1
|
Jiang J, Liu Y, Yang H, Ma Z, Liu W, Zhao M, Peng X, Qin X, Xia Y. Dietary fiber intake, genetic predisposition of gut microbiota, and the risk of metabolic dysfunction-associated steatotic liver disease. Food Res Int 2025; 211:116497. [PMID: 40356189 DOI: 10.1016/j.foodres.2025.116497] [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: 06/17/2024] [Revised: 04/16/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025]
Abstract
This study aimed to explore the association between dietary fiber intake and the risk of metabolic dysfunction-associated steatotic liver disease (MASLD), as well as liver fat content, while considering genetic predispositions of MASLD, gut microbial abundance, and butyrate levels. This study analyzed data from 190,276 participants in the UK Biobank. Dietary fiber intake was assessed using 24-h dietary recall. MASLD cases were diagnosed through hospital admission records and death registries, and liver fat content was measured via magnetic resonance imaging. The genetic predispositions of MASLD, gut microbial abundance, and butyrate levels were evaluated using single nucleotide polymorphisms. Cox proportional hazards models were used to calculate hazard ratios (HRs) and 95 % confidence intervals (CIs). Over a median follow-up of 10.49 years, 1423 MASLD cases were recorded. Elevated dietary fiber intake was associated with a reduced risk of MASLD (HR: 0.72; 95 % CI: 0.58, 0.90) and a lower level of liver fat content (β: -0.97; 95 % CI: -1.21, -0.73) (all P for trend <0.05). Restricted cubic spline analyses further confirmed the linear inverse associations between fiber intake and the risk of MASLD. Notably, the negative associations between dietary fiber intake and both MASLD and liver fat content were consistent across different genetic predispositions of gut microbial abundance and butyrate levels. Moreover, the inverse association between dietary fiber intake and liver fat was strengthened by high genetic susceptibility of MASLD and elevated body mass index (both P for interaction <0.05). Overall, increased dietary fiber consumption was associated with a lower MASLD risk and decreased liver fat content regardless of genetic predispositions of gut microbial abundance and butyrate levels.
Collapse
Affiliation(s)
- Jinguo Jiang
- School of Public Health, Shenyang Medical College, Shenyang, China; Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China.
| | - Yang Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, 38# Xueyuan Road, Haidian District, Beijing 100191, China.
| | - Honghao Yang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China; Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Liaoning Province, Shenyang, China.
| | - Zheng Ma
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China; Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Liaoning Province, Shenyang, China.
| | - Wenqi Liu
- School of Public Health, Shenyang Medical College, Shenyang, China; Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China.
| | - Maoxiang Zhao
- Interventional Center of Valvular Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing 100011, China.
| | - Xinyi Peng
- Hypertension Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Center for Cardiovascular Diseases of China, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China.
| | - Xueying Qin
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, 38# Xueyuan Road, Haidian District, Beijing 100191, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, China.
| | - Yang Xia
- School of Public Health, Shenyang Medical College, Shenyang, China; Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
2
|
Jung YS, Radhakrishnan K, Noh JR, Kim YH, Lee CH, Choi HS. Hepatic estrogen-related receptor gamma is a key regulator of GDF15 production in acute and chronic liver injury. Mol Cell Endocrinol 2025; 606:112572. [PMID: 40379080 DOI: 10.1016/j.mce.2025.112572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 04/21/2025] [Accepted: 05/14/2025] [Indexed: 05/19/2025]
Abstract
AIMS Growth differentiation factor 15 (GDF15) is a stress-induced hepatokine with emerging roles in liver injury. Estrogen-related receptor γ (ERRγ), a nuclear receptor regulating mitochondrial function and metabolic stress, has also been implicated in various liver injury conditions. However, the regulatory interplay between ERRγ and GDF15 remains unclear. This study investigates the molecular mechanisms underlying GDF15 expression and secretion in the liver, focusing on the role of ERRγ during acute and chronic liver injury. MATERIALS AND METHODS Wild-type and hepatocyte-specific ERRγ knockout (ERRγ-LKO) mice were administered with a single dose of carbon tetrachloride (CCl4) or fed an alcohol-containing diet for 4 weeks to establish acute or chronic liver injury models, respectively. ERRγ was overexpressed through an adenoviral construct (Ad-ERRγ). The ERRγ-specific inverse agonist GSK5182 was employed to inhibit the transactivation of ERRγ. The luciferase reporter assays were used to assess the binding of ERRγ protein to the regulatory region of GDF15 gene. KEY FINDINGS Hepatic ERRγ and GDF15 gene expression, and GDF15 protein secretion were significantly elevated in both acute and chronic liver injury. Adenovirus-mediated overexpression of ERRγ is sufficient to substantially increase hepatic GDF15 expression and secretion. Genetic ablation of ERRγ expression or pharmacological inhibition of ERRγ transactivation substantially inhibited the upregulation of hepatic GDF15 expression and production in both acute and chronic liver injury. Furthermore, reporter assays showed that ERRγ, but not ERRα or ERRβ, directly binds to and activates the GDF15 gene promoter. SIGNIFICANCE Our findings highlight the crucial role of ERRγ in transcriptional regulation of GDF15 gene expression and production in response to liver damage. Understanding the regulatory mechanisms of GDF15 expression could lead to new therapeutic targets for protecting the liver from various types of injuries and associated diseases.
Collapse
Affiliation(s)
- Yoon Seok Jung
- Host-Directed Antiviral Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Kamalakannan Radhakrishnan
- Host-Directed Antiviral Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jung-Ran Noh
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Yong-Hoon Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| | - Hueng-Sik Choi
- Host-Directed Antiviral Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
3
|
Schultheiß C, Binder M. nACh receptors engage a nice ChAT with the liver: B cells serving up regeneration. Immunity 2025; 58:1177-1179. [PMID: 40367919 DOI: 10.1016/j.immuni.2025.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/16/2025]
Abstract
Liver regeneration is a remarkable and unique biological process, orchestrated by an intricate cellular crosstalk of (non-)parenchymal, immune, and nerve cells. In this issue of Immunity, Modares et al. uncover the pivotal role of choline acetyltransferase (ChAT+)-expressing B cells as key orchestrators of liver regeneration.
Collapse
Affiliation(s)
- Christoph Schultheiß
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland; Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland; Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland; Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg, Germany.
| |
Collapse
|
4
|
Abhinaya SV, Garg N, Sindwani G, Arora MK, Thapar S, Thomas S, Pamecha V, Sarin SK. Comparison of the effect of isoflurane and propofol on liver regeneration after donor hepatectomy - A randomized controlled trial. J Clin Anesth 2025; 104:111859. [PMID: 40347559 DOI: 10.1016/j.jclinane.2025.111859] [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: 08/06/2024] [Revised: 03/12/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND The biotransformation of inhalational anesthetic agents can result in production of hepatotoxic metabolites, which may potentially impair liver regeneration. Thus, isoflurane and propofol may have differential impacts on liver regeneration after donor hepatectomy. STUDY OBJECTIVE To compare the regeneration liver volume (RgLV) via computed tomography (CT) volumetry on post-operative day (POD) 14 in isoflurane and propofol groups. DESIGN Randomized controlled pilot trial. PATIENTS Sixty donors, who underwent donor hepatectomy. INTERVENTIONS Patients were randomized into isoflurane and propofol group using computer generated random number tables. In isoflurane group, anesthesia was maintained with isoflurane 1-2 % in air‑oxygen mixture. In propofol group, anesthesia was maintained with the target-controlled infusion (TCI) of propofol using a TCI system (Perfusor® Space- B. Braun) at a plasma target concentration of 3-6 μg.ml-1. BIS was recorded in all patients. MEASUREMENTS Liver CT volumetry was assessed at POD 14. MAIN RESULTS RgLV on POD14 was comparable in two groups [449.3(170) & 437.8 (177.8) cm3, [Mean Difference (MD) -11.904 95 % Confidence Interval(CI) -101.83, 78.03 respectively; p = 0.79]. CONCLUSION Administering propofol or isoflurane may not have differential effect on liver regeneration after donor hepatectomy.
Collapse
Affiliation(s)
- S V Abhinaya
- Department of Anaesthesia and Critical care, Institute of liver and biliary sciences, New Delhi, India
| | - Neha Garg
- Department of Anaesthesia, Institute of liver and biliary sciences, New Delhi, India.
| | - Gaurav Sindwani
- Department of Anaesthesia, Institute of liver and biliary sciences, New Delhi, India
| | - Mahesh Kumar Arora
- Department of Anaesthesia, Institute of liver and biliary sciences, New Delhi, India
| | - Shalini Thapar
- Department of Radiology, Institute of liver and biliary sciences, New Delhi, India
| | - Sherin Thomas
- Department of Biochemistry, Institute of liver and biliary sciences, New Delhi, India
| | - Viniyendra Pamecha
- Department of HPB Surgery, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shiv Kumar Sarin
- Department of Hepatology, Director and Chancellor, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
5
|
Zou Y, Wu J, Cheng S, Cheng D, Chen T, Guo X, Tang L, Su X, Zhang M, Zhang X, Liu Y, Zhang J, Bao Q, Hou S, Sun P, Li Y, Han B. Hepatic stellate cell-specific Kcnma1 deletion mitigates metabolic dysfunction-associated steatotic liver disease progression via upregulating Amphiregulin secretion. Mol Metab 2025; 97:102164. [PMID: 40348016 DOI: 10.1016/j.molmet.2025.102164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/25/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
OBJECTIVE Metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing global health concern, with limited effective treatments. KCNMA1 potassium channel has been implicated in the pathogenesis of various metabolic diseases. However, whether and how KCNMA1 regulates MASLD have been elusive. METHODS Global, hepatic stellate cells (HSCs)-specific, and hepatocyte-specific Kcnma1 knockout mice were fed either a standard chow or a high-fat diet (HFD). Serum and liver tissues were collected and analyzed by biochemical assay, histology, qPCR and western blotting. HSCs conditioned medium (CM) treatment hepatocytes experiment model and three-dimensional (3D) hepatocytes-HSCs spheroids were employed to study lipid accumulation in hepatocytes. A Cytokine Antibody Array was used to analyze the cytokine profile. RESULTS Our study demonstrated that global Kcnma1 deletion prevented diet-induced hepatic steatosis and improved insulin sensitivity. Further analyses using HSC-specific and hepatocyte-specific Kcnma1 knockout MASLD mouse models revealed that the protective effect against hepatic steatosis was predominantly mediated by Kcnma1 deletion in HSCs, rather than in hepatocytes. CM transfer experiment and 3D spheroid studies show Kcnma1 deletion effectively prevents lipid accumulation in hepatocytes. Mechanically, Kcnma1-deficient HSCs secrete Amphiregulin (AREG) to regulate lipid metabolism in hepatocytes via epidermal growth factor receptor (EGFR) signaling. Of clinical significance, AREG levels were notably reduced in the liver tissue of MASLD patients, while injection of recombinant AREG protein significantly ameliorated MASLD in mice. CONCLUSIONS Our study uncovers a novel mechanism in which Kcnma1 deletion in HSCs enhances AREG secretion, thereby reducing lipid accumulation in hepatocytes through the AREG/EGFR signaling, ultimately inhibiting the progression of MASLD.
Collapse
Affiliation(s)
- Yunhan Zou
- Hongqiao International Institute of Medicine, Tongren Hospital and Faculty of Basic Medicine, Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiaoxiang Wu
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China; Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Sheng Cheng
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China; Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Daqing Cheng
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Taoying Chen
- School of Food Science and Engineering, Guiyang University, Guizhou, 550005, China
| | - Xirong Guo
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Li Tang
- Rehabilitation Department, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200336, China
| | - Xianbin Su
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Man Zhang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xin Zhang
- Institute of Translational Medicine, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ying Liu
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Jin Zhang
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Qun Bao
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Shangwei Hou
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China.
| | - Peng Sun
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China.
| | - Yong Li
- Hongqiao International Institute of Medicine, Tongren Hospital and Faculty of Basic Medicine, Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Bo Han
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China; Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China.
| |
Collapse
|
6
|
Zhan X, Bai Y, Zhu Q, Gao Y, Li F, Bu Q, Zhu Z, Rao Z, Zhou H. Macrophage ATG16L1 promotes liver regeneration after partial hepatectomy. JHEP Rep 2025; 7:101330. [PMID: 40290519 PMCID: PMC12023798 DOI: 10.1016/j.jhepr.2025.101330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 04/30/2025] Open
Abstract
Background & Aims Autophagy plays an important role in liver regeneration. However, most studies are limited to hepatocytes, and the function and mechanism of macrophage autophagy in liver regeneration remain unclear. This study investigated the role of the essential autophagy gene encoding autophagy-related 16-like 1 (ATG16L1), which regulates the macrophage phenotype in liver regeneration. Methods We generated FloxP-Atg16l1 (Atg16l1 FL/FL ), Lyz2-Cre Atg16l1 knockout (KO) (Atg16l1 M-KO ), and myeloid-specific Atg16l1-overexpression-knock-in (Atg16l1 OE ) mice. These mice were subjected to 70% partial hepatectomy to demonstrate the role of ATG16L1 in macrophages during liver regeneration. Results ATG16L1 expression was significantly upregulated in macrophages during the early stages of liver regeneration. ATG16L1 deletion in macrophages substantially delayed liver regeneration in mice and caused a marked imbalance in Ly6Chi and Ly6Clo macrophage proportions in the liver. RNA-sequencing analysis revealed that, compared with macrophages isolated from Atg16l1 FL/FL mice, those from Atg16l1 M-KO mice exhibited significant downregulation of genes associated with oxidative phosphorylation and upregulation of proinflammatory gene expression. Mechanistically, ATG16L1 loss impaired mitophagy in macrophages, leading to the accumulation of mitochondrial damage and a metabolic shift that promoted proinflammatory macrophage polarization. ATG16L1 deficiency not only promoted macrophage mitochondrial (mt)DNA release and cyclic GMP-AMP synthase-stimulator of interferon genes (STING) activation, but also suppressed STING degradation. Sustained STING hyperactivation and subsequent increased release of downstream interferons further contributed to the inhibition of liver regeneration. Notably, pharmacological activation or genetic overexpression of ATG16L1 significantly enhanced liver regeneration in mice. Conclusions ATG16L1 has a pivotal role in liver regeneration by affecting the phenotype and function of macrophages. Thus, targeting ATG16L1 in macrophages could present a novel strategy for promoting liver regeneration. Impact and implications The autophagy-related gene ATG16L1 mediates mitophagy, facilitating the clearance of damaged mitochondria in macrophages following partial hepatectomy and maintaining a reparative macrophage phenotype. ATG16L1 deficiency triggers excessive STING activation and inhibits its degradation, thereby suppressing liver regeneration. Thus, targeting ATG16L1 in macrophages could represent a novel strategy to promote liver regeneration.
Collapse
Affiliation(s)
- Xinyu Zhan
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Yan Bai
- Department of Anesthesiology, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, 210029, China
| | - Qing Zhu
- Department of Anesthesiology, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, 210029, China
| | - Yiyun Gao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Fan Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Qingfa Bu
- Department of General Surgery, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zeyu Zhu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Zhuqing Rao
- Department of Anesthesiology, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, 210029, China
| | - Haoming Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| |
Collapse
|
7
|
Vosough M, Shokouhian B, Sharbaf MA, Solhi R, Heidari Z, Seydi H, Hassan M, Devaraj E, Najimi M. Role of mitogens in normal and pathological liver regeneration. Hepatol Commun 2025; 9:e0692. [PMID: 40304568 PMCID: PMC12045551 DOI: 10.1097/hc9.0000000000000692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/31/2025] [Indexed: 05/02/2025] Open
Abstract
The liver has a unique ability to regenerate to meet the body's metabolic needs, even following acute or chronic injuries. The cellular and molecular mechanisms underlying normal liver regeneration have been well investigated to improve organ transplantation outcomes. Once liver regeneration is impaired, pathological regeneration occurs, and the underlying cellular and molecular mechanisms require further investigations. Nevertheless, a plethora of cytokines and growth factor-mediated pathways have been reported to modulate physiological and pathological liver regeneration. Regenerative mitogens play an essential role in hepatocyte proliferation. Accelerator mitogens in synergism with regenerative ones promote liver regeneration following hepatectomy. Finally, terminator mitogens restore the proliferating status of hepatocytes to a differentiated and quiescent state upon completion of regeneration. Chronic loss of hepatocytes, which can manifest in chronic liver disorders of any etiology, often has undesired structural consequences, including fibrosis, cirrhosis, and liver neoplasia due to the unregulated proliferation of remaining hepatocytes. In fact, any impairment in the physiological function of the terminator mitogens results in the progression of pathological liver regeneration. In the current review, we intend to highlight the updated cellular and molecular mechanisms involved in liver regeneration and discuss the impairments in central regulating mechanisms responsible for pathological liver regeneration.
Collapse
Affiliation(s)
- Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bahare Shokouhian
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Amin Sharbaf
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Roya Solhi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Heidari
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Homeyra Seydi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ezhilarasan Devaraj
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| |
Collapse
|
8
|
De Rudder M, Manco R, Coubeau L, Fontaine A, Bertrand C, Leclercq IA, Dili A. Vascular damage and excessive proliferation compromise liver function after extended hepatectomy in mice. Hepatology 2025; 81:1468-1484. [PMID: 38661628 DOI: 10.1097/hep.0000000000000900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/27/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND AND AIMS Surgical resection remains the gold standard for liver tumor treatment, yet the emergence of postoperative liver failure, known as the small-for-size syndrome (SFSS), poses a significant challenge. The activation of hypoxia sensors in an SFSS liver remnant initiated early angiogenesis, improving the vascular architecture, safeguarding against liver failure, and reducing mortality. The study aimed to elucidate vascular remodeling mechanisms in SFSS and their impact on hepatocyte function and subsequent liver failure. APPROACH AND RESULTS Mice underwent extended partial hepatectomy to induce SFSS, with a subset exposed to hypoxia immediately after surgery. Hypoxia bolstered posthepatectomy survival rates. The early proliferation of liver sinusoidal cells, coupled with recruitment of putative endothelial progenitor cells, increased vascular density, improved lobular perfusion, and limited hemorrhagic events in the regenerating liver under hypoxia. Administration of granulocyte colony-stimulating factor in hepatectomized mice mimicked the effects of hypoxia on vascular remodeling and endothelial progenitor cell recruitment but failed to rescue survival. Compared to normoxia, hypoxia favored hepatocyte function over proliferation, promoting functional preservation in the regenerating remnant. Injection of Adeno-associated virus serotype 8-thyroxine-binding globulin-hepatocyte nuclear factor 4 alpha virus for hepatocyte-specific overexpression of hepatocyte nuclear factor 4 alpha, the master regulator of hepatocyte function, enforced functionality in proliferating hepatocytes but did not rescue survival. The combination of hepatocyte nuclear factor 4 alpha overexpression and granulocyte colony-stimulating factor treatment rescued survival after SFSS-setting hepatectomy. CONCLUSIONS In summary, SFSS arises from an imbalance and desynchronized interplay between functional regeneration and vascular restructuring. To improve survival following SFSS hepatectomy, it is essential to adopt a 2-pronged strategy aimed at preserving the function of proliferating parenchymal cells and simultaneously attenuating vascular damage.
Collapse
Affiliation(s)
- Maxime De Rudder
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Rita Manco
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Laurent Coubeau
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Surgery, University Clinics of St Luc, UCLouvain, Brussels, Belgium
| | - Alix Fontaine
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Claude Bertrand
- Department of Surgery, University Hospital of UCLouvain-Namur, Site of Godinne, Yvoir, Belgium
| | - Isabelle A Leclercq
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Alexandra Dili
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Surgery, University Hospital of UCLouvain-Namur, Site of Godinne, Yvoir, Belgium
| |
Collapse
|
9
|
Ortuño-Costela MC, Pinzani M, Vallier L. Cell therapy for liver disorders: past, present and future. Nat Rev Gastroenterol Hepatol 2025; 22:329-342. [PMID: 40102584 DOI: 10.1038/s41575-025-01050-2] [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: 02/11/2025] [Indexed: 03/20/2025]
Abstract
The liver fulfils a plethora of vital functions and, due to their importance, liver dysfunction has life-threatening consequences. Liver disorders currently account for more than two million deaths annually worldwide and can be classified broadly into three groups, considering their onset and aetiology, as acute liver diseases, inherited metabolic disorders and chronic liver diseases. In the most advanced and severe forms leading to liver failure, liver transplantation is the only treatment available, which has many associated drawbacks, including a shortage of organ donors. Cell therapy via fully mature cell transplantation is an advantageous alternative that may be able to restore a damaged organ's functionality or serve as a bridge until regeneration can occur. Pioneering work has shown that transplanting adult hepatocytes can support liver recovery. However, primary hepatocytes cannot be grown extensively in vitro as they rapidly lose their metabolic activity. Therefore, different cell sources are currently being tested as alternatives to primary cells. Human pluripotent stem cell-derived cells, chemically induced liver progenitors, or 'liver' organoids, hold great promise for developing new cell therapies for acute and chronic liver diseases. This Review focuses on the advantages and drawbacks of distinct cell sources and the relative strategies to address different therapeutic needs in distinct liver diseases.
Collapse
Affiliation(s)
- M Carmen Ortuño-Costela
- Berlin Institute of Health, BIH Centre for Regenerative Therapies, Charité-Universitätsmedizin, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Massimo Pinzani
- University College London Institute for Liver and Digestive Health, Division of Medicine, Royal Free Hospital, London, UK
- University of Pittsburgh Medical Center-Mediterranean Institute for Transplantation and Highly Specialized Therapies (UPMC-ISMETT), Palermo, Italy
| | - Ludovic Vallier
- Berlin Institute of Health, BIH Centre for Regenerative Therapies, Charité-Universitätsmedizin, Berlin, Germany.
- Max Planck Institute for Molecular Genetics, Berlin, Germany.
| |
Collapse
|
10
|
Lee LR, Guillotin B, Rahni R, Hutchison C, Desvoyes B, Gutierrez C, Birnbaum KD. Glutathione accelerates the cell cycle and cellular reprogramming in plant regeneration. Dev Cell 2025; 60:1153-1167.e6. [PMID: 39755116 DOI: 10.1016/j.devcel.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/01/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025]
Abstract
The plasticity of plant cells underlies their wide capacity to regenerate, with increasing evidence in plants and animals implicating cell-cycle dynamics in cellular reprogramming. To investigate the cell cycle during cellular reprogramming, we developed a comprehensive set of cell-cycle-phase markers in the Arabidopsis root. Using single-cell RNA sequencing profiles and live imaging during regeneration, we found that a subset of cells near an ablation injury dramatically increases division rate by truncating G1 phase. Cells in G1 undergo a transient nuclear peak of glutathione (GSH) prior to coordinated entry into S phase, followed by rapid divisions and cellular reprogramming. A symplastic block of the ground tissue impairs regeneration, which is rescued by exogenous GSH. We propose a model in which GSH from the outer tissues is released upon injury, licensing an exit from G1 near the wound to induce rapid cell division and reprogramming.
Collapse
Affiliation(s)
- Laura R Lee
- New York University, Center for Genomics and Systems Biology, Department of Biology, New York, NY 10003, USA
| | - Bruno Guillotin
- New York University, Center for Genomics and Systems Biology, Department of Biology, New York, NY 10003, USA
| | - Ramin Rahni
- New York University, Center for Genomics and Systems Biology, Department of Biology, New York, NY 10003, USA
| | - Chanel Hutchison
- New York University, Center for Genomics and Systems Biology, Department of Biology, New York, NY 10003, USA
| | | | | | - Kenneth D Birnbaum
- New York University, Center for Genomics and Systems Biology, Department of Biology, New York, NY 10003, USA.
| |
Collapse
|
11
|
Junrungsee S, Vipudhamorn W, Lapisatepun W, Thepbunchonchai A, Chotirosniramit A, Lapisatepun W, Ko-Iam W. Portal flow modulation by splenic artery ligation to prevent posthepatectomy liver failure: A randomized controlled trial. Surgery 2025:109351. [PMID: 40204604 DOI: 10.1016/j.surg.2025.109351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Posthepatectomy liver failure is a serious clinical issue with high mortality, similar in pathophysiology to small-for-size syndrome seen in liver transplantation. This study evaluates the efficacy of splenic artery ligation in reducing posthepatectomy liver failure in patients with portal venous pressure >15 mm Hg after hepatectomy. METHODS This single-center, randomized controlled trial was conducted from May 2019 to November 2023. Eligible participants were patients scheduled for open hepatectomy for any indication. Patients with a portal venous pressure >15 mm Hg were randomized into splenic artery ligation and control groups in a 1:1 ratio. The primary outcomes were posthepatectomy liver failure grades B and C (International Study group of Liver Surgery criteria), and secondary outcomes included 90-day mortality, comprehensive complication index, and ascites volume. RESULTS The study was terminated early, before reaching the calculated sample size, because the primary outcome in the intervention group demonstrated statistically significant results. Of the 92 patients, 36 had elevated portal venous pressure, which was associated with greater rates of posthepatectomy liver failure grades B and C (41.67% vs 3.57%, P < .001), increased ascites volume (5,340 mL vs 1,055 mL, P < .001), and a greater comprehensive complication index (20.90 vs 8.70, P < .001). In the randomized subset, splenic artery ligation significantly reduced portal venous pressure and the portal venous pressure-central venous pressure gradient compared with both presplenic artery ligation values and the control group and significantly lowered the incidence of posthepatectomy liver failure grades B and C (16.67% vs 66.67%, P = .006), comprehensive complication index (8.70 vs 20.90, P = .034). Splenic artery ligation was identified as an independent factor in reducing posthepatectomy liver failure (adjusted relative risk, 0.29). CONCLUSION Splenic artery ligation is effective in reducing posthepatectomy liver failure in patients with high portal venous pressure after hepatectomy.
Collapse
Affiliation(s)
- Sunhawit Junrungsee
- Division of Hepatobiliary Pancreatic Surgery, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Clinical Surgical Research Center, Chiang Mai University, Chiang Mai, Thailand.
| | - Witcha Vipudhamorn
- Division of Colorectal Surgery, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Worakitti Lapisatepun
- Division of Hepatobiliary Pancreatic Surgery, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand. https://twitter.com/WallEe19880
| | - Asara Thepbunchonchai
- Division of Hepatobiliary Pancreatic Surgery, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand. https://twitter.com/AsaraOum
| | - Anon Chotirosniramit
- Division of Hepatobiliary Pancreatic Surgery, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Warangkana Lapisatepun
- Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wasana Ko-Iam
- Clinical Surgical Research Center, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
12
|
Guerin DJ, Gutierrez B, Zhang B, Tseng KAS. Notch Is Required for Neural Progenitor Proliferation During Embryonic Eye Regrowth. Int J Mol Sci 2025; 26:2637. [PMID: 40141279 PMCID: PMC11942531 DOI: 10.3390/ijms26062637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
The ability of an organism to regrow tissues is regulated by various signaling pathways. One such pathway that has been studied widely both in the context of regeneration and development is the Notch signaling pathway. Notch is required for the development of the eye and regeneration of tissues in multiple organisms, but it is unknown if Notch plays a role in the regulation of Xenopus laevis embryonic eye regrowth. We found that Notch1 is required for eye regrowth and regulates retinal progenitor cell proliferation. Chemical and molecular inhibition of Notch1 significantly decreased eye regrowth by reducing retinal progenitor cell proliferation without affecting retinal differentiation. Temporal inhibition studies showed that Notch function is required during the first day of regrowth. Interestingly, Notch1 loss-of-function phenocopied the effects of the inhibition of the proton pump, vacuolar-type ATPase (V-ATPase), where retinal proliferation but not differentiation was blocked during eye regrowth. Overexpression of a form of activated Notch1, the Notch intracellular domain (NICD) rescued the loss of eye regrowth due to V-ATPase inhibition. These findings highlight the importance of the Notch signaling pathway in eye regeneration and its role in inducing retinal progenitor cell proliferation in response to injury.
Collapse
|
13
|
Reddy MS, Gopal PV. Small for Size Syndrome in Living Donor Liver Transplantation- Prevention and Management. J Clin Exp Hepatol 2025; 15:102458. [PMID: 39722782 PMCID: PMC11666951 DOI: 10.1016/j.jceh.2024.102458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/13/2024] [Indexed: 12/28/2024] Open
Abstract
Small-for-size syndrome is a clinical syndrome of early allograft dysfunction usually following living donor liver transplantation due to a mismatch between recipient metabolic and functional requirements and the graft's functional capacity. While graft size relative to the recipient size is the most commonly used parameter to predict risk, small-for-size syndrome is multifactorial and its development depends on a number of inter-dependant factors only some of which are modifiable. Intra-operative monitoring of portal haemodynamics and portal flow modulation is widely recommended though there is wide variation in clinical practice. Management of established small-for-size syndrome centres around meticulous patient care, infection prevention, fluid management and identifying correctable technical complications. However, retransplantation is the only treatment in severe cases. While small-for-size syndrome per se is associated with increased peri-operative mortality, the contribution of non-hepatic organ failure in determining patient outcomes needs further studies.
Collapse
Affiliation(s)
- Mettu Srinivas Reddy
- Star Institute for Advanced Liver Care & Transplantation, Star Hospitals, Hyderabad, Rainbow Children's Hospital, Hyderabad, India
| | - Prasanna V. Gopal
- Institute of Liver Disease & Transplantation, Gleneagles Health City, Chennai, India
| |
Collapse
|
14
|
Zhang J, Qiu X, Lei Y, Chen H, Wu D, Wang T, Sui X, Xiao J, Jiang C, Zhang H, Liu Y, Liu X, Zhang Y, Che X, Lin Y, Yao J, Pan Z, Li R, Zheng J. Engineered EVs from LncEEF1G - overexpressing MSCs promote fibrotic liver regeneration by upregulating HGF release from hepatic stellate cells. Exp Mol Med 2025; 57:584-600. [PMID: 40025174 PMCID: PMC11958733 DOI: 10.1038/s12276-025-01413-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 11/30/2024] [Accepted: 12/12/2024] [Indexed: 03/04/2025] Open
Abstract
Fibrosis is a disease that negatively affects liver regeneration, resulting in severe complications after liver surgery. However, there is still no clinically effective treatment for promoting fibrotic liver regeneration because the underlying hepatocellular mechanism remains poorly understood. Through microRNA microarrays combined with the application of AAV6, we found that high expression of miR-181a-5p in activated hepatic stellate cells (HSCs) suppressed the expression of hepatic growth factor (HGF) and partially contributed to impaired regeneration potential in mice with hepatic fibrosis that had undergone two-thirds partial hepatectomy. As nanotherapeutics, mesenchymal stem-cell-derived extracellular vesicles (MSC-EVs) have been verified as effective treatments for liver regeneration. Here we observe that MSC-EVs can also promote fibrotic liver regeneration via enriched lncEEF1G, which acts as a competing endogenous RNA to directly sponge miR-181a-5p, leading to the upregulated expression of HGF in HSCs. Finally, engineered MSC-EVs with high expression of lncEEF1G (lncEEF1GOE-EVs) were constructed, suggesting greater potential for this model. In summary, our findings indicate that lncEEF1GOE-EVs have a nanotherapeutic effect on promoting regeneration of fibrotic livers by modulating the miR-181a-5p/HGF pathway in HSCs, which highlights the potential of extracellular vesicle engineering technology for patients with hepatic fibrosis who have undergone hepatic surgery. Engineered mesenchymal stem cells that overexpress lncEEF1G can secrete extracellular vesicles that are rich in lncEEF1G (lncEEF1GOE-EVs). Upon injection of lncEEF1GOE-EVs into a fibrotic 70% partial hepatectomy mouse model, lncEEF1G competitively binds to miR-181a-5p in hepatic stellate cells, preventing the interaction between miR-181a-5p and the messenger RNA of hepatocyte growth factor. This consequently leads to an increase in the secretion of hepatocyte growth factor and the promotion of hepatocyte proliferation.
Collapse
Affiliation(s)
- Jiebin Zhang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaotong Qiu
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yunguo Lei
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haitian Chen
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dongwei Wu
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tingting Wang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xin Sui
- Surgical ICU, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiaqi Xiao
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chenhao Jiang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huayao Zhang
- Shaoguan Maternal and Child Health Hospital, Shaoguan, Guangdong, China
| | - Yasong Liu
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoquan Liu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yingcai Zhang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xu Che
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Ye Lin
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jia Yao
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Zihao Pan
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Rong Li
- Guangdong Provincial Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Jun Zheng
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Liver Disease Research, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
15
|
Huang J, Zheng Y, Ma W, Han Y, Xue J, Huan Z, Wu C, Zhu Y. SiO 2-based inorganic nanofiber aerogel with rapid hemostasis and liver wound healing functions. Acta Biomater 2025; 194:483-497. [PMID: 39826855 DOI: 10.1016/j.actbio.2025.01.027] [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: 09/25/2024] [Revised: 12/31/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Non-compressible hemostasis and promoting tissue healing are important in soft tissue trauma repair. Inorganic aerogels show superior performance in rapid hemostasis or promoting tissue healing, but simultaneously promoting non-compressive hemostasis and soft tissue healing still remains a challenge. Herein, SiO2-based inorganic nanofiber aerogels (M2+@SiO2, M=Ca, Mg, and Sr) were prepared by freeze-drying the mixture of bioactive silicates-deposited SiO2 nanofibers and SiO2 sol. These M2+@SiO2 aerogels have a three-dimensional highly-interconnected porous structure, remarkable flexibility, high absorption, good hydrophilicity, negative zeta potential, and bioactive ions releasing capacity. M2+@SiO2 aerogels not only exhibited satisfactory hemostasis activities in vitro, but also possessed high hemostatic efficacy in compressible rabbit femoral artery injury bleeding model and non-compressible rat liver puncture bleeding model compared to medical gauze and gelatin sponge. M2+@SiO2 aerogel had low blood clotting index of Ca. 10 % and short partial thromboplastin time of ca. 82 s in vitro, and could greatly short bleeding time by >50 % and decrease blood loss by about 80 % compared to medical gauze and gelatin sponge in non-compressible hemostasis. Sr2+@SiO2 aerogel showed optimal bioactivities on promoting cell proliferation, cell migration, and the expression of liver function and angiogenesis related genes and proteins in vitro. Importantly, Sr2+@SiO2 aerogel possessed a noteworthy function to promote liver soft tissue healing in vivo by releasing bioactive ions and providing a highly-interconnected porous structure to support vascular development and tissue regeneration. Overall, Sr2+@SiO2 aerogel has great potential for integrated rapid hemostasis and soft tissue healing, which is promising in soft tissue trauma therapy. STATEMENT OF SIGNIFICANCE: Non-compressible hemorrhage and soft tissue impairment are the main causes of mortality in emergency trauma. Inorganic aerogels with high porosity and outstanding flexibility can rapidly absorb blood to pro-coagulation and fill in irregular trauma without compression, but the low bioactivity limited the ability to promote soft tissue healing. Herein, SiO2-based inorganic nanofiber aerogels (M2+@SiO2, M=Ca, Mg, and Sr) were prepared by freeze-drying the mixture of bioactive silicates-deposited SiO2 nanofibers and SiO2 sol. M2+@SiO2 aerogels possessed high bioactivity and exhibited superior hemostatic performance in compressible and non-compressible bleeding model. Furthermore, Sr2+@SiO2 aerogel showed optimal bioactivities on cell responses and effectively promoted liver healing by releasing bioactive ions and providing highly-interconnected porous support structure for vascular development and tissue regeneration.
Collapse
Affiliation(s)
- Jimin Huang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Yi Zheng
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Wenping Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Yahui Han
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Jianmin Xue
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Zhiguang Huan
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China.
| | - Yufang Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China.
| |
Collapse
|
16
|
Graham RE, Zheng R, Wagner J, Unciti-Broceta A, Hay DC, Forbes SJ, Gadd VL, Carragher NO. Single-cell morphological tracking of cell states to identify small-molecule modulators of liver differentiation. iScience 2025; 28:111871. [PMID: 39995868 PMCID: PMC11848441 DOI: 10.1016/j.isci.2025.111871] [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: 02/06/2024] [Revised: 07/24/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
We have developed a single-cell assay that combines Cell Painting-a morphological profiling assay-with trajectory inference analysis. We have applied this morphological trajectory inference to the bi-potent HepaRG liver progenitor cell line allowing us to track liver cell fate and map small-molecule-induced changes using a morphological atlas of liver cell differentiation. Our overarching goal is to demonstrate the potential of Cell Painting to study biological processes as continuous trajectories at the single-cell level, enhancing resolution and biological understanding. This work has identified small-molecule Src family kinase inhibitors that promote the differentiation of HepaRG cells toward a hepatocyte-like lineage as well as primary human hepatic progenitor cells toward a hepatocyte-like phenotype in vitro. These findings could significantly advance research on liver cell regeneration mechanisms and facilitate the development of cell-based and small-molecule therapies.
Collapse
Affiliation(s)
- Rebecca E. Graham
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Runshi Zheng
- Centre for Regenerative Medicine, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jesko Wagner
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Asier Unciti-Broceta
- Edinburgh Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
- Cancer Research UK Scotland Centre, Edinburgh EH4 2XU, UK
| | - David C. Hay
- Centre for Regenerative Medicine, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Stuart J. Forbes
- Centre for Regenerative Medicine, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Victoria L. Gadd
- Centre for Regenerative Medicine, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Neil O. Carragher
- Edinburgh Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
- Cancer Research UK Scotland Centre, Edinburgh EH4 2XU, UK
| |
Collapse
|
17
|
Dong Q, Liu Z, Ma Y, Chen X, Wang X, Tang J, Ma K, Liang C, Wang M, Wu X, Liu Y, Zhou Y, Yang H, Gao M. Adipose tissue deficiency impairs transient lipid accumulation and delays liver regeneration following partial hepatectomy in male Seipin knockout mice. Clin Transl Med 2025; 15:e70238. [PMID: 39980067 PMCID: PMC11842221 DOI: 10.1002/ctm2.70238] [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: 10/08/2024] [Revised: 01/30/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Liver diseases pose significant health challenges, underscoring the importance of understanding liver regeneration mechanisms. Systemic adipose tissue is thought to be a primary source of lipids and energy during this process; however, empirical data on the effects of adipose tissue deficiency are limited. This study investigates the role of adipose tissue in liver regeneration, focusing on transient regeneration-associated steatosis (TRAS) and hepatocyte proliferation using a Seipin knockout mouse model that mimics severe human lipodystrophy. Additionally, the study explores therapeutic strategies through adipose tissue transplantation. METHODS Male Seipin knockout (Seipin-/-) and wild-type (WT) mice underwent 2/3 partial hepatectomy (PHx). Liver and plasma samples were collected at various time points post-surgery. Histological assessments, lipid accumulation analyses and measurements of hepatocyte proliferation markers were conducted. Additionally, normal adipose tissue was transplanted into Seipin-/- mice to evaluate the restoration of liver regeneration. RESULTS Seipin-/- mice exhibited significantly reduced liver regeneration rates and impaired TRAS, as evidenced by histological and lipid measurements. While WT mice demonstrated extensive hepatocyte proliferation at 48 and 72 h post-PHx, characterised by increased mitotic cells, elevated proliferating cell nuclear antigen and Ki67 expression, Seipin-/- mice showed delayed hepatocyte proliferation. Notably, adipose tissue transplantation into Seipin-/- mice restored TRAS and improved liver regeneration and hepatocyte proliferation. Conversely, liver-specific overexpression of Seipin in Seipin-/- mice did not affect TRAS or liver regeneration, indicating that the observed effects are primarily due to adipose tissue deficiency rather than hepatic Seipin itself. CONCLUSIONS Systemic adipose tissue is essential for TRAS and effective liver regeneration following PHx. Its deficiency impairs these processes, while adipose tissue transplantation can restore normal liver function. These findings underscore the critical role of adipose tissue in liver recovery and suggest potential therapeutic strategies for liver diseases associated with lipodystrophies. KEY POINTS Seipin-/- mice, which lack adipose tissue, exhibit significantly impaired TRAS and delayed liver regeneration following partial hepatectomy. Transplantation of normal adipose tissue into Seipin-/- mice restores TRAS and enhances liver regeneration, highlighting the essential role of adipose tissue in these processes. Liver-specific overexpression of Seipin has no effect on TRAS and liver regeneration in Seipin-/- mice.
Collapse
Affiliation(s)
- Qianqian Dong
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
- Department of Clinical LaboratoryThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Ziwei Liu
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
- Department of Clinical LaboratoryBethune International Peace HospitalShijiazhuangHebeiChina
| | - Yidan Ma
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
| | - Xin Chen
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
- Department of General SurgeryThe First Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Xiaowei Wang
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
| | - Jinye Tang
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
| | - Kexin Ma
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
| | - Chenxi Liang
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
| | - Mengyu Wang
- Department of CardiologyFirst Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xiaoqin Wu
- Department of Integrative Biology and PharmacologyUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Yang Liu
- Department of Integrative Biology and PharmacologyUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Yaru Zhou
- Department of EndocrinologyThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Hongyuan Yang
- Department of Integrative Biology and PharmacologyUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Mingming Gao
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of Education, The Key Laboratory of Vascular Biology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical UniversityShijiazhuangHebeiChina
- Department of Integrative Biology and PharmacologyUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| |
Collapse
|
18
|
Gong D, Mo J, Zhai M, Zhou F, Wang G, Ma S, Dai X, Deng X. Advances, challenges and future applications of liver organoids in experimental regenerative medicine. Front Med (Lausanne) 2025; 11:1521851. [PMID: 39927267 PMCID: PMC11804114 DOI: 10.3389/fmed.2024.1521851] [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: 11/05/2024] [Accepted: 12/20/2024] [Indexed: 02/11/2025] Open
Abstract
The liver is a vital organ responsible for numerous metabolic processes in the human body, including the metabolism of drugs and nutrients. After liver damage, the organ can rapidly return to its original size if the causative factor is promptly eliminated. However, when the harmful stimulus persists, the liver's regenerative capacity becomes compromised. Substantial theoretical feasibility has been demonstrated at the levels of gene expression, molecular interactions, and intercellular dynamics, complemented by numerous successful animal studies. However, a robust model and carrier that closely resemble human physiology are still lacking for translating these theories into practice. The potential for liver regeneration has been a central focus of ongoing research. Over the past decade, the advent of organoid technology has provided improved models and materials for advancing research efforts. Liver organoid technology represents a novel in vitro culture system. After several years of refinement, human liver organoids can now accurately replicate the liver's morphological structure, nutrient and drug metabolism, gene expression, and secretory functions, providing a robust model for liver disease research. Regenerative medicine aims to replicate human organ or tissue functions to repair or replace damaged tissues, restore their structure or function, or stimulate the regeneration of tissues or organs within the body. Liver organoids possess the same structure and function as liver tissue, offering the potential to serve as a viable replacement for the liver, aligning with the goals of regenerative medicine. This review examines the role of liver organoids in regenerative medicine.
Collapse
Affiliation(s)
- Da Gong
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Jiaye Mo
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People’s Hospital, Shenzhen, China
- Guangxi University of Chinese Medicine, Nanning, China
| | - Mei Zhai
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Fulin Zhou
- Department of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Guocai Wang
- Department of Physiology, School of Medicine and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Shaohua Ma
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Tsinghua University Shenzhen International Graduate School, Guangdong, China
| | - Xiaoyong Dai
- Department of Physiology, School of Medicine and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Tsinghua University Shenzhen International Graduate School, Guangdong, China
| | - Xuesong Deng
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People’s Hospital, Shenzhen, China
| |
Collapse
|
19
|
Szternel Ł, Sobucki B, Wieprzycka L, Krintus M, Panteghini M. Golgi protein 73 in liver fibrosis. Clin Chim Acta 2025; 565:119999. [PMID: 39401651 DOI: 10.1016/j.cca.2024.119999] [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: 09/11/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/17/2024]
Abstract
Golgi protein 73 (GP73) is implicated in key pathogenic processes, particularly those related to inflammation and fibrogenesis. In the last years, its measurement has emerged as a promising biomarker for detection of liver fibrosis (LF), a common consequence of chronic liver disease that can progress to cirrhosis and eventually hepatocellular carcinoma. GP73 concentrations in blood appear significantly increased in LF patients, correlating with disease severity, making this biomarker a possible non-invasive alternative for detecting and monitoring this condition regardless of etiology. Understanding the molecular mechanisms involving GP73 expression could also lead to new therapeutic strategies aimed at modulating its synthesis or function to prevent or reverse LF. Despite its clinical potential, GP73 as a LF biomarker faces several challenges. The lack of demonstrated comparability among different assays as well as the lack of knowledge of individual variability can make difficult the result interpretation. Further research is therefore needed focusing on robust clinical validation of GP73 as a LF biomarker. Addressing analytical, biological, and clinical limitations will be critical to exploiting its potential for improving detection and monitoring of advanced LF.
Collapse
Affiliation(s)
- Łukasz Szternel
- Department of Laboratory Medicine, Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Poland
| | - Bartłomiej Sobucki
- Department of Laboratory Medicine, Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Poland
| | - Laura Wieprzycka
- Department of Laboratory Medicine, Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Poland
| | - Magdalena Krintus
- Department of Laboratory Medicine, Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Poland.
| | - Mauro Panteghini
- Department of Laboratory Medicine, Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Poland
| |
Collapse
|
20
|
Eyraud D, Philippe A, Guerin C, Sarmiento I, Suner L, Puybasset L, Bertil S, Vaillant JC, Helley D, Granger B, Smadja DM, Gaussem P. Cirrhotic Patients Exhibit Remarkable Vascular Regenerative Profile One Month after Liver Transplantation. Stem Cell Rev Rep 2025; 21:276-279. [PMID: 39377987 DOI: 10.1007/s12015-024-10796-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 01/26/2025]
Affiliation(s)
- Daniel Eyraud
- Université Pierre et Marie Curie, Paris, France, Department of Anesthesiology and Reanimation, AP-HP, Pitié-Salpêtrière University Hospital, Paris, F-75013, France
| | - Aurélien Philippe
- Service d'hématologie biologique, AP-HP, Université de Paris Cité, Hôpital Européen Georges Pompidou, Paris, F-75015, France
- Université de Paris Cité, Innovative Therapies in Haemostasis, INSERM, Paris, F-75006, France
| | - Coralie Guerin
- Université de Paris Cité, Innovative Therapies in Haemostasis, INSERM, Paris, F-75006, France
- Cytometry Platform, Curie CoreTech, Institut Curie, Paris, F-75005, France
| | - Ignacio Sarmiento
- Université Pierre et Marie Curie, Paris, France, Department of Anesthesiology and Reanimation, AP-HP, Pitié-Salpêtrière University Hospital, Paris, F-75013, France
| | - Ludovic Suner
- Service d'hématologie biologique, AP-HP, Université de Paris Cité, Hôpital Européen Georges Pompidou, Paris, F-75015, France
| | - Louis Puybasset
- Université Pierre et Marie Curie, Paris, France, Department of Anesthesiology and Reanimation, AP-HP, Pitié-Salpêtrière University Hospital, Paris, F-75013, France
| | - Sébastien Bertil
- Service d'hématologie biologique, AP-HP, Université de Paris Cité, Hôpital Européen Georges Pompidou, Paris, F-75015, France
| | - Jean-Christophe Vaillant
- Université Pierre et Marie Curie, Paris, France, Department of Digestive, HPB Surgery, and Liver Transplantation, AP-HP, Pitié-Salpêtrière University Hospital, Paris, F-75013, France
| | - Dominique Helley
- Service d'hématologie biologique, AP-HP, Université de Paris Cité, Hôpital Européen Georges Pompidou, Paris, F-75015, France
- Université de Paris Cité, Paris Cardiovascular Research Center, Paris, F-75015, France
| | - Benjamin Granger
- Université Pierre et Marie Curie, Paris, France, Department of statistics, Clinical Research Unit, AP-HP, Pitié-Salpêtrière University Hospital, Paris, F-75013, France
| | - David M Smadja
- Service d'hématologie biologique, AP-HP, Université de Paris Cité, Hôpital Européen Georges Pompidou, Paris, F-75015, France.
- Université de Paris Cité, Innovative Therapies in Haemostasis, INSERM, Paris, F-75006, France.
| | - Pascale Gaussem
- Service d'hématologie biologique, AP-HP, Université de Paris Cité, Hôpital Européen Georges Pompidou, Paris, F-75015, France
- Université de Paris Cité, Innovative Therapies in Haemostasis, INSERM, Paris, F-75006, France
| |
Collapse
|
21
|
McGinn M, Rabender C, Mikkelsen R, Yakovlev V. Hepatocyte-derived extracellular vesicles regulate liver regeneration through a negative feedback mechanism. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70023. [PMID: 39554866 PMCID: PMC11565257 DOI: 10.1002/jex2.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/24/2024] [Accepted: 11/03/2024] [Indexed: 11/19/2024]
Abstract
While significant progress has been made in understanding various aspects of liver regeneration, the molecular mechanisms responsible for the initiation and termination of cell proliferation in the liver following massive tissue loss or injury of liver remain unknown. As it was previously shown, the loss of liver mass affects putative hepatocyte-specific mitogenic inhibitors in the blood. Although the presence of these putative inhibitors regulating precise liver regeneration has been described in numerous publications, they have never been identified. Extracellular vesicles (EVs) are nano-sized, membrane-limited structures secreted by cells into the extracellular space. Their proposed role is stable intercellular carriers of proteins and RNAs, predominantly micro-RNA, from secreted to recipient cells. Upon uptake by the recipient cells, EVs can significantly modulate their biological functions. In the present study, using in vivo and in vitro models, we demonstrate that hepatocyte proliferation and liver regeneration are regulated by EVs secreted by hepatocytes into the bloodstream. This regulation occurs through a negative feedback mechanism, which explains the precise regeneration of liver tissue after massive damage. We also demonstrate that an essential component of this mechanism is RNA carried by hepatocyte-derived EVs. Our findings open up a new and unexplored area of liver biology regarding the mechanisms involved in the precise regulation of liver regeneration after a massive tissue loss or injury. Further study of this mechanism will have a great influence on the development of new approaches to liver transplantation, various liver pathologies, and hepatic tumors.
Collapse
Affiliation(s)
- Mina McGinn
- Department of Radiation Oncology, Massey Comprehensive Cancer CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Christopher Rabender
- Department of Radiation Oncology, Massey Comprehensive Cancer CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Ross Mikkelsen
- Department of Radiation Oncology, Massey Comprehensive Cancer CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Vasily Yakovlev
- Department of Radiation Oncology, Massey Comprehensive Cancer CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
| |
Collapse
|
22
|
Mo D, Lv M, Mao X. Using different zebrafish models to explore liver regeneration. Front Cell Dev Biol 2024; 12:1485773. [PMID: 39544362 PMCID: PMC11560876 DOI: 10.3389/fcell.2024.1485773] [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: 08/24/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
The liver possesses an impressive capability to regenerate following various injuries. Given its profound implications for the treatment of liver diseases, which afflict millions globally, liver regeneration stands as a pivotal area of digestive organ research. Zebrafish (Danio rerio) has emerged as an ideal model organism in regenerative medicine, attributed to their remarkable ability to regenerate tissues and organs, including the liver. Many fantastic studies have been performed to explore the process of liver regeneration using zebrafish, especially the extreme hepatocyte injury model. Biliary-mediated liver regeneration was first discovered in the zebrafish model and then validated in mammalian models and human patients. Considering the notable expansion of biliary epithelial cells in many end-stage liver diseases, the promotion of biliary-mediated liver regeneration might be another way to treat these refractory liver diseases. To date, a comprehensive review discussing the current advancements in zebrafish liver regeneration models is lacking. Therefore, this review aims to investigate the utility of different zebrafish models in exploring liver regeneration, highlighting the genetic and cellular insights gained and discussing the potential translational impact on human health.
Collapse
Affiliation(s)
- Dashuang Mo
- Department of Immunology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Mengzhu Lv
- Department of Immunology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xiaoyu Mao
- College of Language Intelligence, Sichuan International Studies University, Chongqing, China
| |
Collapse
|
23
|
Alatas FS, Yamaza T, Matsuura T, Ongko L, Kadim M, Ohga S, Taguchi T, Tajiri T. Potential role of stem cells from human exfoliated deciduous teeth in inducing liver regeneration. J Gastroenterol Hepatol 2024; 39:2190-2196. [PMID: 38859685 DOI: 10.1111/jgh.16651] [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: 11/27/2023] [Revised: 04/11/2024] [Accepted: 05/28/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND AND AIM Even with advancement of medical technologies, liver transplantation still faces several major challenges. Hence, other treatment modalities are urgently needed for patients with end-stage liver disease. Stem cells from human exfoliated deciduous teeth (SHED) was discovered to have highly proliferative and pluripotent properties; including differentiation into hepatocyte-like cells. This study aims to investigate the capability of intrasplenic transplanted SHED and SHED-Hep cells in inducing proliferation of stem cells and native hepatocytes in order to accelerate liver regeneration in liver fibrosis mice models. METHODS Three carbon tetrachloride (CCl4)-injured male mice groups were used in this study. Two of those groups were transplanted with either SHED or SHED-Hep, while the other did not undergo transplantation. One age- and sex- matched healthy mice group was used as control. All specimens were immunohistochemically stained with anti-Ki-67 antibodies and anti-proliferating cell nuclear antigen (PCNA) antibodies before counter stained with hematoxylin-eosin. RESULTS Anti-Ki-67 antibodies staining: at both 8 and 12 weeks, proliferating activity was predominantly seen on both SHED- and SHED-Hep-transplanted CCl4-injured mice groups, while control and non-transplanted CCl4-injured mice group showed little to no sign of proliferation activity. Anti-PCNA staining: at both 8 and 12 weeks, significant proliferating activity was detected by PCNA staining, mainly on stem cells population area on SHED- and SHED-Hep-treated group. CONCLUSIONS In conclusion, this study has provided the evidence that transplantation of SHED or SHED-Hep on liver-injured mice induced proliferation of both transplanted stem cells and native liver cells in order to accelerate liver regeneration.
Collapse
Affiliation(s)
- Fatima Safira Alatas
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Child Health, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Takayoshi Yamaza
- Departments of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, Fukuoka, Japan
| | - Toshiharu Matsuura
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Lukito Ongko
- Department of Child Health, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Muzal Kadim
- Department of Child Health, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Shouichi Ohga
- Departments of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoaki Taguchi
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Fukuoka College of Health Sciences, Fukuoka, Japan
| | - Tatsuro Tajiri
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
24
|
Liang Y, Mei Q, He E, Ballar P, Wei C, Wang Y, Dong Y, Zhou J, Tao X, Qu W, Zhao M, Chhetri G, Wei L, Shao J, Shen Y, Liu J, Feng L, Shen Y. MANF serves as a novel hepatocyte factor to promote liver regeneration after 2/3 partial hepatectomy via doubly targeting Wnt/β-catenin signaling. Cell Death Dis 2024; 15:681. [PMID: 39289348 PMCID: PMC11408687 DOI: 10.1038/s41419-024-07069-8] [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: 04/23/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
Liver regeneration is an intricate pathophysiological process that has been a subject of great interest to the scientific community for many years. The capacity of liver regeneration is very critical for patients with liver diseases. Therefore, exploring the mechanisms of liver regeneration and finding good ways to improve it are very meaningful. Mesencephalic astrocyte-derived neurotrophic factor (MANF), a member of newly identified neurotrophic factors (NTFs) family, extensively expresses in the liver and has demonstrated cytoprotective effects during ER stress and inflammation. However, the role of MANF in liver regeneration remains unclear. Here, we used hepatocyte-specific MANF knockout (MANFHep-/-) mice to investigate the role of MANF in liver regeneration after 2/3 partial hepatectomy (PH). Our results showed that MANF expression was up-regulated in a time-dependent manner, and the peak level of mRNA and protein appeared at 24 h and 36 h after 2/3 PH, respectively. Notably, MANF knockout delayed hepatocyte proliferation, and the peak proliferation period was delayed by 24 h. Mechanistically, our in vitro results showed that MANF physically interacts with LRP5 and β-catenin, two essential components of Wnt/β-catenin pathway. Specifically, as a cofactor, MANF binds to the extracellular segment of LRP5 to activate Wnt/β-catenin signaling. On the other hand, MANF interacts with β-catenin to stabilize cytosolic β-catenin level and promote its nuclear translocation, which further enhance the Wnt/β-catenin signaling. We also found that MANF knockout does not affect the c-Met/β-catenin complex after 2/3 PH. In summary, our study confirms that MANF may serve as a novel hepatocyte factor that is closely linked to the activation of the Wnt/β-catenin pathway via intracellular and extracellular targets.
Collapse
Affiliation(s)
- Yanyan Liang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Qiong Mei
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Enguang He
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Petek Ballar
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Izmir, 35100, Turkey
| | - Chuansheng Wei
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Yue Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Yue Dong
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Jie Zhou
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Xiaofang Tao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Wenyan Qu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Mingxia Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Goma Chhetri
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Limeng Wei
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Juntang Shao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Yujun Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Jun Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Lijie Feng
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Yuxian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.
- Department of General Surgery, The First Affiliated Hospital, Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
25
|
Su R, Ai Y, Wang J, Wu L, Sun H, Ding M, Xie R, Liang Q. Engineered Microfibers for Tissue Engineering. ACS APPLIED BIO MATERIALS 2024; 7:5823-5840. [PMID: 39145987 DOI: 10.1021/acsabm.4c00615] [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: 08/16/2024]
Abstract
Hydrogel microfibers are hydrogel materials engineered into fiber structures. Techniques such as wet spinning, microfluidic spinning, and 3D bioprinting are often used to prepare microfibers due to their ability to precisely control the size, morphology, and structure of the microfibers. Microfibers with different structural morphologies have different functions; they provide a flow-through culture environment for cells to improve viability, and can also be used to induce the differentiation of cells such as skeletal muscle and cardiac muscle cells to eventually form functional organs in vitro through special morphologies. This Review introduces recent advances in microfluidics, 3D bioprinting, and wet spinning in the preparation of microfibers, focusing on the materials and fabrication methods. The applications of microfibers in tissue engineering are highlighted by summarizing their contributions in engineering biomimetic blood vessels, vascularized tissues, bone, heart, pancreas, kidney, liver, and fat. Furthermore, applications of engineered fibers in tissue repair and drug screening are also discussed.
Collapse
Affiliation(s)
- Riguga Su
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Yongjian Ai
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Jingyu Wang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Lei Wu
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Hua Sun
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Mingyu Ding
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Ruoxiao Xie
- Department of Materials, Design and Manufacturing Engineering, School of Engineering, University of Liverpool, Liverpool L69 3BX, U.K
| | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| |
Collapse
|
26
|
Wu Y, Li L, Li W, Li N, Zhang X, Zheng L, Zhong S, Lü S, Shu X, Zhou J, Ai D, Gao M, Liu S, Lü D, Long M. Stretch-induced hepatic endothelial mechanocrine promotes hepatocyte proliferation. Hepatology 2024:01515467-990000000-01018. [PMID: 39250438 DOI: 10.1097/hep.0000000000001082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 08/20/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND AND AIMS Partial hepatectomy-induced liver regeneration causes the increase in relative blood flow rate within the liver, which dilates hepatic sinusoids and applies mechanical stretch on liver sinusoidal endothelial cells (LSECs). Heparin-binding EGF-like growth factor is a crucial growth factor during liver regeneration. We aimed to investigate whether this sinusoidal dilation-induced stretch promotes HB-EGF secretion in LSECs and what the related molecular mechanism is. APPROACH AND RESULTS In vivo partial hepatectomy, ex vivo liver perfusion, and in vitro LSEC mechanical stretch were applied to detect HB-EGF expression in LSECs and hepatocyte proliferation. Knockdown or inhibition of mechanosensitive proteins was used to unravel the molecular mechanism in response to stretch. This stretch triggers amplitude-dependent and duration-dependent HB-EGF upregulation in LSECs, which is mediated by Yes-associated protein (YAP) nuclear translocation and binding to TEA domain family. This YAP translocation is achieved in 2 ways: On one hand, F-actin polymerization-mediated expansion of nuclear pores promotes YAP entry into nucleus passively. On the other hand, F-actin polymerization upregulates the expression of BAG family molecular chaperone regulator 3, which binds with YAP to enter the nucleus cooperatively. In this process, β1-integrin serves as a target mechanosensory in stretch-induced signaling pathways. This HB-EGF secretion-promoted liver regeneration after 2/3 partial hepatectomy is attenuated in endothelial cell-specific Yap1 -deficient mice. CONCLUSIONS Our findings indicate that mechanical stretch-induced HB-EGF upregulation in LSECs through YAP translocation can promote hepatocyte proliferation during liver regeneration through a mechanocrine manner, which deepens the understanding of the mechanical-biological coupling in liver regeneration.
Collapse
Affiliation(s)
- Yi Wu
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Linda Li
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Wang Li
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Ning Li
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyu Zhang
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Lu Zheng
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Shaoyu Zhong
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Shouqin Lü
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Xinyu Shu
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jin Zhou
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Ding Ai
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Ming Gao
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Sijin Liu
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Dongyuan Lü
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Mian Long
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
27
|
Li P, Ma X, Huang D, Gu X. Exploring the roles of non-coding RNAs in liver regeneration. Noncoding RNA Res 2024; 9:945-953. [PMID: 38680418 PMCID: PMC11046251 DOI: 10.1016/j.ncrna.2024.04.003] [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: 02/14/2024] [Revised: 03/26/2024] [Accepted: 04/16/2024] [Indexed: 05/01/2024] Open
Abstract
Liver regeneration (LR) is a complex process encompassing three distinct phases: priming, proliferation phase and restoration, all influenced by various regulatory factors. After liver damage or partial resection, the liver tissue demonstrates remarkable restorative capacity, driven by cellular proliferation and repair mechanisms. The essential roles of non-coding RNAs (ncRNAs), predominantly microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNA (circRNA), in regulating LR have been vastly studied. Additionally, the impact of ncRNAs on LR and their abnormal expression profiles during this process have been extensively documented. Mechanistic investigations have revealed that ncRNAs interact with genes involved in proliferation to regulate hepatocyte proliferation, apoptosis and differentiation, along with liver progenitor cell proliferation and migration. Given the significant role of ncRNAs in LR, an in-depth exploration of their involvement in the liver's self-repair capacity can reveal promising therapeutic strategies for LR and liver-related diseases. Moreover, understanding the unique regenerative potential of the adult liver and the mechanisms and regulatory factors of ncRNAs in LR are crucial for improving current treatment strategies and exploring new therapeutic approaches for various liver-related diseases. This review provides a brief overview of the LR process and the ncRNA expression profiles during this process. Furthermore, we also elaborate on the specific molecular mechanisms through which multiple key ncRNAs regulate the LR process. Finally, based on the expression characteristics of ncRNAs and their interactions with proliferation-associated genes, we explore their potential clinical application, such as developing predictive indicators reflecting liver regenerative activity and manipulating LR processes for therapeutic purposes.
Collapse
Affiliation(s)
- Penghui Li
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Xiao Ma
- Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, China
| | - Di Huang
- Department of Child Health Care, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| |
Collapse
|
28
|
Wu D, van de Graaf SFJ. Maladaptive regeneration and metabolic dysfunction associated steatotic liver disease: Common mechanisms and potential therapeutic targets. Biochem Pharmacol 2024; 227:116437. [PMID: 39025410 DOI: 10.1016/j.bcp.2024.116437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
The normal liver has an extraordinary capacity of regeneration. However, this capacity is significantly impaired in steatotic livers. Emerging evidence indicates that metabolic dysfunction associated steatotic liver disease (MASLD) and liver regeneration share several key mechanisms. Some classical liver regeneration pathways, such as HGF/c-Met, EGFR, Wnt/β-catenin and Hippo/YAP-TAZ are affected in MASLD. Some recently established therapeutic targets for MASH such as the Thyroid Hormone (TH) receptors, Glucagon-like protein 1 (GLP1), Farnesoid X receptor (FXR), Peroxisome Proliferator-Activated Receptors (PPARs) as well as Fibroblast Growth Factor 21 (FGF21) are also reported to affect hepatocyte proliferation. With this review we aim to provide insight into common molecular pathways, that may ultimately enable therapeutic strategies that synergistically ameliorate steatohepatitis and improve the regenerating capacity of steatotic livers. With the recent rise of prolonged ex-vivo normothermic liver perfusion prior to organ transplantation such treatment is no longer restricted to patients undergoing major liver resection or transplantation, but may eventually include perfused (steatotic) donor livers or even liver segments, opening hitherto unexplored therapeutic avenues.
Collapse
Affiliation(s)
- Dandan Wu
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, the Netherlands
| | - Stan F J van de Graaf
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, the Netherlands.
| |
Collapse
|
29
|
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
|
30
|
Martucci NJ, Stoops J, Bowen W, Orr A, Cotner MC, Michalopoulos GK, Bhushan B, Mars WM. A Novel Role for the Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Delta Isoform in Hepatocellular Proliferation. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1511-1527. [PMID: 38705383 PMCID: PMC11393825 DOI: 10.1016/j.ajpath.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/09/2024] [Accepted: 03/22/2024] [Indexed: 05/07/2024]
Abstract
The phosphatidylinositol-4,5-bisphosphate 3-kinase delta isoform (Pik3cd), usually considered immune-specific, was unexpectedly identified as a gene potentially related to either regeneration and/or differentiation in animals lacking hepatocellular Integrin Linked Kinase (ILK). Since a specific inhibitor (Idelalisib, or CAL101) for the catalytic subunit encoded by Pik3cd (p110δ) has reported hepatotoxicity when used for treating chronic lymphocytic leukemia and other lymphomas, the authors aimed to elucidate whether there is a role for p110δ in normal liver function. To determine the effect on normal liver regeneration, partial hepatectomy (PHx) was performed using mice in which p110δ was first inhibited using CAL101. Inhibition led to over a 50% decrease in proliferating hepatocytes in the first 2 days after PHx. This difference correlated with phosphorylation changes in the HGF and EGF receptors (MET and EGFR, respectively) and NF-κB signaling. Ingenuity Pathway Analyses implicated C/EBPβ, HGF, and the EGFR heterodimeric partner, ERBB2, as three of the top 20 regulators downstream of p110δ signaling because their pathways were suppressed in the presence of CAL101 at 1 day post-PHx. A regulatory role for p110δ signaling in mouse and rat hepatocytes through MET and EGFR was further verified using hepatocyte primary cultures, in the presence or absence of CAL101. Combined, these data support a role for p110δ as a downstream regulator of normal hepatocytes when stimulated to proliferate.
Collapse
Affiliation(s)
- Nicole J Martucci
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John Stoops
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William Bowen
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anne Orr
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mary-Claire Cotner
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Bharat Bhushan
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wendy M Mars
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
31
|
Perez-Gutierrez L, Li P, Ferrara N. Endothelial cell diversity: the many facets of the crystal. FEBS J 2024; 291:3287-3302. [PMID: 36266750 DOI: 10.1111/febs.16660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 10/03/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Endothelial cells (ECs) form the inner lining of blood vessels and play crucial roles in angiogenesis. While it has been known for a long time that there are considerable differences among ECs from lymphatic and blood vessels, as well as among arteries, veins and capillaries, the full repertoire of endothelial diversity is only beginning to be elucidated. It has become apparent that the role of ECs is not just limited to their exchange functions. Indeed, a multitude of organ-specific functions, including release of growth factors, regulation of immune functions, have been linked to ECs. Recent years have seen a surge into the identification of spatiotemporal molecular and functional heterogeneity of ECs, supported by technologies such as single-cell RNA sequencing (scRNA-seq), lineage tracing and intersectional genetics. Together, these techniques have spurred the generation of epigenomic, transcriptomic and proteomic signatures of ECs. It is now clear that ECs across organs and in different vascular beds, but even within the same vessel, have unique molecular identities and employ specialized molecular mechanisms to fulfil highly specialized needs. Here, we focus on the molecular heterogeneity of the endothelium in different organs and pathological conditions.
Collapse
Affiliation(s)
- Lorena Perez-Gutierrez
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Pin Li
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Napoleone Ferrara
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| |
Collapse
|
32
|
Sande-Melon M, Bergemann D, Fernández-Lajarín M, González-Rosa JM, Cox AG. Development of a hepatic cryoinjury model to study liver regeneration. Development 2024; 151:dev203124. [PMID: 38975841 PMCID: PMC11318111 DOI: 10.1242/dev.203124] [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: 05/29/2024] [Accepted: 06/20/2024] [Indexed: 07/09/2024]
Abstract
The liver is a remarkable organ that can regenerate in response to injury. Depending on the extent of injury, the liver can undergo compensatory hyperplasia or fibrosis. Despite decades of research, the molecular mechanisms underlying these processes are poorly understood. Here, we developed a new model to study liver regeneration based on cryoinjury. To visualise liver regeneration at cellular resolution, we adapted the CUBIC tissue-clearing approach. Hepatic cryoinjury induced a localised necrotic and apoptotic lesion characterised by inflammation and infiltration of innate immune cells. After this initial phase, we observed fibrosis, which resolved as regeneration re-established homeostasis in 30 days. Importantly, this approach enables the comparison of healthy and injured parenchyma within an individual animal, providing unique advantages to previous models. In summary, the hepatic cryoinjury model provides a fast and reproducible method for studying the cellular and molecular pathways underpinning fibrosis and liver regeneration.
Collapse
Affiliation(s)
- Marcos Sande-Melon
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - David Bergemann
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13th Street, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Miriam Fernández-Lajarín
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13th Street, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA 02467, USA
| | - Juan Manuel González-Rosa
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13th Street, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA 02467, USA
| | - Andrew G. Cox
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3000, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, Victoria 3000, Australia
| |
Collapse
|
33
|
Brazovskaja A, Gomes T, Holtackers R, Wahle P, Körner C, He Z, Schaffer T, Eckel JC, Hänsel R, Santel M, Seimiya M, Denecke T, Dannemann M, Brosch M, Hampe J, Seehofer D, Damm G, Camp JG, Treutlein B. Cell atlas of the regenerating human liver after portal vein embolization. Nat Commun 2024; 15:5827. [PMID: 38992008 PMCID: PMC11239663 DOI: 10.1038/s41467-024-49236-7] [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: 05/22/2023] [Accepted: 05/28/2024] [Indexed: 07/13/2024] Open
Abstract
The liver has the remarkable capacity to regenerate. In the clinic, regeneration is induced by portal vein embolization, which redirects portal blood flow, resulting in liver hypertrophy in locations with increased blood supply, and atrophy of embolized segments. Here, we apply single-cell and single-nucleus transcriptomics on healthy, hypertrophied, and atrophied patient-derived liver samples to explore cell states in the regenerating liver. Our data unveils pervasive upregulation of genes associated with developmental processes, cellular adhesion, and inflammation in post-portal vein embolization liver, disrupted portal-central hepatocyte zonation, and altered cell subtype composition of endothelial and immune cells. Interlineage crosstalk analysis reveals mesenchymal cells as an interaction hub between immune and endothelial cells, and highlights the importance of extracellular matrix proteins in liver regeneration. Moreover, we establish tissue-scale iterative indirect immunofluorescence imaging for high-dimensional spatial analysis of perivascular microenvironments, uncovering changes to tissue architecture in regenerating liver lobules. Altogether, our data is a rich resource revealing cellular and histological changes in human liver regeneration.
Collapse
Affiliation(s)
| | - Tomás Gomes
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
| | - Rene Holtackers
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Philipp Wahle
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Christiane Körner
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
| | - Zhisong He
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Theresa Schaffer
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Julian Connor Eckel
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
| | - René Hänsel
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), Leipzig University, Leipzig, Germany
| | - Malgorzata Santel
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Makiko Seimiya
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Timm Denecke
- Department of Diagnostic and Interventional Radiology, Leipzig University, Leipzig, Germany
| | - Michael Dannemann
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mario Brosch
- Medical Department 1, University Hospital Dresden, Technical University Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technical University Dresden, Dresden, Germany
| | - Jochen Hampe
- Medical Department 1, University Hospital Dresden, Technical University Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technical University Dresden, Dresden, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany.
| | - J Gray Camp
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany.
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland.
| | - Barbara Treutlein
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany.
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
| |
Collapse
|
34
|
Nishikawa Y. Aberrant differentiation and proliferation of hepatocytes in chronic liver injury and liver tumors. Pathol Int 2024; 74:361-378. [PMID: 38837539 PMCID: PMC11551836 DOI: 10.1111/pin.13441] [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: 03/09/2024] [Revised: 04/29/2024] [Accepted: 05/12/2024] [Indexed: 06/07/2024]
Abstract
Chronic liver injury induces liver cirrhosis and facilitates hepatocarcinogenesis. However, the effects of this condition on hepatocyte proliferation and differentiation are unclear. We showed that rodent hepatocytes display a ductular phenotype when they are cultured within a collagenous matrix. This process involves transdifferentiation without the emergence of hepatoblastic features and is at least partially reversible. During the ductular reaction in chronic liver diseases with progressive fibrosis, some hepatocytes, especially those adjacent to ectopic ductules, demonstrate ductular transdifferentiation, but the majority of increased ductules originate from the existing bile ductular system that undergoes extensive remodeling. In chronic injury, hepatocyte proliferation is weak but sustained, and most regenerative nodules in liver cirrhosis are composed of clonally proliferating hepatocytes, suggesting that a small fraction of hepatocytes maintain their proliferative capacity in chronic injury. In mouse hepatocarcinogenesis models, hepatocytes activate the expression of various fetal/neonatal genes, indicating that these cells undergo dedifferentiation. Hepatocyte-specific somatic integration of various oncogenes in mice demonstrated that hepatocytes may be the cells of origin for a broad spectrum of liver tumors through transdifferentiation and dedifferentiation. In conclusion, the phenotypic plasticity and heterogeneity of mature hepatocytes are important for understanding the pathogenesis of chronic liver diseases and liver tumors.
Collapse
Affiliation(s)
- Yuji Nishikawa
- President's OfficeAsahikawa Medical UniversityAsahikawaHokkaidoJapan
| |
Collapse
|
35
|
Song G, Feng G, Li Q, Peng J, Ge W, Long Y, Cui Z. Transcriptomic Characterization of Key Factors and Signaling Pathways for the Regeneration of Partially Hepatectomized Liver in Zebrafish. Int J Mol Sci 2024; 25:7212. [PMID: 39000319 PMCID: PMC11241411 DOI: 10.3390/ijms25137212] [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: 05/08/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Liver regeneration induced by partial hepatectomy (PHx) has attracted intensive research interests due to the great significance for liver resection and transplantation. The zebrafish (Danio rerio) is an excellent model to study liver regeneration. In the fish subjected to PHx (the tip of the ventral lobe was resected), the lost liver mass could be fully regenerated in seven days. However, the regulatory mechanisms underlying the liver regeneration remain largely unknown. In this study, gene expression profiles during the regeneration of PHx-treated liver were explored by RNA sequencing (RNA-seq). The genes responsive to the injury of PHx treatment were identified and classified into different clusters based on the expression profiles. Representative gene ontology (GO) enrichments for the early responsive genes included hormone activity, ribosome biogenesis and rRNA processing, etc., while the late responsive genes were enriched in biological processes such as glutathione metabolic process, antioxidant activity and cellular detoxification. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichments were also identified for the differentially expressed genes (DEGs) between the time-series samples and the sham controls. The proteasome was overrepresented by the up-regulated genes at all of the sampling time points. Inhibiting proteasome activity by the application of MG132 to the fish enhanced the expression of Pcna (proliferating cell nuclear antigen), an indicator of hepatocyte proliferation after PHx. Our data provide novel insights into the molecular mechanisms underlying the regeneration of PHx-treated liver.
Collapse
Affiliation(s)
- Guili Song
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Guohui Feng
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Qing Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Jinrong Peng
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei Ge
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Yong Long
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Zongbin Cui
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| |
Collapse
|
36
|
Li T, Zhong W, Li M, Shao Z, Zhang G, Wang W, Gao Z, Tan X, Xu Z, Luo F, Song G. TRIM26 deficiency enhancing liver regeneration through macrophage polarization and β-catenin pathway activation. Cell Death Dis 2024; 15:453. [PMID: 38926362 PMCID: PMC11208526 DOI: 10.1038/s41419-024-06798-0] [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: 12/12/2023] [Revised: 05/25/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024]
Abstract
Liver regeneration is a complex process involving the crosstalk between parenchymal and non-parenchymal cells, especially macrophages. However, the underlying mechanisms remain incompletely understood. Here, we identify the E3 ubiquitin ligase TRIM26 as a crucial regulator of liver regeneration. Following partial hepatectomy or acute liver injury induced by carbon tetrachloride, Trim26 knockout mice exhibit enhanced hepatocyte proliferation compared to wild-type controls, while adeno-associated virus (AAV)-mediated overexpression of Trim26 reverses the promotional effects. Mechanistically, Trim26 deficiency promotes the recruitment of macrophages to the liver and their polarization towards pro-inflammatory M1 phenotype. These M1 macrophages secrete Wnts, including Wnt2, which subsequently stimulate hepatocyte proliferation through the activation of Wnt/β-catenin signaling. In hepatocytes, Trim26 knockdown reduces the ubiquitination and degradation of β-catenin, thereby further enhancing Wnt/β-catenin signaling. Pharmacological inhibition of Wnt/β-catenin pathway by ICG-001 or depletion of macrophages by clodronate liposomes diminishes the pro-regenerative effects of Trim26 deficiency. Moreover, bone marrow transplantation experiments provide evidence that Trim26 knockout in myeloid cells alone can also promote liver regeneration, highlighting the critical role of macrophage Trim26 in this process. Taken together, our study uncovers TRIM26 as a negative regulator of liver regeneration by modulating macrophage polarization and Wnt/β-catenin signaling in hepatocytes, providing a potential therapeutic target for promoting liver regeneration in clinical settings.
Collapse
Affiliation(s)
- Tingting Li
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Wei Zhong
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Mengqi Li
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Zile Shao
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Gongye Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Weiwei Wang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Zhixing Gao
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Xuemei Tan
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Ziyi Xu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Fanghong Luo
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Gang Song
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
37
|
McGinn M, Rabender C, Mikkelsen R, Yakovlev V. Hepatocyte-derived extracellular vesicles regulate liver regeneration after partial hepatectomy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600679. [PMID: 38979255 PMCID: PMC11230358 DOI: 10.1101/2024.06.25.600679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
While significant progress has been made in understanding different aspects of liver regeneration, the molecular mechanisms responsible for the initiation and termination of cell proliferation in the liver after massive loss or injury of liver tissue remain unknown. The loss of liver mass affects tissue-specific mitogenic inhibitors in the blood, which in turn regulate the proliferation of remaining hepatocytes and liver regeneration. Although well described in a number of publications, which inhibitory substances or "sensor molecules" control the regeneration mechanisms to properly maintain liver size remain unknown. Extracellular vesicles (EVs) are nano-sized, membrane-limited structures secreted by cells into the extracellular space. Their proposed role is stable intercellular carriers of proteins and RNAs, mostly micro-RNA, from secreted to recipient cells. Taken up by the recipient cells, EVs can significantly modulate their biological functions. In the present study, using in vivo and in vitro models, we demonstrate that hepatocyte proliferation and liver regeneration are regulated by EVs secreted by hepatocytes into the bloodstream. This regulation is carried out through a negative feedback mechanism, which explains the very precise regeneration of liver tissue after massive damage. We also demonstrate that an essential component of this mechanism is RNA carried by hepatocyte-derived EVs. These findings open up a new and unexplored area of biology regarding the mechanisms involved in the homeostasis regulation of various constantly renewing tissues by maintaining the optimal size and correct ratio between differentiating and proliferating cells.
Collapse
Affiliation(s)
- Mina McGinn
- Department of Radiation Oncology, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Christopher Rabender
- Department of Radiation Oncology, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Ross Mikkelsen
- Department of Radiation Oncology, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Vasily Yakovlev
- Department of Radiation Oncology, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
38
|
Cao Y, Wang S, Zhang M, Lai B, Liang Y. PFKFB3-mediated glycolysis in hepatic stellate cells promotes liver regeneration. Biochem Biophys Res Commun 2024; 712-713:149958. [PMID: 38640731 DOI: 10.1016/j.bbrc.2024.149958] [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: 03/15/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
Hepatic stellate cells (HSCs) perform a significant function in liver regeneration (LR) by becoming active. We propose to investigate if activated HSCs enhance glycolysis via PFKFB3, an essential glycolytic regulator, and whether targeting this pathway could be beneficial for LR. The liver and isolated HSCs of mice subjected to 2/3 partial hepatectomy (PHx) exhibited a significant rise in PFKFB3 expression, as indicated by quantitative RT-PCR analyses and Western blotting. Also, the primary HSCs of mice subjected to PHx have a significant elevation of the glycolysis level. Knocking down PFKFB3 significantly diminished the enhancement of glycolysis by PDGF in human LX2 cells. The hepatocyte proliferation in mice treated with PHx was almost completely prevented when the PFKFB3 inhibitor 3PO was administered, emerging that PFKFB3 is essential in LR. Furthermore, there was a decline in mRNA expression of immediate early genes and proinflammatory cytokines. In terms of mechanism, both the p38 MAP kinase and ERK1/2 phosphorylation in LO2 cells and LO2 proliferation were significantly reduced by the conditioned medium (CM) obtained from LX2 cells with either PFKFB3 knockdown or inhibition. Compared to the control group, isolated hepatocytes from 3PO-treated mice showed decreased p38 MAP kinase and ERK1/2 phosphorylation and proliferation. Thus, LR after PHx involves the activation of PFKFB3 in HSCs, which enhances glycolysis and promotes lactate production, thereby facilitating hepatocyte proliferation via the p38/ERK MAPK signaling pathway.
Collapse
Affiliation(s)
- Yapeng Cao
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Siyu Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Min Zhang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Baochang Lai
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yanni Liang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xian Yang, 712046, China.
| |
Collapse
|
39
|
Sande-Melon M, Bergemann D, Fernández-Lajarín M, González-Rosa JM, Cox AG. Development of a hepatic cryoinjury model to study liver regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.24.550437. [PMID: 38948752 PMCID: PMC11212901 DOI: 10.1101/2023.07.24.550437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The liver is a remarkable organ that can regenerate in response to injury. Depending on the extent of injury, the liver can undergo compensatory hyperplasia or fibrosis. Despite decades of research, the molecular mechanisms underlying these processes are poorly understood. Here, we developed a new model to study liver regeneration based on cryoinjury. To visualise liver regeneration at cellular resolution, we adapted the CUBIC tissue-clearing approach. Hepatic cryoinjury induced a localised necrotic and apoptotic lesion characterised by inflammation and infiltration of innate immune cells. Following this initial phase, we observed fibrosis, which resolved as regeneration re-established homeostasis in 30 days. Importantly, this approach enables the comparison of healthy and injured parenchyma with an individual animal, providing unique advantages to previous models. In summary, the hepatic cryoinjury model provides a fast and reproducible method for studying the cellular and molecular pathways underpinning fibrosis and liver regeneration.
Collapse
Affiliation(s)
- Marcos Sande-Melon
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - David Bergemann
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13 Street, 02129 MA, USA
- Harvard Medical School
| | - Miriam Fernández-Lajarín
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13 Street, 02129 MA, USA
- Harvard Medical School
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA 02467
| | - Juan Manuel González-Rosa
- Cardiovascular Research Centre, Massachusetts General Hospital Research Institute, Charlestown Navy Yard Campus, 149, 13 Street, 02129 MA, USA
- Harvard Medical School
- Biology Department, Morrissey College of Arts and Sciences, Boston College, Chestnut Hill, MA 02467
| | - Andrew G. Cox
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, 3000, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, Victoria, 3000, Australia
| |
Collapse
|
40
|
Yu T, Ma X, Cheng Y, Wang Z, Zhang G, Ding H, Yin J, Wang Y, Hu S. Amelioration of NAFLD by sleeve gastrectomy-triggered hepatocyte regeneration in mice - experimental research. Int J Surg 2024; 110:3307-3325. [PMID: 38573134 PMCID: PMC11175824 DOI: 10.1097/js9.0000000000001387] [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: 11/20/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Sleeve gastrectomy (SG) is known to alleviate non-alcoholic fatty liver disease (NAFLD) and restore liver function; however, its underlying mechanism remains unclear. MATERIALS AND METHODS We investigated the effect of SG on the metabolic phenotype of diet-induced obese (DIO) mice. Postoperative stained liver images were analyzed to determine the hepatocyte proliferation phenotype. Single-cell RNA sequencing was used to characterize the regeneration signals of the liver after SG in DIO mice, and real-time quantitative reverse transcription PCR was performed to verify the above results. We employed Olink proteomics to capture serum element changes and investigated the role of Yes1 protein in liver regeneration and carcinogenesis through the Hippo-YAP pathway. DIO mice were treated with YAP inhibitor verteporfin after SG mice to clarify whether SG-induced liver regeneration is related to the YAP signaling pathway. RESULTS SG significantly reduced NAFLD-associated dysfunction in hepatocytes and replaced them with fully functional hepatocytes, which have a high regenerative capacity across the entire liver. SG also enhanced the hepatic regenerative capacity, as demonstrated by SG combined with hepatic lobectomy in healthy mice. Yes1 protein was identified as the signaling molecule most closely related to classical regeneration signals. Our study showed that SG-enhanced proliferation and improved metabolism did not depend on YAP signaling. CONCLUSION SG can enhance hepatic regenerative capacity and improve liver metabolism. This study provides a better understanding of the mechanisms underlying SG-induced metabolic improvements.
Collapse
Affiliation(s)
- Tianming Yu
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University
| | - Xiaomin Ma
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan
| | - Yang Cheng
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University
| | - Zeyu Wang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan
| | - Guangyong Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan
| | - Huanxin Ding
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University
| | - Jialuo Yin
- College of Chemical Engineering, Qingdao University of Science and Technology
| | - Yifei Wang
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong Province
| | - Sanyuan Hu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| |
Collapse
|
41
|
Xue Y, Ruan Y, Wang Y, Xiao P, Xu J. Signaling pathways in liver cancer: pathogenesis and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:20. [PMID: 38816668 PMCID: PMC11139849 DOI: 10.1186/s43556-024-00184-0] [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: 01/04/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
Liver cancer remains one of the most prevalent malignancies worldwide with high incidence and mortality rates. Due to its subtle onset, liver cancer is commonly diagnosed at a late stage when surgical interventions are no longer feasible. This situation highlights the critical role of systemic treatments, including targeted therapies, in bettering patient outcomes. Despite numerous studies on the mechanisms underlying liver cancer, tyrosine kinase inhibitors (TKIs) are the only widely used clinical inhibitors, represented by sorafenib, whose clinical application is greatly limited by the phenomenon of drug resistance. Here we show an in-depth discussion of the signaling pathways frequently implicated in liver cancer pathogenesis and the inhibitors targeting these pathways under investigation or already in use in the management of advanced liver cancer. We elucidate the oncogenic roles of these pathways in liver cancer especially hepatocellular carcinoma (HCC), as well as the current state of research on inhibitors respectively. Given that TKIs represent the sole class of targeted therapeutics for liver cancer employed in clinical practice, we have particularly focused on TKIs and the mechanisms of the commonly encountered phenomena of its resistance during HCC treatment. This necessitates the imperative development of innovative targeted strategies and the urgency of overcoming the existing limitations. This review endeavors to shed light on the utilization of targeted therapy in advanced liver cancer, with a vision to improve the unsatisfactory prognostic outlook for those patients.
Collapse
Affiliation(s)
- Yangtao Xue
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yeling Ruan
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yali Wang
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Peng Xiao
- Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Junjie Xu
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China.
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China.
- Zhejiang University Cancer Center, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China.
| |
Collapse
|
42
|
Zhang Q, Yu T, Tan H, Shi H. Hepatic recruitment of myeloid-derived suppressor cells upon liver injury promotes both liver regeneration and fibrosis. BMC Gastroenterol 2024; 24:163. [PMID: 38745150 PMCID: PMC11092103 DOI: 10.1186/s12876-024-03245-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND The liver regeneration is a highly complicated process depending on the close cooperations between the hepatocytes and non-parenchymal cells involving various inflammatory cells. Here, we explored the role of myeloid-derived suppressor cells (MDSCs) in the processes of liver regeneration and liver fibrosis after liver injury. METHODS We established four liver injury models of mice including CCl4-induced liver injury model, bile duct ligation (BDL) model, concanavalin A (Con A)-induced hepatitis model, and lipopolysaccharide (LPS)-induced hepatitis model. The intrahepatic levels of MDSCs (CD11b+Gr-1+) after the liver injury were detected by flow cytometry. The effects of MDSCs on liver tissues were analyzed in the transwell co-culture system, in which the MDSCs cytokines including IL-10, VEGF, and TGF-β were measured by ELISA assay and followed by being blocked with specific antibodies. RESULTS The intrahepatic infiltrations of MDSCs with surface marker of CD11b+Gr-1+ remarkably increased after the establishment of four liver injury models. The blood served as the primary reservoir for hepatic recruitment of MDSCs during the liver injury, while the bone marrow appeared play a compensated role in increasing the number of MDSCs at the late stage of the inflammation. The recruited MDSCs in injured liver were mainly the M-MDSCs (CD11b+Ly6G-Ly6Chigh) featured by high expression levels of cytokines including IL-10, VEGF, and TGF-β. Co-culture of the liver tissues with MDSCs significantly promoted the proliferation of both hepatocytes and hepatic stellate cells (HSCs). CONCLUSIONS The dramatically and quickly infiltrated CD11b+Gr-1+ MDSCs in injured liver not only exerted pro-proliferative effects on hepatocytes, but also accounted for the activation of profibrotic HSCs.
Collapse
Affiliation(s)
- Qiongwen Zhang
- Department of Head and Neck Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Ting Yu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Huaicheng Tan
- Department of Head and Neck Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Huashan Shi
- Department of Head and Neck Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China.
| |
Collapse
|
43
|
de Haan LR, van Golen RF, Heger M. Molecular Pathways Governing the Termination of Liver Regeneration. Pharmacol Rev 2024; 76:500-558. [PMID: 38697856 DOI: 10.1124/pharmrev.123.000955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/24/2024] [Accepted: 02/08/2024] [Indexed: 05/05/2024] Open
Abstract
The liver has the unique capacity to regenerate, and up to 70% of the liver can be removed without detrimental consequences to the organism. Liver regeneration is a complex process involving multiple signaling networks and organs. Liver regeneration proceeds through three phases: the initiation phase, the growth phase, and the termination phase. Termination of liver regeneration occurs when the liver reaches a liver-to-body weight that is required for homeostasis, the so-called "hepatostat." The initiation and growth phases have been the subject of many studies. The molecular pathways that govern the termination phase, however, remain to be fully elucidated. This review summarizes the pathways and molecules that signal the cessation of liver regrowth after partial hepatectomy and answers the question, "What factors drive the hepatostat?" SIGNIFICANCE STATEMENT: Unraveling the pathways underlying the cessation of liver regeneration enables the identification of druggable targets that will allow us to gain pharmacological control over liver regeneration. For these purposes, it would be useful to understand why the regenerative capacity of the liver is hampered under certain pathological circumstances so as to artificially modulate the regenerative processes (e.g., by blocking the cessation pathways) to improve clinical outcomes and safeguard the patient's life.
Collapse
Affiliation(s)
- Lianne R de Haan
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, China (L.R.d.H., M.H.); Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands (L.R.d.H.); Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands (R.F.v.G.); Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands (M.H.); and Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands (M.H.)
| | - Rowan F van Golen
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, China (L.R.d.H., M.H.); Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands (L.R.d.H.); Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands (R.F.v.G.); Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands (M.H.); and Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands (M.H.)
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, China (L.R.d.H., M.H.); Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands (L.R.d.H.); Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands (R.F.v.G.); Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands (M.H.); and Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands (M.H.)
| |
Collapse
|
44
|
Elchaninov A, Vishnyakova P, Kuznetsova M, Lokhonina A, Soboleva A, Trofimov D, Fatkhudinov T, Sukhikh G. Mimicking the cellular environment does not cause monocyte-derived macrophages to become phenotypically similar to Kupffer cells. Immunol Cell Biol 2024; 102:381-395. [PMID: 38629182 DOI: 10.1111/imcb.12746] [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: 10/03/2023] [Revised: 02/24/2024] [Accepted: 03/19/2024] [Indexed: 05/12/2024]
Abstract
Resident macrophages of various mammalian organs are characterized by several distinctive features in their gene expression profile and phenotype, including involvement in the regulation of organ functions, as well as reduced sensitivity to proinflammatory activation factors. The reasons for the formation of such a specific phenotype remain the subject of intensive research. Some papers emphasize the role of the origin of organ macrophages. Other studies indicate that monocytes that develop in the red bone marrow are also able to form resident macrophages with a phenotype characteristic of a particular organ, but this requires appropriate microenvironmental conditions. In this article, we studied the possibility of differentiation of monocyte-derived macrophages into cells with a Kupffer-like phenotype under the influence of the main stromal components of Kupffer cells macrophage niche: Ito cells, liver sinusoid endotheliocytes and hepatocyte growth factor (HGF). It was found that Kupffer cells are characterized by several features, including increased expression of transcription factors Spic and Id3, as well as MUP family genes, Clusterin and Ngp genes. In addition, Kupffer cells were characterized by a higher proliferative activity. The expression of marker genes of Kupffer cells (i.e. Id3, Spic, Marco and Timd4) increased in monocyte-derived macrophages during coculture with Ito cells, liver sinusoid endothelial cells, macrophage colony-stimulating factor and HGF cells only by 3 days. However, the expression level of these genes was always higher in Kupffer cells. In addition, a complete coincidence of the expressed gene profile in monocyte-derived macrophages and Kupffer cells did not occur even after 3 days of culturing.
Collapse
Affiliation(s)
- Andrey Elchaninov
- Laboratory of Growth and Development, Avtsyn Research Institute of Human Morphology of FSBI, Petrovsky National Research Centre of Surgery, Moscow, Russia
- Histology Department, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Polina Vishnyakova
- Laboratory of Regenerative Medicine, Institute of Translational Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
- Histology Department, Medical Institute, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| | - Maria Kuznetsova
- Laboratory of molecular research methods, Institute of Reproductive Genetics, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Anastasiya Lokhonina
- Laboratory of Regenerative Medicine, Institute of Translational Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
- Histology Department, Medical Institute, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| | - Anna Soboleva
- Laboratory of Growth and Development, Avtsyn Research Institute of Human Morphology of FSBI, Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - Dmitry Trofimov
- Laboratory of molecular research methods, Institute of Reproductive Genetics, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Timur Fatkhudinov
- Laboratory of Growth and Development, Avtsyn Research Institute of Human Morphology of FSBI, Petrovsky National Research Centre of Surgery, Moscow, Russia
- Histology Department, Medical Institute, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| | - Gennady Sukhikh
- Laboratory of Regenerative Medicine, Institute of Translational Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| |
Collapse
|
45
|
Takahashi K, Gosho M, Miyazaki Y, Nakahashi H, Shimomura O, Furuya K, Doi M, Owada Y, Ogawa K, Ohara Y, Akashi Y, Enomoto T, Hashimoto S, Oda T. Preoperative albumin-bilirubin score and liver resection percentage determine postoperative liver regeneration after partial hepatectomy. World J Gastroenterol 2024; 30:2006-2017. [PMID: 38681122 PMCID: PMC11045494 DOI: 10.3748/wjg.v30.i14.2006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/05/2024] [Accepted: 03/25/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND The success of liver resection relies on the ability of the remnant liver to regenerate. Most of the knowledge regarding the pathophysiological basis of liver regeneration comes from rodent studies, and data on humans are scarce. Additionally, there is limited knowledge about the preoperative factors that influence postoperative regeneration. AIM To quantify postoperative remnant liver volume by the latest volumetric software and investigate perioperative factors that affect posthepatectomy liver regeneration. METHODS A total of 268 patients who received partial hepatectomy were enrolled. Patients were grouped into right hepatectomy/trisegmentectomy (RH/Tri), left hepatectomy (LH), segmentectomy (Seg), and subsegmentectomy/nonanatomical hepatectomy (Sub/Non) groups. The regeneration index (RI) and late regeneration rate were defined as (postoperative liver volume)/[total functional liver volume (TFLV)] × 100 and (RI at 6-months - RI at 3-months)/RI at 6-months, respectively. The lower 25th percentile of RI and the higher 25th percentile of late regeneration rate in each group were defined as "low regeneration" and "delayed regeneration". "Restoration to the original size" was defined as regeneration of the liver volume by more than 90% of the TFLV at 12 months postsurgery. RESULTS The numbers of patients in the RH/Tri, LH, Seg, and Sub/Non groups were 41, 53, 99 and 75, respectively. The RI plateaued at 3 months in the LH, Seg, and Sub/Non groups, whereas the RI increased until 12 months in the RH/Tri group. According to our multivariate analysis, the preoperative albumin-bilirubin (ALBI) score was an independent factor for low regeneration at 3 months [odds ratio (OR) 95%CI = 2.80 (1.17-6.69), P = 0.02; per 1.0 up] and 12 months [OR = 2.27 (1.01-5.09), P = 0.04; per 1.0 up]. Multivariate analysis revealed that only liver resection percentage [OR = 1.03 (1.00-1.05), P = 0.04] was associated with delayed regeneration. Furthermore, multivariate analysis demonstrated that the preoperative ALBI score [OR = 2.63 (1.00-1.05), P = 0.02; per 1.0 up] and liver resection percentage [OR = 1.02 (1.00-1.05), P = 0.04; per 1.0 up] were found to be independent risk factors associated with volume restoration failure. CONCLUSION Liver regeneration posthepatectomy was determined by the resection percentage and preoperative ALBI score. This knowledge helps surgeons decide the timing and type of rehepatectomy for recurrent cases.
Collapse
Affiliation(s)
- Kazuhiro Takahashi
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| | - Masahiko Gosho
- Department of Biostatistics, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| | - Yoshihiro Miyazaki
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| | - Hiromitsu Nakahashi
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| | - Osamu Shimomura
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| | - Kinji Furuya
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| | - Manami Doi
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| | - Yohei Owada
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| | - Koichi Ogawa
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| | - Yusuke Ohara
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| | - Yoshimasa Akashi
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| | - Tsuyoshi Enomoto
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| | - Shinji Hashimoto
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| | - Tatsuya Oda
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, University of Tsukuba, Tsukuba 3058-575, Ibaraki, Japan
| |
Collapse
|
46
|
Tan VWT, Salmi TM, Karamalakis AP, Gillespie A, Ong AJS, Balic JJ, Chan YC, Bladen CE, Brown KK, Dawson MA, Cox AG. SLAM-ITseq identifies that Nrf2 induces liver regeneration through the pentose phosphate pathway. Dev Cell 2024; 59:898-910.e6. [PMID: 38366599 DOI: 10.1016/j.devcel.2024.01.024] [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: 04/28/2023] [Revised: 12/07/2023] [Accepted: 01/24/2024] [Indexed: 02/18/2024]
Abstract
The liver exhibits a remarkable capacity to regenerate following injury. Despite this unique attribute, toxic injury is a leading cause of liver failure. The temporal processes by which the liver senses injury and initiates regeneration remain unclear. Here, we developed a transgenic zebrafish model wherein hepatocyte-specific expression of uracil phosphoribosyltransferase (UPRT) enabled the implementation of SLAM-ITseq to investigate the nascent transcriptome during initiation of liver injury and regeneration. Using this approach, we identified a rapid metabolic transition from the fed to the fasted state that was followed by induction of the nuclear erythroid 2-related factor (Nrf2) antioxidant program. We find that activation of Nrf2 in hepatocytes is required to induce the pentose phosphate pathway (PPP) and improve survival following liver injury. Mechanistically, we demonstrate that inhibition of the PPP disrupts nucleotide biosynthesis to prevent liver regeneration. Together, these studies provide fundamental insights into the mechanism by which early metabolic adaptation to injury facilitates tissue regeneration.
Collapse
Affiliation(s)
- Vicky W T Tan
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Talhah M Salmi
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Anthony P Karamalakis
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Andrea Gillespie
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Athena Jessica S Ong
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Jesse J Balic
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Yih-Chih Chan
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Cerys E Bladen
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Kristin K Brown
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia; Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mark A Dawson
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia; Department of Clinical Haematology, Peter MacCallum Cancer Centre & Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; Centre for Cancer Research, The University of Melbourne, Melbourne, VIC 3000, Australia.
| | - Andrew G Cox
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia; Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
47
|
Wang WC, Wu TH, Hung HC, Lee JC, Cheng CH, Wang YC, Lee CF, Wu TJ, Chou HS, Chan KM, Lee WC. Liver regeneration of living donor after liver donation for transplantation: Disparity in the left and right remnant liver. Medicine (Baltimore) 2024; 103:e37632. [PMID: 38579088 PMCID: PMC10994454 DOI: 10.1097/md.0000000000037632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/26/2024] [Indexed: 04/07/2024] Open
Abstract
Donor safety is crucial for living donor liver transplantation (LDLT), and sufficient liver regeneration significantly affects outcomes of living donors. This study aimed to investigate clinical factors associated with liver regeneration in living donors. The study retrospectively reviewed 380 living donors who underwent liver donation at Chang Gung Memorial Hospital in Linkou. The clinical characteristics and medical parameters of donors were analyzed and compared according to liver donation graft type. There were 355 donors (93.4%) with right hemi-liver donations and 25 donors (6.6%) with left hemi-liver donations. Left hemi-liver donors had a higher body mass index (BMI) and a larger ratio of remnant liver volume (RLV) to total liver volume (TLV). However, the 2 groups showed no significant difference in the liver regeneration ratio. The type of remnant liver (P < .001), RLV/body weight (P = .027), RLV/TLV (P < .001), serum albumin on postoperative day 7 and total bilirubin levels on postoperative day 30 were the most significant factors affecting liver regeneration in living donors. In conclusion, adequate liver regeneration is essential for donor outcome after liver donation. The remnant liver could eventually regenerate to an adequate volume similar to the initial TLV before liver donation. However, the remnant left hemi-liver had a faster growth rate than the remnant right hemi-liver in donors.
Collapse
Affiliation(s)
- Wei-Cheng Wang
- Department of General Surgery and Chang Gung Transplantation Institute, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Tsung-Han Wu
- Department of General Surgery and Chang Gung Transplantation Institute, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Hao-Chien Hung
- Department of General Surgery and Chang Gung Transplantation Institute, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Jin-Chiao Lee
- Department of General Surgery and Chang Gung Transplantation Institute, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chih-Hsien Cheng
- Department of General Surgery and Chang Gung Transplantation Institute, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yu-Chao Wang
- Department of General Surgery and Chang Gung Transplantation Institute, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chen-Fang Lee
- Department of General Surgery and Chang Gung Transplantation Institute, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ting-Jung Wu
- Department of General Surgery and Chang Gung Transplantation Institute, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Hong-Shiue Chou
- Department of General Surgery and Chang Gung Transplantation Institute, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Kun-Ming Chan
- Department of General Surgery and Chang Gung Transplantation Institute, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Wei-Chen Lee
- Department of General Surgery and Chang Gung Transplantation Institute, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
48
|
Namoto K, Baader C, Orsini V, Landshammer A, Breuer E, Dinh KT, Ungricht R, Pikiolek M, Laurent S, Lu B, Aebi A, Schönberger K, Vangrevelinghe E, Evrova O, Sun T, Annunziato S, Lachal J, Redmond E, Wang L, Wetzel K, Capodieci P, Turner J, Schutzius G, Unterreiner V, Trunzer M, Buschmann N, Behnke D, Machauer R, Scheufler C, Parker CN, Ferro M, Grevot A, Beyerbach A, Lu WY, Forbes SJ, Wagner J, Bouwmeester T, Liu J, Sohal B, Sahambi S, Greenbaum LE, Lohmann F, Hoppe P, Cong F, Sailer AW, Ruffner H, Glatthar R, Humar B, Clavien PA, Dill MT, George E, Maibaum J, Liberali P, Tchorz JS. NIBR-LTSi is a selective LATS kinase inhibitor activating YAP signaling and expanding tissue stem cells in vitro and in vivo. Cell Stem Cell 2024; 31:554-569.e17. [PMID: 38579685 DOI: 10.1016/j.stem.2024.03.003] [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: 07/26/2022] [Revised: 01/24/2024] [Accepted: 03/06/2024] [Indexed: 04/07/2024]
Abstract
The YAP/Hippo pathway is an organ growth and size regulation rheostat safeguarding multiple tissue stem cell compartments. LATS kinases phosphorylate and thereby inactivate YAP, thus representing a potential direct drug target for promoting tissue regeneration. Here, we report the identification and characterization of the selective small-molecule LATS kinase inhibitor NIBR-LTSi. NIBR-LTSi activates YAP signaling, shows good oral bioavailability, and expands organoids derived from several mouse and human tissues. In tissue stem cells, NIBR-LTSi promotes proliferation, maintains stemness, and blocks differentiation in vitro and in vivo. NIBR-LTSi accelerates liver regeneration following extended hepatectomy in mice. However, increased proliferation and cell dedifferentiation in multiple organs prevent prolonged systemic LATS inhibition, thus limiting potential therapeutic benefit. Together, we report a selective LATS kinase inhibitor agonizing YAP signaling and promoting tissue regeneration in vitro and in vivo, enabling future research on the regenerative potential of the YAP/Hippo pathway.
Collapse
Affiliation(s)
- Kenji Namoto
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland.
| | - Clara Baader
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Vanessa Orsini
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Eva Breuer
- University Hospital Zurich (USZ), Zurich, Switzerland
| | - Kieu Trinh Dinh
- German Cancer Research Center (DKFZ) Heidelberg, Research Group Experimental Hepatology, Inflammation and Cancer, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | | | | | | | - Bo Lu
- Biomedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Alexandra Aebi
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | | | - Olivera Evrova
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Tianliang Sun
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland; Division of Liver Diseases, Institute for Regenerative Medicine, Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Julie Lachal
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Emily Redmond
- Biomedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Louis Wang
- Biomedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Kristie Wetzel
- Biomedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | | | | | - Gabi Schutzius
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Markus Trunzer
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Dirk Behnke
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | | | | | - Magali Ferro
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Armelle Grevot
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Wei-Yu Lu
- University of Edinburgh, Center for Inflammation Research, Edinburgh, UK
| | - Stuart J Forbes
- University of Edinburgh, Center for Regenerative Medicine, Edinburgh, UK
| | - Jürgen Wagner
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Jun Liu
- Biomedical Research, Novartis Pharma AG, La Jolla, CA, USA
| | - Bindi Sohal
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | | | - Felix Lohmann
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Philipp Hoppe
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Feng Cong
- Biomedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | | | - Heinz Ruffner
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Ralf Glatthar
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Bostjan Humar
- University Hospital Zurich (USZ), Zurich, Switzerland
| | | | - Michael T Dill
- German Cancer Research Center (DKFZ) Heidelberg, Research Group Experimental Hepatology, Inflammation and Cancer, Heidelberg, Germany; Department of Gastroenterology, Infectious Diseases and Intoxication, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Jürgen Maibaum
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Jan S Tchorz
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland.
| |
Collapse
|
49
|
Guo Q, Wang ML, Zhong K, Li JL, Jiang TM, Wen H, Aji T, Shao YM. Portal vein embolization combined with ex vivo liver resection and autotransplantation: A novel treatment strategy for end-stage and metastatic hepatic alveolar echinococcosis. Hepatobiliary Pancreat Dis Int 2024; 23:210-216. [PMID: 37295974 DOI: 10.1016/j.hbpd.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 05/24/2023] [Indexed: 06/12/2023]
Affiliation(s)
- Qiang Guo
- Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China; Clinical Medical Research Center of Echinococcosis and Hepatobiliary Disease of Xinjiang Uygur Autonomous Region, Urumqi 830054, China
| | - Mao-Lin Wang
- Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China; Clinical Medical Research Center of Echinococcosis and Hepatobiliary Disease of Xinjiang Uygur Autonomous Region, Urumqi 830054, China
| | - Kai Zhong
- Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China; Clinical Medical Research Center of Echinococcosis and Hepatobiliary Disease of Xinjiang Uygur Autonomous Region, Urumqi 830054, China
| | - Jia-Long Li
- Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China; Clinical Medical Research Center of Echinococcosis and Hepatobiliary Disease of Xinjiang Uygur Autonomous Region, Urumqi 830054, China
| | - Tie-Min Jiang
- Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China; Clinical Medical Research Center of Echinococcosis and Hepatobiliary Disease of Xinjiang Uygur Autonomous Region, Urumqi 830054, China
| | - Hao Wen
- Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China; Clinical Medical Research Center of Echinococcosis and Hepatobiliary Disease of Xinjiang Uygur Autonomous Region, Urumqi 830054, China; State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi 830054, China
| | - Tuerganaili Aji
- Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China; Clinical Medical Research Center of Echinococcosis and Hepatobiliary Disease of Xinjiang Uygur Autonomous Region, Urumqi 830054, China
| | - Ying-Mei Shao
- Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China; Clinical Medical Research Center of Echinococcosis and Hepatobiliary Disease of Xinjiang Uygur Autonomous Region, Urumqi 830054, China.
| |
Collapse
|
50
|
Martins BS, Fogelman N, Tate M, Hermes GH, Sinha R. Effects of prazosin treatment on liver enzymes are moderated by alcohol withdrawal symptoms in individuals with alcohol use disorder. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:507-515. [PMID: 38258493 PMCID: PMC10939766 DOI: 10.1111/acer.15263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/24/2024]
Abstract
BACKGROUND Alcohol use disorder (AUD) is associated with significant liver pathology marked by elevated liver enzymes. Prazosin, an alpha1-noradrenergic antagonist significantly improves alcohol drinking outcomes in individuals with alcohol withdrawal symptoms (AW), but effects on liver enzymes are unknown. We assessed the effects of prazosin treatment on the liver enzymes alanine transaminase (ALT), aspartate transaminase (AST), and gamma-glutamyltransferase (GGT) in individuals with AUD. METHODS Participants (N=100) with AUD were enrolled in a 12-week randomized controlled trial and received either placebo or 16 mg/day of prazosin. Whole blood was drawn from 92 participants to measure liver enzyme levels every 4 weeks, and severity of AW was assessed weekly. Analysis predicting liver function outcomes used linear mixed effects models. RESULTS Controlling for alcohol consumption, a significant AW × treatment effect was seen for ALT (p < 0.05), AST (p < 0.05) and GGT (p < 0.01). Additionally, AST (b = 0.2, p < 0.01), ALT (b = 0.2, p < 0.05), and GGT (b = 0.3, p < 0.01) were elevated in individuals with higher AW in the placebo but not in the prazosin group (AST: p > 0.66; ALT: p > 0.65). Only in the prazosin group were lower GGT levels associated with higher withdrawal severity (b = -0.16, p < 0.05). CONCLUSIONS We found an interaction of alcohol withdrawal symptoms and prazosin treatment on liver enzyme levels, which were not influenced by week in the trial or the amount of alcohol consumed. Together, these findings suggest that prazosin treatment reduces liver enzymes over the course of AUD treatment among individuals with significant AW, though replication to establish clinical utility is needed.
Collapse
Affiliation(s)
- Bradford S. Martins
- Yale Stress Center, Yale University, 2 Church St. South Suite 209, New Haven, CT, 06519. USA
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Nia Fogelman
- Yale Stress Center, Yale University, 2 Church St. South Suite 209, New Haven, CT, 06519. USA
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Marshall Tate
- Yale Stress Center, Yale University, 2 Church St. South Suite 209, New Haven, CT, 06519. USA
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Gretchen H. Hermes
- Yale Stress Center, Yale University, 2 Church St. South Suite 209, New Haven, CT, 06519. USA
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Rajita Sinha
- Yale Stress Center, Yale University, 2 Church St. South Suite 209, New Haven, CT, 06519. USA
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| |
Collapse
|