liver. Type 1 diabetes (T1D) results from immune-mediated beta cell destruction. The more prevalent type 2 diabetes (T2D) is a heterogeneous disorder characterized by varying degrees of beta cell dysfunction in concert with insulin resistance. The strong association between obesity and T2D involves pathways regulated by the central nervous system governing food intake and energy expenditure, integrating inputs from peripheral organs and the environment. The risk of developing diabetes or its complications represents interactions between genetic susceptibility and environmental factors, including the availability of nutritious food and other social determinants of health. This perspective reviews recent advances in understanding the pathophysiology and treatment of diabetes and its complications, which could alter the course of this prevalent disorder.
Collapse
Affiliation(s)
- E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | - Anna L Gloyn
- Department of Pediatrics, Division of Endocrinology & Diabetes, Department of Genetics, Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joshua J Joseph
- Division of Endocrinology, Diabetes and Metabolism, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Shivani Misra
- Department of Metabolism, Digestion and Reproduction, Imperial College London, and Imperial College NHS Trust, London, UK
| | - Utpal B Pajvani
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Judith Simcox
- Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
34
|
Cheng Z, Chu H, Seki E, Lin R, Yang L. Hepatocyte programmed cell death: the trigger for inflammation and fibrosis in metabolic dysfunction-associated steatohepatitis. Front Cell Dev Biol 2024; 12:1431921. [PMID: 39071804 PMCID: PMC11272544 DOI: 10.3389/fcell.2024.1431921] [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: 05/13/2024] [Accepted: 06/28/2024] [Indexed: 07/30/2024] Open
Abstract
By replacing and removing defective or infected cells, programmed cell death (PCD) contributes to homeostasis maintenance and body development, which is ubiquitously present in mammals and can occur at any time. Besides apoptosis, more novel modalities of PCD have been described recently, such as necroptosis, pyroptosis, ferroptosis, and autophagy-dependent cell death. PCD not only regulates multiple physiological processes, but also participates in the pathogenesis of diverse disorders, including metabolic dysfunction-associated steatotic liver disease (MASLD). MASLD is mainly classified into metabolic dysfunction-associated steatotic liver (MASL) and metabolic dysfunction-associated steatohepatitis (MASH), and the latter putatively progresses to cirrhosis and hepatocellular carcinoma. Owing to increased incidence and obscure etiology of MASH, its management still remains a tremendous challenge. Recently, hepatocyte PCD has been attracted much attention as a potent driver of the pathological progression from MASL to MASH, and some pharmacological agents have been proved to exert their salutary effects on MASH partly via the regulation of the activity of hepatocyte PCD. The current review recapitulates the pathogenesis of different modalities of PCD, clarifies the mechanisms underlying how metabolic disorders in MASLD induce hepatocyte PCD and how hepatocyte PCD contributes to inflammatory and fibrotic progression of MASH, discusses several signaling pathways in hepatocytes governing the execution of PCD, and summarizes some potential pharmacological agents for MASH treatment which exert their therapeutic effects partly via the regulation of hepatocyte PCD. These findings indicate that hepatocyte PCD putatively represents a new therapeutic point of intervention for MASH.
Collapse
Affiliation(s)
- Zilu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Rong Lin
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
35
|
Vesković M, Pejović M, Šutulović N, Hrnčić D, Rašić-Marković A, Stanojlović O, Mladenović D. Exploring Fibrosis Pathophysiology in Lean and Obese Metabolic-Associated Fatty Liver Disease: An In-Depth Comparison. Int J Mol Sci 2024; 25:7405. [PMID: 39000518 PMCID: PMC11242866 DOI: 10.3390/ijms25137405] [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: 05/30/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
While obesity-related nonalcoholic fatty liver disease (NAFLD) is linked with metabolic dysfunctions such as insulin resistance and adipose tissue inflammation, lean NAFLD more often progresses to liver fibrosis even in the absence of metabolic syndrome. This review aims to summarize the current knowledge regarding the mechanisms of liver fibrosis in lean NAFLD. The most commonly used lean NAFLD models include a methionine/choline-deficient (MCD) diet, a high-fat diet with carbon tetrachloride (CCl4), and a high-fructose and high-cholesterol diet. The major pro-fibrogenic mechanisms in lean NAFLD models include increased activation of the extracellular signal-regulated kinase (ERK) pathway, elevated expression of α-smooth muscle actin (α-SMA), collagen type I, and TGF-β, and modulation of fibrogenic markers such as tenascin-X and metalloproteinase inhibitors. Additionally, activation of macrophage signaling pathways promoting hepatic stellate cell (HSC) activation further contributes to fibrosis development. Animal models cannot cover all clinical features that are evident in patients with lean or obese NAFLD, implicating the need for novel models, as well as for deeper comparisons of clinical and experimental studies. Having in mind the prevalence of fibrosis in lean NAFLD patients, by addressing specific pathways, clinical studies can reveal new targeted therapies along with novel biomarkers for early detection and enhancement of clinical management for lean NAFLD patients.
Collapse
Affiliation(s)
- Milena Vesković
- Institute of Pathophysiology, Faculty of Medicine, University of Belgrade, Dr Subotića 9, 11000 Belgrade, Serbia
| | - Milka Pejović
- Primary Health Center “Vračar”, Velimira Bate Živojinovića 16, 11000 Belgrade, Serbia
| | - Nikola Šutulović
- Institute of Medical Physiology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000 Belgrade, Serbia
| | - Dragan Hrnčić
- Institute of Medical Physiology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000 Belgrade, Serbia
| | - Aleksandra Rašić-Marković
- Institute of Medical Physiology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000 Belgrade, Serbia
| | - Olivera Stanojlović
- Institute of Medical Physiology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000 Belgrade, Serbia
| | - Dušan Mladenović
- Institute of Pathophysiology, Faculty of Medicine, University of Belgrade, Dr Subotića 9, 11000 Belgrade, Serbia
| |
Collapse
|
36
|
Shang T, Jiang T, Cui X, Pan Y, Feng X, Dong L, Wang H. Diverse functions of SOX9 in liver development and homeostasis and hepatobiliary diseases. Genes Dis 2024; 11:100996. [PMID: 38523677 PMCID: PMC10958229 DOI: 10.1016/j.gendis.2023.03.035] [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: 07/26/2022] [Revised: 02/13/2023] [Accepted: 03/19/2023] [Indexed: 03/26/2024] Open
Abstract
The liver is the central organ for digestion and detoxification and has unique metabolic and regenerative capacities. The hepatobiliary system originates from the foregut endoderm, in which cells undergo multiple events of cell proliferation, migration, and differentiation to form the liver parenchyma and ductal system under the hierarchical regulation of transcription factors. Studies on liver development and diseases have revealed that SRY-related high-mobility group box 9 (SOX9) plays an important role in liver embryogenesis and the progression of hepatobiliary diseases. SOX9 is not only a master regulator of cell fate determination and tissue morphogenesis, but also regulates various biological features of cancer, including cancer stemness, invasion, and drug resistance, making SOX9 a potential biomarker for tumor prognosis and progression. This review systematically summarizes the latest findings of SOX9 in hepatobiliary development, homeostasis, and disease. We also highlight the value of SOX9 as a novel biomarker and potential target for the clinical treatment of major liver diseases.
Collapse
Affiliation(s)
- Taiyu Shang
- School of Life Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Tianyi Jiang
- National Center for Liver Cancer, The Naval Medical University, Shanghai 201805, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Xiaowen Cui
- National Center for Liver Cancer, The Naval Medical University, Shanghai 201805, China
| | - Yufei Pan
- National Center for Liver Cancer, The Naval Medical University, Shanghai 201805, China
| | - Xiaofan Feng
- National Center for Liver Cancer, The Naval Medical University, Shanghai 201805, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Liwei Dong
- National Center for Liver Cancer, The Naval Medical University, Shanghai 201805, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Hongyang Wang
- School of Life Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
- National Center for Liver Cancer, The Naval Medical University, Shanghai 201805, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
- Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Second Military Medical University & Ministry of Education, Shanghai 200438, China
| |
Collapse
|
37
|
Li S, Xiong F, Zhang S, Liu J, Gao G, Xie J, Wang Y. Oligonucleotide therapies for nonalcoholic steatohepatitis. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102184. [PMID: 38665220 PMCID: PMC11044058 DOI: 10.1016/j.omtn.2024.102184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Nonalcoholic steatohepatitis (NASH) represents a severe disease subtype of nonalcoholic fatty liver disease (NAFLD) that is thought to be highly associated with systemic metabolic abnormalities. It is characterized by a series of substantial liver damage, including hepatocellular steatosis, inflammation, and fibrosis. The end stage of NASH, in some cases, may result in cirrhosis and hepatocellular carcinoma (HCC). Nowadays a large number of investigations are actively under way to test various therapeutic strategies, including emerging oligonucleotide drugs (e.g., antisense oligonucleotide, small interfering RNA, microRNA, mimic/inhibitor RNA, and small activating RNA) that have shown high potential in treating this fatal liver disease. This article systematically reviews the pathogenesis of NASH/NAFLD, the promising druggable targets proven by current studies in chemical compounds or biological drug development, and the feasibility and limitations of oligonucleotide-based therapeutic approaches under clinical or pre-clinical studies.
Collapse
Affiliation(s)
- Sixu Li
- Department of Pathophysiology, West China College of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610066, China
| | - Feng Xiong
- Department of Cardiology, The Third People’s Hospital of Chengdu, Chengdu 610031, China
| | - Songbo Zhang
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Jinghua Liu
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Viral Vector Core, University of Massachusetts Chan Medical, School, Worcester, MA 01605, USA
| | - Jun Xie
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Viral Vector Core, University of Massachusetts Chan Medical, School, Worcester, MA 01605, USA
| | - Yi Wang
- Department of Pathophysiology, West China College of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610066, China
| |
Collapse
|
38
|
Chen J, Zhang J, Xia Y, Li J, Jia Q, Zhang Z, Jing X, Xu Y, Zou L, Wang L, Song H, Li J, Liu Q, Xiong Y, Tang Q, Chen W, Yang N, Xu H, Li Y, He J. Reactive Oxygen Species-Responsive Delivery of a Notch Inhibitor to Alleviate Nonalcoholic Steatohepatitis by Inhibiting Hepatic de Novo Lipogenesis and Inflammation. Mol Pharm 2024; 21:2922-2936. [PMID: 38751169 DOI: 10.1021/acs.molpharmaceut.4c00070] [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] [Indexed: 06/04/2024]
Abstract
With the increased prevalence of nonalcoholic steatohepatitis (NASH) in the world, effective pharmacotherapy in clinical practice is still lacking. Previous studies have shown that dibenzazepine (DBZ), a Notch inhibitor, could alleviate NASH development in a mouse model. However, low bioavailability, poor water solubility, and extrahepatic side effects restrict its clinical application. To overcome these barriers, we developed a reactive oxygen species (ROS)-sensitive nanoparticle based on the conjugation of bilirubin to poly(ethylene glycol) (PEG) chains, taking into account the overaccumulation of hepatic ROS in the pathologic state of nonalcoholic steatohepatitis (NASH). The PEGylated bilirubin can self-assemble into nanoparticles in an aqueous solution and encapsulate insoluble DBZ into its hydrophobic cavity. DBZ nanoparticles (DBZ Nps) had good stability, rapidly released DBZ in response to H2O2, and effectively scavenged intracellular ROS of hepatocytes. After systemic administration, DBZ Nps could accumulate in the liver of the NASH mice, extend persistence in circulation, and improve the bioavailability of DBZ. Furthermore, DBZ Nps significantly improved glucose intolerance, relieved hepatic lipid accumulation and inflammation, and ameliorated NASH-induced liver fibrosis. Additionally, DBZ Nps had no significant extrahepatic side effects. Taken together, our results highlight the potential of the ROS-sensitive DBZ nanoparticle as a promising therapeutic strategy for NASH.
Collapse
Affiliation(s)
- Jiahao Chen
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Jinhang Zhang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Yan Xia
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Jiahui Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Qingyi Jia
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Zijing Zhang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Xiandan Jing
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Ying Xu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Ling Zou
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Lingling Wang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Haiying Song
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Jingwei Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Qinhui Liu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Yimin Xiong
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Qin Tang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Wenfei Chen
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Na Yang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Haixia Xu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Yanping Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Jinhan He
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital,Sichuan University, Chengdu, Sichuan Province 610041, China
| |
Collapse
|
39
|
Xu C, Tai H, Chu Y, Liu Y, He J, Wang Y, Su B, Li S. Gossypetin targets the liver-brain axis to alleviate pre-existing liver fibrosis and hippocampal neuroinflammation in mice. Front Pharmacol 2024; 15:1385330. [PMID: 38860164 PMCID: PMC11163038 DOI: 10.3389/fphar.2024.1385330] [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: 02/12/2024] [Accepted: 04/24/2024] [Indexed: 06/12/2024] Open
Abstract
Liver fibrosis occurs in response to chronic damage and inflammation to the liver. Leaving untreated, it can lead to decreased liver function and can eventually progress to cirrhosis, a more advanced and irreversible state of liver damage. Clinical investigations showed that chronic liver disease associated with neurological symptoms including anxiety, depression, and cognitive decline. However, few therapeutic options are available for treating liver and related brain pathologies simultaneously. In this study, we aim to find therapeutic candidates that target the liver-brain axis. Gossypetin, a flavonoid from sedum, shows promising capability in treating liver and brain pathologies in CCl4-induced mouse model. Short term of gossypetin administration is sufficient to ameliorate impaired liver function and pre-existing liver fibrosis, suppress MKK3/6-p38 MAPK and p53 activation, and abolish the activation of hepatic stellate cells and Kupffer cells. Although we observe no neuronal loss in the brain of mice with liver fibrosis, we do observe astrogliosis and microglial activation in certain brain regions, especially the hippocampus. Brief gossypetin administration also shows potential in alleviating neuroinflammation in these regions. These results suggest that gossypetin can target the liver-brain axis and be a promising candidate for treating chronic liver fibrosis patients with neurological symptoms.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bingyin Su
- Development and Regeneration Key Lab of Sichuan Province, Department of Histology and Embryology, Department of Pathology, Chengdu Medical College, Chengdu, China
| | - Shurong Li
- Development and Regeneration Key Lab of Sichuan Province, Department of Histology and Embryology, Department of Pathology, Chengdu Medical College, Chengdu, China
| |
Collapse
|
40
|
Sokal-Dembowska A, Jarmakiewicz-Czaja S, Ferenc K, Filip R. Can Nutraceuticals Support the Treatment of MASLD/MASH, and thus Affect the Process of Liver Fibrosis? Int J Mol Sci 2024; 25:5238. [PMID: 38791276 PMCID: PMC11120776 DOI: 10.3390/ijms25105238] [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: 03/30/2024] [Revised: 04/30/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Currently, metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH) are considered to be the main causes of fibrosis. In turn, fibrosis may lead to the development of hepatocellular carcinoma or advanced cirrhosis, i.e., potentially life-threatening conditions. It is likely that therapy aimed at reducing the risk of developing hepatic steatosis and inflammation could be helpful in minimizing the threat/probability of organ fibrosis. In recent years, increasing attention has been paid to the influence of nutraceuticals in the prevention and treatment of liver diseases. Therefore, the aim of this review was to describe the precise role of selected ingredients such as vitamin C, beta-carotene, omega-3 fatty acids, and curcumin. It is likely that the use of these ingredients in the treatment of patients with MASLD/MASH, along with behavioral and pharmacological therapy, may have a beneficial effect on combating inflammation, reducing oxidative stress, and thereby preventing liver damage.
Collapse
Affiliation(s)
- Aneta Sokal-Dembowska
- Institute of Health Sciences, Medical College, Rzeszow University, 35-959 Rzeszow, Poland
| | | | - Katarzyna Ferenc
- Institute of Medicine, Medical College, Rzeszow University, 35-959 Rzeszow, Poland
| | - Rafał Filip
- Institute of Medicine, Medical College, Rzeszow University, 35-959 Rzeszow, Poland
- Department of Gastroenterology with IBD Unit, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| |
Collapse
|
41
|
Elseweidy MM, Ali AEM, Hassanin SM, Mahmoud YK. Empagliflozin ameliorates liver fibrosis in NASH rat model via targeting hepatic NF-κB/SOX9/OPN signaling and osteocalcin level. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3449-3459. [PMID: 37962587 PMCID: PMC11074015 DOI: 10.1007/s00210-023-02826-6] [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: 09/07/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) may be associated with tissue fibrotic changes and can be treated via different therapeutic tools which may however either initiate weak or long-term side effects that minimize its use. Empagliflozin (EMPA) is an oral anti-diabetic drug which has characteristic effects during hepatic steatosis regarding lipid accumulation and insulin resistance. In this study, we aimed to investigate an additional mechanism through which EMPA can exert and potentiate its anti-inflammatory and anti-fibrotic effects in NASH rat model. Male Wistar albino rats fed on high fat diet (HFD) and 20% fructose in drinking water for 18 weeks and received EMPA (30 mg/kg/day, orally) starting from week 11. Body and liver weights, homeostatic model assessment of insulin resistance (HOMA-IR), lipid profile, liver function tests, other biochemical and histological parameters were determined. HFD joined with fructose intake significantly increased body and liver weights, HOMA-IR value, hepatic inflammatory and fibrotic markers, liver transaminases, hepatic expression of nuclear factor-kappa B (NF-κB), sex determining region Y box 9 (SOX 9), and osteopontin (OPN) with significant decrease in hepatic osteocalcin (OCN). Intense hepatic lesions with severe microsteatosis and deposition of collagen fibers were clearly observed. Effectively, EMPA restored the normal liver functions, downregulated hepatic inflammatory cytokines, NF-κB, SOX 9, OPN, and increased OCN level. These results highlight another pathway illustrated the anti-fibrotic effects of EMPA against liver fibrosis probably through downregulation of NF-κB/SOX 9/OPN signaling along with upregulation of hepatic OCN which may potentiate the valuable anti-inflammatory and anti-fibrotic effects of EMPA.
Collapse
Affiliation(s)
- Mohamed M Elseweidy
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Abd El-Monem Ali
- Pathology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Sara M Hassanin
- Zagazig University Hospitals, Zagazig University, Zagazig, Egypt
| | - Yasmin K Mahmoud
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
42
|
Buniatian GH, Schwinghammer U, Tremmel R, Cynis H, Weiss TS, Weiskirchen R, Lauschke VM, Youhanna S, Ramos I, Valcarcel M, Seferyan T, Rahfeld J, Rieckmann V, Klein K, Buadze M, Weber V, Kolak V, Gebhardt R, Friedman SL, Müller UC, Schwab M, Danielyan L. Consequences of Amyloid-β Deficiency for the Liver. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307734. [PMID: 38430535 PMCID: PMC11095235 DOI: 10.1002/advs.202307734] [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: 10/15/2023] [Revised: 01/27/2024] [Indexed: 03/04/2024]
Abstract
The hepatic content of amyloid beta (Aβ) decreases drastically in human and rodent cirrhosis highlighting the importance of understanding the consequences of Aβ deficiency in the liver. This is especially relevant in view of recent advances in anti-Aβ therapies for Alzheimer's disease (AD). Here, it is shown that partial hepatic loss of Aβ in transgenic AD mice immunized with Aβ antibody 3D6 and its absence in amyloid precursor protein (APP) knockout mice (APP-KO), as well as in human liver spheroids with APP knockdown upregulates classical hallmarks of fibrosis, smooth muscle alpha-actin, and collagen type I. Aβ absence in APP-KO and deficiency in immunized mice lead to strong activation of transforming growth factor-β (TGFβ), alpha secretases, NOTCH pathway, inflammation, decreased permeability of liver sinusoids, and epithelial-mesenchymal transition. Inversely, increased systemic and intrahepatic levels of Aβ42 in transgenic AD mice and neprilysin inhibitor LBQ657-treated wild-type mice protect the liver against carbon tetrachloride (CCl4)-induced injury. Transcriptomic analysis of CCl4-treated transgenic AD mouse livers uncovers the regulatory effects of Aβ42 on mitochondrial function, lipid metabolism, and its onco-suppressive effects accompanied by reduced synthesis of extracellular matrix proteins. Combined, these data reveal Aβ as an indispensable regulator of cell-cell interactions in healthy liver and a powerful protector against liver fibrosis.
Collapse
Affiliation(s)
- Gayane Hrachia Buniatian
- Department of Clinical PharmacologyUniversity Hospital of TuebingenAuf der Morgenstelle 872076TuebingenGermany
| | - Ute Schwinghammer
- Department of Clinical PharmacologyUniversity Hospital of TuebingenAuf der Morgenstelle 872076TuebingenGermany
| | - Roman Tremmel
- Dr. Margarete Fischer‐Bosch Institute of Clinical PharmacologyAuerbachstr. 11270376StuttgartGermany
- University of Tuebingen72074TuebingenGermany
| | - Holger Cynis
- Department of Drug Design and Target ValidationFraunhofer Institute for Cell Therapy and ImmunologyWeinbergweg 2206120Halle (Saale)Germany
- Junior Research Group, Immunomodulation in Pathophysiological ProcessesFaculty of MedicineMartin‐Luther‐University Halle‐WittenbergWeinbergweg 2206120Halle (Saale)Germany
| | - Thomas S. Weiss
- Children's University Hospital (KUNO)University Hospital RegensburgFranz‐Josef‐Strauss‐Allee 1193053RegensburgGermany
| | - Ralf Weiskirchen
- Institute of Molecular PathobiochemistryExperimental Gene Therapy and Clinical ChemistryRWTH University Hospital AachenPauwelsstr. 3052074AachenGermany
| | - Volker M. Lauschke
- Dr. Margarete Fischer‐Bosch Institute of Clinical PharmacologyAuerbachstr. 11270376StuttgartGermany
- University of Tuebingen72074TuebingenGermany
- Department of Physiology and Pharmacology Karolinska InstituteStockholm171 77Sweden
| | - Sonia Youhanna
- Department of Physiology and Pharmacology Karolinska InstituteStockholm171 77Sweden
| | - Isbaal Ramos
- Innovative Technologies in Biological Systems SL (INNOPROT)BizkaiaDerio48160Spain
| | - Maria Valcarcel
- Innovative Technologies in Biological Systems SL (INNOPROT)BizkaiaDerio48160Spain
| | - Torgom Seferyan
- H. Buniatian Institute of BiochemistryNational Academy of Sciences of the Republic of Armenia (NAS RA)5/1 Paruir Sevak St.Yerevan0014Armenia
| | - Jens‐Ulrich Rahfeld
- Department of Drug Design and Target ValidationFraunhofer Institute for Cell Therapy and ImmunologyWeinbergweg 2206120Halle (Saale)Germany
| | - Vera Rieckmann
- Department of Drug Design and Target ValidationFraunhofer Institute for Cell Therapy and ImmunologyWeinbergweg 2206120Halle (Saale)Germany
| | - Kathrin Klein
- Dr. Margarete Fischer‐Bosch Institute of Clinical PharmacologyAuerbachstr. 11270376StuttgartGermany
- University of Tuebingen72074TuebingenGermany
| | - Marine Buadze
- Department of Clinical PharmacologyUniversity Hospital of TuebingenAuf der Morgenstelle 872076TuebingenGermany
| | - Victoria Weber
- Department of Clinical PharmacologyUniversity Hospital of TuebingenAuf der Morgenstelle 872076TuebingenGermany
| | - Valentina Kolak
- Department of Clinical PharmacologyUniversity Hospital of TuebingenAuf der Morgenstelle 872076TuebingenGermany
| | - Rolf Gebhardt
- Rudolf‐Schönheimer Institute of BiochemistryFaculty of MedicineUniversity of LeipzigJohannisstraße 3004103LeipzigGermany
| | - Scott L. Friedman
- Division of Liver DiseasesIcahn School of Medicine at Mount Sinai1425 Madison AveNew YorkNY10029USA
| | - Ulrike C. Müller
- Institute for Pharmacy and Molecular Biotechnology IPMBDepartment of Functional GenomicsUniversity of HeidelbergIm Neuenheimer Feld 36469120HeidelbergGermany
| | - Matthias Schwab
- Department of Clinical PharmacologyUniversity Hospital of TuebingenAuf der Morgenstelle 872076TuebingenGermany
- Dr. Margarete Fischer‐Bosch Institute of Clinical PharmacologyAuerbachstr. 11270376StuttgartGermany
- Departments of Biochemistry and Clinical Pharmacologyand Neuroscience LaboratoryYerevan State Medical University2‐ Koryun StYerevan0025Armenia
- Cluster of Excellence iFIT (EXC2180) “Image‐guided and Functionally Instructed Tumor Therapies”University of Tübingen72076TübingenGermany
| | - Lusine Danielyan
- Department of Clinical PharmacologyUniversity Hospital of TuebingenAuf der Morgenstelle 872076TuebingenGermany
- Departments of Biochemistry and Clinical Pharmacologyand Neuroscience LaboratoryYerevan State Medical University2‐ Koryun StYerevan0025Armenia
| |
Collapse
|
43
|
Yan T, Yan N, Xia Y, Sawaswong V, Zhu X, Dias HB, Aibara D, Takahashi S, Hamada K, Saito Y, Li G, Liu H, Yan H, Velenosi TJ, Krausz KW, Huang J, Kimura S, Rotman Y, Qu A, Hao H, Gonzalez FJ. Hepatocyte-specific CCAAT/enhancer binding protein α restricts liver fibrosis progression. J Clin Invest 2024; 134:e166731. [PMID: 38557493 PMCID: PMC10977981 DOI: 10.1172/jci166731] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/09/2024] [Indexed: 04/04/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) - previously described as nonalcoholic steatohepatitis (NASH) - is a major driver of liver fibrosis in humans, while liver fibrosis is a key determinant of all-cause mortality in liver disease independent of MASH occurrence. CCAAT/enhancer binding protein α (CEBPA), as a versatile ligand-independent transcriptional factor, has an important function in myeloid cells, and is under clinical evaluation for cancer therapy. CEBPA is also expressed in hepatocytes and regulates glucolipid homeostasis; however, the role of hepatocyte-specific CEBPA in modulating liver fibrosis progression is largely unknown. Here, hepatic CEBPA expression was found to be decreased during MASH progression both in humans and mice, and hepatic CEBPA mRNA was negatively correlated with MASH fibrosis in the human liver. CebpaΔHep mice had markedly enhanced liver fibrosis induced by a high-fat, high-cholesterol, high-fructose diet or carbon tetrachloride. Temporal and spatial hepatocyte-specific CEBPA loss at the progressive stage of MASH in CebpaΔHep,ERT2 mice functionally promoted liver fibrosis. Mechanistically, hepatocyte CEBPA directly repressed Spp1 transactivation to reduce the secretion of osteopontin, a fibrogenesis inducer of hepatic stellate cells. Forced hepatocyte-specific CEBPA expression reduced MASH-associated liver fibrosis. These results demonstrate an important role for hepatocyte-specific CEBPA in liver fibrosis progression, and may help guide the therapeutic discoveries targeting hepatocyte CEBPA for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Tingting Yan
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
- State Key Laboratory of Natural Medicines, Laboratory of Metabolic Regulation and Drug Target Discovery, China Pharmaceutical University, Nanjing, China
| | - Nana Yan
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
- State Key Laboratory of Natural Medicines, Laboratory of Metabolic Regulation and Drug Target Discovery, China Pharmaceutical University, Nanjing, China
| | - Yangliu Xia
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Vorthon Sawaswong
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Xinxin Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, and Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Henrique Bregolin Dias
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Daisuke Aibara
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Shogo Takahashi
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Keisuke Hamada
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Yoshifumi Saito
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Hui Liu
- Department of Pathology, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Hualong Yan
- Cancer and Stem Cell Epigenetics, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute and
| | - Thomas J. Velenosi
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kristopher W. Krausz
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jing Huang
- Cancer and Stem Cell Epigenetics, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute and
| | - Shioko Kimura
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Yaron Rotman
- Liver and Energy Metabolism Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, and Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Laboratory of Metabolic Regulation and Drug Target Discovery, China Pharmaceutical University, Nanjing, China
| | - Frank J. Gonzalez
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
44
|
Steffani M, Geng Y, Pajvani UB, Schwabe RF. Protective hepatocyte signals restrain liver fibrosis in metabolic dysfunction-associated steatohepatitis. J Clin Invest 2024; 134:e179710. [PMID: 38557494 PMCID: PMC10977975 DOI: 10.1172/jci179710] [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] [Indexed: 04/04/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) affects nearly 40% of the global adult population and may progress to metabolic dysfunction-associated steatohepatitis (MASH), and MASH-associated liver fibrosis and cirrhosis. Despite numerous studies unraveling the mechanism of hepatic fibrogenesis, there are still no approved antifibrotic therapies. The development of MASLD and liver fibrosis results from complex cell-cell interactions that often initiate within hepatocytes but remain incompletely understood. In this issue of the JCI, Yan and colleagues describe an ATF3/HES1/CEBPA/OPN pathway that links hepatocyte signals to fibrogenic activation of hepatic stellate cells and may provide new perspectives on therapeutic options for MASLD-induced liver fibrosis.
Collapse
Affiliation(s)
- Marcella Steffani
- Department of Medicine, Columbia University, New York, New York, USA
| | - Yana Geng
- Department of Medicine, Columbia University, New York, New York, USA
| | - Utpal B. Pajvani
- Department of Medicine, Columbia University, New York, New York, USA
- Institute of Human Nutrition, New York, New York, USA
- Columbia University Digestive and Liver Disease Research Center, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, New York, New York, USA
| | - Robert F. Schwabe
- Department of Medicine, Columbia University, New York, New York, USA
- Institute of Human Nutrition, New York, New York, USA
- Columbia University Digestive and Liver Disease Research Center, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, New York, New York, USA
| |
Collapse
|
45
|
Mao Q, Lin B, Zhang W, Zhang Y, Zhang Y, Cao Q, Xu M. Understanding the role of ursodeoxycholic acid and gut microbiome in non-alcoholic fatty liver disease: current evidence and perspectives. Front Pharmacol 2024; 15:1371574. [PMID: 38576492 PMCID: PMC10991717 DOI: 10.3389/fphar.2024.1371574] [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: 01/23/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, resulting in a huge medical burden worldwide. Accumulating evidence suggests that the gut microbiome and bile acids play pivotal roles during the development of NAFLD. Patients with NAFLD exhibit unique signatures of the intestinal microbiome marked by the priority of Gram-negative bacteria, decreased ratio of Firmicutes/Bacteroidetes (F/B), and increased Prevotella and Lachnospiraceae. The intestinal microbiota is involved in the metabolism of bile acids. Ursodeoxycholic acid (UDCA) is a key determinant in maintaining the dynamic communication between the host and gut microbiota. It generally shows surprising therapeutic potential in NAFLD with several mechanisms, such as improving cellular autophagy, apoptosis, and mitochondrial functions. This action is based on its direct or indirect effect, targeting the farnesoid X receptor (FXR) and various other nuclear receptors. This review aims to discuss the current studies on the involvement of the microbiome-UDCA interface in NAFLD therapy and provide prospective insights into future preventative and therapeutic approaches for NAFLD.
Collapse
Affiliation(s)
- Qingyi Mao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Beibei Lin
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wenluo Zhang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yu Zhang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yu Zhang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Qian Cao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Mengque Xu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| |
Collapse
|
46
|
Suda N, Bartolomé A, Liang J, Son J, Yagishita Y, Siebel C, Accili D, Ding H, Pajvani UB. β-cell Jagged1 is sufficient but not necessary for islet Notch activity and insulin secretory defects in obese mice. Mol Metab 2024; 81:101894. [PMID: 38311286 PMCID: PMC10877406 DOI: 10.1016/j.molmet.2024.101894] [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: 11/22/2023] [Revised: 01/22/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024] Open
Abstract
OBJECTIVE Notch signaling, re-activated in β cells from obese mice and causal to β cell dysfunction, is determined in part by transmembrane ligand availability in a neighboring cell. We hypothesized that β cell expression of Jagged1 determines the maladaptive Notch response and resultant insulin secretory defects in obese mice. METHODS We assessed expression of Notch pathway components in high-fat diet-fed (HFD) or leptin receptor-deficient (db/db) mice, and performed single-cell RNA sequencing (scRNA-Seq) in islets from patients with and without type 2 diabetes (T2D). We generated and performed glucose tolerance testing in inducible, β cell-specific Jagged1 gain-of- and loss-of-function mice. We also tested effects of monoclonal neutralizing antibodies to Jagged1 in glucose-stimulated insulin secretion (GSIS) assays in isolated islets. RESULTS Jag1 was the only Notch ligand that tracked with increased Notch activity in HFD-fed and db/db mice, as well as in metabolically-inflexible β cells enriched in patients with T2D. Neutralizing antibodies to block Jagged1 in islets isolated from HFD-fed and db/db mice potentiated GSIS ex vivo. To demonstrate if β cell Jagged1 is sufficient to cause glucose tolerance in vivo, we generated inducible β cell-specific Jag1 transgenic (β-Jag1TG) and loss-of-function (iβ-Jag1KO) mice. While forced Jagged1 impaired glucose intolerance due to reduced GSIS, loss of β cell Jagged1 did not protect against HFD-induced insulin secretory defects. CONCLUSIONS Jagged1 is increased in islets from obese mice and in patients with T2D, and neutralizing Jagged1 antibodies lead to improved GSIS, suggesting that inhibition of Jagged1-Notch signaling may have therapeutic benefit. However, genetic loss-of-function experiments suggest that β cells are not a likely source of the Jagged1 signal.
Collapse
Affiliation(s)
- Nina Suda
- Department of Medicine, Columbia University, New York, NY, USA
| | | | - Jiani Liang
- Department of Medicine, Columbia University, New York, NY, USA
| | - Jinsook Son
- Department of Medicine, Columbia University, New York, NY, USA
| | - Yoko Yagishita
- Department of Medicine, Columbia University, New York, NY, USA
| | - Christian Siebel
- Department of Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Domenico Accili
- Department of Medicine, Columbia University, New York, NY, USA
| | - Hongxu Ding
- Department of Pharmacy Practice & Science, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Utpal B Pajvani
- Department of Medicine, Columbia University, New York, NY, USA.
| |
Collapse
|
47
|
Shao C, Xu H, Sun X, Pan Y, Liang X, Huang J, He Y, Guo W, Ye L, Zhang J. Jiawei Taohe Chengqi decoction inhibition of the notch signal pathway affects macrophage reprogramming to inhibit HSCs activation for the treatment of hepatic fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117486. [PMID: 38030027 DOI: 10.1016/j.jep.2023.117486] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/12/2023] [Accepted: 11/20/2023] [Indexed: 12/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiawei Taohe Chengqi Tang (JTCD) is a modified formulation of Traditional Chinese Medicine (TCM) known as Taohe Chengqi Decoction, which has been described in the ancient TCM literature "Treatise on Febrile Diseases". As a formula that can activate blood circulation and eliminate blood stasis and regulate Yin and Yang in traditional Chinese medicine applications, JTCD has been reported to be effective in the treatment of chronic liver disease and hepatic fibrosis (HF). AIM OF STUDY The current study aimed to evaluate the effectiveness of JTCD in modulating hepatic macrophages by regulating the Notch signal pathway, and to further investigate the mechanisms underlying macrophage reprogramming that leads to HF. MATERIALS AND METHODS Molecular assays were performed using in vitro cultures of human mononuclear THP-1 cells and human-derived hepatic stellate cells LX-2. CCl4-induced mice were utilized as an in vivo model to simulate HF. RESULTS Our results demonstrated that JTCD exhibited dual effects by inhibiting hepatic stellate cell (HSCs) activation and modulating the polarisation of macrophages towards the M2 phenotype while decreasing the M1 phenotype. Network pharmacological analyses and molecular docking studies revealed that the Notch signal pathway was significantly enriched and played a crucial role in the therapeutic response of JTCD against HF. Moreover, through the establishment of a co-culture model, we validated that JTCD inhibited the Notch signal pathway in macrophages, leading to alterations in macrophage reprogramming, subsequent inhibition of HSC activation, and ultimately exerting anti-HF effects. CONCLUSION In conclusion, our findings provide solid evidence for JTCD in treating HF, as it suppresses the Notch signal pathway in macrophages, regulates macrophage reprogramming, and inhibits HSC activation.
Collapse
Affiliation(s)
- Chang Shao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Huihui Xu
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Xiguang Sun
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Yun Pan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Xiaofan Liang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Jiaxin Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Yi He
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Wenqin Guo
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Linmao Ye
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Junjie Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
48
|
Kei N, Cheung KK, Ma KL, Yau TK, Lauw S, Wong VWS, You L, Cheung PCK. Effects of Oat β-Glucan and Inulin on Alleviation of Nonalcoholic Steatohepatitis Aggravated by Circadian Disruption in C57BL/6J Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:3520-3535. [PMID: 38333950 DOI: 10.1021/acs.jafc.3c08028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
This was the first study that examined the effects of oat β-glucan and inulin on diet-induced nonalcoholic steatohepatitis (NASH) in circadian-disrupted (CD)-male C57BL/6J mice. CD intensified NASH, significantly increasing alanine aminotransferase and upregulating hepatic tumor necrosis factor α (TNFα) and transforming growth factor β 1 (TGFβ1). However, these observations were significantly alleviated by oat β-glucan and inulin treatments. Compared to CD NASH mice, oat β-glucan significantly decreased the liver index, aspartate aminotransferase (AST), and insulin. In prebiotic-treated and CD NASH mice, significant negative correlations were found between enrichment of Muribaculaceae bacterium Isolate-036 (Harlan), Muribaculaceae bacterium Isolate-001 (NCI), and Bacteroides ovatus after oat β-glucan supplementation with TNFα and TGFβ1 levels; and enrichment of Muribaculaceae bacterium Isolate-110 (HZI) after inulin supplementation with AST level. In conclusion, oat β-glucan and inulin exhibited similar antiliver injury, anti-inflammatory, and antifibrotic activities but had no effect on cecal short-chain fatty acids and gut microbiota diversity in CD NASH mice.
Collapse
Affiliation(s)
- Nelson Kei
- Food and Nutritional Sciences Program, School of Life Sciences, The Chinese University of Hong Kong, New Territories, Hong Kong SAR, China
| | - Kam Kuen Cheung
- Food and Nutritional Sciences Program, School of Life Sciences, The Chinese University of Hong Kong, New Territories, Hong Kong SAR, China
- Food Research Centre, The Chinese University of Hong Kong, New Territories, Hong Kong SAR , China
| | - Ka Lee Ma
- Food and Nutritional Sciences Program, School of Life Sciences, The Chinese University of Hong Kong, New Territories, Hong Kong SAR, China
| | - Tsz Kwan Yau
- Cell and Molecular Biology Program, School of Life Sciences, The Chinese University of Hong Kong, New Territories, Hong Kong SAR, China
| | - Susana Lauw
- Food and Nutritional Sciences Program, School of Life Sciences, The Chinese University of Hong Kong, New Territories, Hong Kong SAR, China
- Food Research Centre, The Chinese University of Hong Kong, New Territories, Hong Kong SAR , China
| | - Vincent Wai Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, New Territories, Hong Kong SAR, China
| | - Lijun You
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Peter Chi Keung Cheung
- Food and Nutritional Sciences Program, School of Life Sciences, The Chinese University of Hong Kong, New Territories, Hong Kong SAR, China
- Food Research Centre, The Chinese University of Hong Kong, New Territories, Hong Kong SAR , China
| |
Collapse
|
49
|
Wang X, Zhang L, Dong B. Molecular mechanisms in MASLD/MASH-related HCC. Hepatology 2024:01515467-990000000-00739. [PMID: 38349726 PMCID: PMC11323288 DOI: 10.1097/hep.0000000000000786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/16/2024] [Indexed: 03/23/2024]
Abstract
Liver cancer is the third leading cause of cancer-related deaths and ranks as the sixth most prevalent cancer type globally. NAFLD or metabolic dysfunction-associated steatotic liver disease, and its more severe manifestation, NASH or metabolic dysfunction-associated steatohepatitis (MASH), pose a significant global health concern, affecting approximately 20%-25% of the population. The increased prevalence of metabolic dysfunction-associated steatotic liver disease and MASH is parallel to the increasing rates of obesity-associated metabolic diseases, including type 2 diabetes, insulin resistance, and fatty liver diseases. MASH can progress to MASH-related HCC (MASH-HCC) in about 2% of cases each year, influenced by various factors such as genetic mutations, carcinogen exposure, immune microenvironment, and microbiome. MASH-HCC exhibits distinct molecular and immune characteristics compared to other causes of HCC and affects both men and women equally. The management of early to intermediate-stage MASH-HCC typically involves surgery and locoregional therapies, while advanced HCC is treated with systemic therapies, including anti-angiogenic therapies and immune checkpoint inhibitors. In this comprehensive review, we consolidate previous research findings while also providing the most current insights into the intricate molecular processes underlying MASH-HCC development. We delve into MASH-HCC-associated genetic variations and somatic mutations, disease progression and research models, multiomics analysis, immunological and microenvironmental impacts, and discuss targeted/combined therapies to overcome immune evasion and the biomarkers to recognize treatment responders. By furthering our comprehension of the molecular mechanisms underlying MASH-HCC, our goal is to catalyze the advancement of more potent treatment strategies, ultimately leading to enhanced patient outcomes.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Liang Zhang
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bingning Dong
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
50
|
Chen L, Guo W, Mao C, Shen J, Wan M. Liver fibrosis: pathological features, clinical treatment and application of therapeutic nanoagents. J Mater Chem B 2024; 12:1446-1466. [PMID: 38265305 DOI: 10.1039/d3tb02790b] [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: 01/25/2024]
Abstract
Liver fibrosis is a reversible damage-repair response, the pathological features of which mainly include damage to hepatocytes, sinusoid capillarization, hepatic stellate cells activation, excessive accumulation of extracellular matrix and inflammatory response. Although some treatments (including drugs and stem cell therapy) for these pathological features have been shown to be effective, more clinical trials are needed to confirm their effectiveness. In recent years, nanomaterials-based therapies have emerged as an innovative and promising alternative to traditional drugs, being explored for the treatment of liver fibrosis diseases. Natural nanomaterials (including extracellular vesicles) and synthetic nanomaterials (including inorganic nanomaterials and organic nanomaterials) are developed to facilitate drug targeting delivery and combination therapy. In this review, the pathological features of liver fibrosis and the current anti-fibrosis drugs in clinical trials are briefly introduced, followed by a detailed introduction of the therapeutic nanoagents for the precise delivery of anti-fibrosis drugs. Finally, the future development trend in this field is discussed.
Collapse
Affiliation(s)
- Lin Chen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Wenyan Guo
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|