1
|
Ding H, Lv H, Sui M, Wang X, Sun Y, Tian M, Ma S, Xue Y, Zhang M, Wang X, Qi J, Wang L, Zhu Q. Interaction of neuropilin-1 and hepatocyte growth factor/C-Met pathway in liver fibrosis progression in hepatocyte-specific NRP-1 knockout mice. J Gastroenterol 2025:10.1007/s00535-025-02262-8. [PMID: 40419692 DOI: 10.1007/s00535-025-02262-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 04/29/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND Hepatocyte growth factor (HGF)/c-Met signaling critically influences liver fibrosis, but its interaction with neuropilin-1 (NRP-1) in hepatocytes remains unclear. We investigated the role of hepatocyte-specific NRP-1 deletion in liver fibrosis progression and its relationship with the HGF/c-Met pathway. METHODS Hepatocyte-specific NRP-1 knockout mice were generated using the Cre-lox system, and liver fibrosis was induced by carbon tetrachloride injections or a methionine- and choline-deficient diet. Fibrosis severity, hepatocyte injury, and cytokine secretion were evaluated via histology, biochemical assays, and molecular analyses in isolated hepatocytes. In vitro experiments were conducted in primary hepatocytes and Huh7 cells using lentiviral overexpression and knockdown of NRP-1. Chromatin immunoprecipitation and dual-luciferase reporter assays were performed to analyze transcription factor binding to the NRP-1 promoter. RESULTS Hepatocyte NRP-1 expression increased significantly during liver fibrosis and was positively correlated with HGF/c-Met expression and fibrosis severity. In vivo, NRP-1 inhibition reduced extracellular matrix accumulation and abnormal angiogenesis in Alb-Cre NRP-1f/f mice. In vitro, NRP-1 blockade inhibited c-Met activation and reduced transforming growth factor-beta and vascular endothelial growth factor secretion in hepatocytes. NRP-1 functioned as a co-receptor for HGF/c-Met, with HGF upregulating NRP-1 expression at transcript and protein levels. NRP-1 promoted fibrosis through the Met/extracellular signal-regulated kinase pathway. Furthermore, HGF increased retinoic acid receptor alpha expression, promoting NRP-1 transcription. CONCLUSIONS HGF-induced upregulation of hepatocyte NRP-1, mediated by RARA binding to its promoter, drives liver fibrosis through c-Met pathway activation, highlighting NRP-1 as a potential therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Han Ding
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan City, Shandong Province, China
| | - Huanran Lv
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan City, Shandong Province, China
| | - Minghao Sui
- Department of Gastroenterology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan City, Shandong Province, China
| | - Xinyu Wang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan City, Shandong Province, China
| | - Yanning Sun
- Urology Department, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan City, Shandong Province, China
| | - Miaomiao Tian
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan City, Shandong Province, China
| | - Shujun Ma
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan City, Shandong Province, China
| | - Yuchan Xue
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan City, Shandong Province, China
| | - Miao Zhang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan City, Shandong Province, China
| | - Xin Wang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan City, Shandong Province, China
| | - Jianni Qi
- Department of Key Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan City, Shandong Province, China
| | - Le Wang
- Department of Geriatrics, Department of Geriatric Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan City, Shandong Province, China.
| | - Qiang Zhu
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Huaiyin District, Jinan City, Shandong Province, China.
| |
Collapse
|
2
|
Wu Q, Ge Z, Lv C, He Q. Interacting roles of gut microbiota and T cells in the development of autoimmune hepatitis. Front Immunol 2025; 16:1584001. [PMID: 40491914 PMCID: PMC12146343 DOI: 10.3389/fimmu.2025.1584001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 04/29/2025] [Indexed: 06/11/2025] Open
Abstract
Autoimmune hepatitis (AIH) is a progressive liver inflammatory disease mediated by an autoimmune response, with an increasing incidence rate. In severe cases, AIH will rapidly progress to liver cirrhosis and liver failure and even lead to death. The gut microbiota is a complex ecosystem that significantly regulates physiological and pathological processes among various digestive system diseases. It is widely acknowledged that there is a critical correlation between AIH and the gut microbiota. Numerous studies have demonstrated that the composition of gut microbiota in individuals with AIH differs markedly from that of healthy subjects. Immune cells, especially T cells, are pivotal in the development of AIH, closely interacting with the gut microbiota. In this review, we discuss the regulatory role of the gut microbiota in T cell-mediated development of AIH, as well as the effect of T cells on the composition of the gut microbiota in AIH. By modulating gut microbiota or immunity pathways, novel opportunities are provided to regulate the balance of the immune-microbial microenvironment, targeting the dual factor for autoimmune hepatitis therapies.
Collapse
Affiliation(s)
| | | | - Chengyu Lv
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qifeng He
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Zhou L, Shi Z, Yang X, Zeng J, You Z, Zhang Y, Zhu Z, Liu Z, Niu Y, Yu H, He J, Long Y, Wu Z, Zhang Y, Lyu C, Deng L, Wang Y, Wu C, Du Y. Tension-induced directional migration of hepatic stellate cells potentially coordinates liver fibrosis progression. Nat Biomed Eng 2025:10.1038/s41551-025-01381-0. [PMID: 40410557 DOI: 10.1038/s41551-025-01381-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 03/25/2025] [Indexed: 05/25/2025]
Abstract
Liver fibrosis is an over-reacted wound healing that becomes lethal in its late stage, when hepatic stellate cells (HSCs) trigger fibrotic response, proliferation of connective tissue and build-up of directional fibrous tissue bands (septa). Current in vitro models of liver fibrosis cannot reproduce liver lobule structure and the dynamic formation of septa at the same time, and the known biochemical cues underlying the progression of liver fibrosis cannot explain directional formation of fibrotic tissue. Here we report a microfabricated in vitro model that reproduces both the hexagonal liver lobule structure and the dynamic directionality of septa formation. By using collagen and primary mouse HSCs or human HSC lines, we found that tension was necessary to coordinate the cell migration that contributes to the band-like cell distribution and that HSCs sensed directional biophysical cues through liquid-liquid phase separation. This system allows the study of the biophysical interaction of HSCs and collagen during the formation of septa structures, and could be used to deepen our understanding of liver fibrosis progression.
Collapse
Affiliation(s)
- Lyu Zhou
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Ziao Shi
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xuesi Yang
- School of Aerospace Engineering, Tsinghua University, Beijing, China
| | - Jia'nan Zeng
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Zhifeng You
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yuying Zhang
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Zhiyue Zhu
- Department of Mechanical and Industrial Engineering, Ted Rogers Centre For Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Zhiqiang Liu
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yudi Niu
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Hongsheng Yu
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Jinliang He
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Yi Long
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Zhaozhao Wu
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yan Zhang
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Cheng Lyu
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Liping Deng
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yuan Wang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Tumor Systems Biology, Beijing, China
| | - Congying Wu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Tumor Systems Biology, Beijing, China
| | - Yanan Du
- School of Biomedical Engineering, Tsinghua Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
4
|
Park S, Jeong I, Kim OK. Ginsenoside Rh2 Mitigates Endoplasmic Reticulum Stress-Induced Apoptosis and Inflammation and Through Inhibition of Hepatocyte-Macrophage Inflammatory Crosstalk. Nutrients 2025; 17:1682. [PMID: 40431422 PMCID: PMC12114235 DOI: 10.3390/nu17101682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2025] [Revised: 05/09/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Endoplasmic reticulum stress (ERS) contributes to hepatocyte inflammation, triggered by prolonged exposure to lipotoxicity, and promotes non-alcoholic fatty liver disease (NAFLD) progression by recruiting and activating hepatic macrophages, which accelerate fibrosis and exacerbate disease progression. Here, we aimed to evaluate the therapeutic potential of ginsenoside Rh2 (Rh2) in a cell model of NAFLD induced by the ERS inducer thapsigargin (THA). Methods: HepG2 cells were treated with THA to induce ERS and mimic NAFLD conditions. The effects of Rh2 on ERS, lipid accumulation, and apoptosis were assessed in HepG2 cells. Additionally, THP-1 cells were used to investigate macrophage activation upon exposure to conditioned medium (CM) from THA- and Rh2-treated HepG2 cells. Gene and protein expression of inflammatory and lipid synthesis markers were analyzed, as well as M1/M2 macrophage polarization markers. Results: Rh2 inhibited THA-induced apoptosis, ERS, and lipid accumulation in HepG2 cells. It also reduced the expression of lipid synthesis genes (SREBF1, FAS) and inflammatory markers (IL-6, IL-1β, TNF-α, MCP-1). CM from Rh2-treated HepG2 cells suppressed macrophage activation in THP-1 cells, decreased M1 polarization markers (CD80, CD86), and increased M2 markers (CD163, Arg1, MRC-1). Conclusions: These results suggest that Rh2 effectively suppresses inflammation and lipid storage in ERS-induced HepG2 cells while modulating the crosstalk between hepatocytes and macrophages. These findings underscore the potential of Rh2 as a promising therapeutic agent for the prevention and early intervention of NAFLD progression.
Collapse
Affiliation(s)
- Shinjung Park
- Division of Food and Nutrition, Chonnam National University, Gwangju 61186, Republic of Korea; (S.P.); (I.J.)
| | - Inae Jeong
- Division of Food and Nutrition, Chonnam National University, Gwangju 61186, Republic of Korea; (S.P.); (I.J.)
| | - Ok-Kyung Kim
- Division of Food and Nutrition, Chonnam National University, Gwangju 61186, Republic of Korea; (S.P.); (I.J.)
- Human Ecology Research Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
5
|
Accacha S, Barillas-Cerritos J, Srivastava A, Ross F, Drewes W, Gulkarov S, De Leon J, Reiss AB. From Childhood Obesity to Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) and Hyperlipidemia Through Oxidative Stress During Childhood. Metabolites 2025; 15:287. [PMID: 40422865 DOI: 10.3390/metabo15050287] [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: 03/10/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND/OBJECTIVES Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is rapidly becoming the most prevalent form of chronic liver disease in both pediatric and adult populations. It encompasses a wide spectrum of liver abnormalities, ranging from simple fat accumulation to severe conditions such as inflammation, fibrosis, cirrhosis, and liver cancer. Major risk factors for MASLD include obesity, insulin resistance, type 2 diabetes, and hypertriglyceridemia. METHODS This narrative review employed a comprehensive search of recent literature to identify the latest studies on the relationship between MAFLD and obesity, the health consequences and the latest treatment options to prevent long-term damage to the liver and other organs. Additionally, the article presents perspectives on diagnostic biomarkers. RESULTS Childhood obesity is linked to a multitude of comorbid conditions and remains a primary risk factor for adult obesity. This abnormal fat accumulation is known to have long-term detrimental effects into adulthood. Scientific evidence unequivocally demonstrates the role of obesity-related conditions, such as insulin resistance, dyslipidemia, and hyperglycemia, in the development and progression of MASLD. Oxidative stress, stemming from mitochondrial dysfunction, is a leading factor in MASLD. This review discusses the interconnections between oxidative stress, obesity, dyslipidemia, and MASLD. CONCLUSIONS Atherogenic dyslipidemia, oxidative stress, inflammation, insulin resistance, endothelial dysfunction, and cytokines collectively contribute to the development of MASLD. Potential treatment targets for MASLD are focused on prevention and the use of drugs to address obesity and elevated blood lipid levels.
Collapse
Affiliation(s)
- Siham Accacha
- Department of Pediatrics, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Julia Barillas-Cerritos
- Department of Pediatrics, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Ankita Srivastava
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Frances Ross
- Department of Pediatrics, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Wendy Drewes
- Department of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Shelly Gulkarov
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Joshua De Leon
- Department of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Allison B Reiss
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA
- Department of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| |
Collapse
|
6
|
McClune ME, Ebohon O, Dressler JM, Davis MM, Tupik JD, Lochhead RB, Booth CJ, Steere AC, Jutras BL. The peptidoglycan of Borrelia burgdorferi can persist in discrete tissues and cause systemic responses consistent with chronic illness. Sci Transl Med 2025; 17:eadr2955. [PMID: 40267217 DOI: 10.1126/scitranslmed.adr2955] [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: 06/24/2024] [Revised: 01/27/2025] [Accepted: 03/06/2025] [Indexed: 04/25/2025]
Abstract
Persistent symptoms after an acute infection is an emerging public health concern, but the pathobiology of such conditions is not well understood. One possible scenario involves the persistence of lingering antigen. We have previously reported that patients with postinfectious Lyme arthritis often harbor the peptidoglycan (PG) cell wall of Borrelia burgdorferi, the Lyme disease agent, in the synovial fluid of their inflamed joints after treatment. However, it is not yet known how B. burgdorferi PG persists, in what form, or if it may play a role in other postinfectious complications after Lyme disease. Using a murine model, we developed a real-time in vivo system to track B. burgdorferi PG as a function of cell wall chemistry and validated our findings using both molecular and cellular approaches. Unlike typical bacterial PG, the unique chemical properties of polymeric B. burgdorferi PG drive murine liver accumulation, where the cell wall material persists for weeks. Kupffer cells and hepatocytes phagocytose and retain B. burgdorferi PG and, although liver occupancy coincides with minimal pathology, both organ-specific and secreted protein profiles produced under these conditions bear some similarities to reported proteins enriched in patients with chronic illness after acute infection. Moreover, transcriptomic profiling indicated that B. burgdorferi PG affects energy metabolism in peripheral blood mononuclear cells. Our findings provide mechanistic insights into how a pathogenic molecule can persist after agent clearance, potentially contributing to illness after infection.
Collapse
Affiliation(s)
- Mecaila E McClune
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Human Center for Immunobiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Osamudiamen Ebohon
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Human Center for Immunobiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Jules M Dressler
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Marisela M Davis
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
| | - Juselyn D Tupik
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
- Department of Biomedical and Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Robert B Lochhead
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Carmen J Booth
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Allen C Steere
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Brandon L Jutras
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Human Center for Immunobiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
- Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
7
|
Jin Z, Li Y, Yi H, Wang M, Wang C, Du S, Zeng W, Zong Z. Pathogenetic development, diagnosis and clinical therapeutic approaches for liver metastasis from colorectal cancer (Review). Int J Oncol 2025; 66:22. [PMID: 39950314 PMCID: PMC11844340 DOI: 10.3892/ijo.2025.5728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/10/2025] [Indexed: 02/23/2025] Open
Abstract
Colorectal cancer (CRC) is a prevalent malignancy and a significant proportion of patients with CRC develop liver metastasis (CRLM), which is a major contributor to CRC‑related mortality. The present review aimed to comprehensively examine the pathogenetic development and diagnosis of CRLM and the clinical therapeutic approaches for treatment of this disease. The molecular mechanisms underlying CRLM were discussed, including the role of the tumour microenvironment and epithelial‑mesenchymal transition. The present review also highlighted the importance of early detection and the current challenges in predicting the development of CRLM. Various treatment strategies were reviewed, including surgical resection, chemotherapy and immunotherapy, and the potential of novel therapies, such as selective internal radiation therapy and Traditional Chinese Medicine. Despite recent advancements in treatment options, the treatment of CRLM remains a therapeutic challenge due to the complexity of the liver microenvironment and the heterogeneity of CRC. The present review emphasized the need for a multidisciplinary approach and the integration of emerging therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Zhenhua Jin
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yin Li
- Huan Kui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hao Yi
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Menghui Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Huan Kui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chaofeng Wang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shaokun Du
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wenjuan Zeng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Huan Kui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
8
|
Zhang R, Wang J, Wang Y, Jing W, Lyu B, Wang S, Li J, Yang H, Yu H. Hydrochloric acid-modified high-purity soybean insoluble dietary fiber from Okara attenuates NO 2 - induced hepatic and renal injury in mice. J Food Sci 2025; 90:e70065. [PMID: 40047324 DOI: 10.1111/1750-3841.70065] [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: 10/18/2024] [Revised: 01/09/2025] [Accepted: 01/28/2025] [Indexed: 05/13/2025]
Abstract
Nitrite, a toxic substance, has been shown to adsorb and reduce organismal damage when natural adsorbents are used. Okara, due to its unique structure and adsorption properties, is one of the most eco-friendly and efficient adsorbents, naturally reducing nitrite-induced biotoxicity without side effects. Consequently, six groups of Kunming (KM) mice (n = 10) were comprehensively evaluated through tests for identifying physicochemical properties and physiological indexes. Additionally, the adsorption effects of high-purity soybean insoluble dietary fiber from Okara (HPSIDF) and hydrochloric acid-modified high-purity soybean insoluble dietary fiber from Okara (HCl-HPSIDF) on nitrite-induced toxicity were explored to ultimately determine their bioavailability potentials. Results indicated a 1.35-fold increase in water-holding capacity, a 1.32-fold increase in oil-holding capacity, and a 1.22-fold increase in water-swelling capacity. These physicochemical properties of HCl-HPSIDF were enhanced to improve the dietary fiber quality. Acid treatment increased HCl-HPSIDF's NO2 -removal rate by 20.87% compared to HPSIDF and enhanced its adsorption capacity by 42.5%. The average particle size of HCl-HPSIDF was reduced by 32.82%, increasing NO2 - contact with its adsorption sites and enhancing adsorption capacity. These improvements in changes in HCl-HPSIDF performance support its use as a natural, side-effect-free adsorbent for nitrite, with acid treatment enhancing dietary fiber functionality and enabling HCl-HPSIDF's application in adsorbents.
Collapse
Affiliation(s)
- Ruining Zhang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, China
| | - Jinyu Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, China
| | - Yi Wang
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, China
- Whole Grain Food Engineering Research Center, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Wendan Jing
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, China
| | - Bo Lyu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, China
| | - Sainan Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, China
| | - Jiaxin Li
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, China
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA) - CITEXVI, Universidade de Vigo, Vigo, Spain
| | - Huanhuan Yang
- College of Life Science Chang Chun Normal University, Changchun, China
| | - Hansong Yu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, China
| |
Collapse
|
9
|
An G, Song J, Ying W, Lim W. Overview of the hazardous impacts of metabolism-disrupting chemicals on the progression of fatty liver diseases. Mol Cell Toxicol 2025; 21:387-397. [PMID: 40160987 PMCID: PMC11947047 DOI: 10.1007/s13273-025-00521-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2025] [Indexed: 04/02/2025]
Abstract
Background Given the global increase in obesity, metabolic dysfunction-associated steatotic liver disease (MASLD) is a major health concern. Because the liver is the primary organ for xenobiotic metabolism, the impact of environmental stressors on liver homeostasis and MASLD has garnered significant interest over the past few decades. The concept of metabolism-disrupting chemicals (MDCs) has been introduced to underscore the importance of environmental factors in metabolic homeostasis. Recent epidemiological and biological studies suggest a causal link between exposure to MDCs and prevalence and progression of MASLD. Objective This review aims to introduce the emerging concept of MDCs and their representative toxic mechanisms. In particular, this review focuses on broadening the understanding of their impacts on MASLD or metabolic dysfunction-associated steatohepatitis (MASH) progression. Result Recent research has highlighted the environmental contaminants, such as heavy metals, microplastics, and pesticides, have the potential to influence hepatic metabolism and aggravate MASLD/MASH progression. These MDCs not only directly affect lipid metabolism in hepatocytes but also affect other cell types, such as immune cells and stellate cells, as well as the gut-liver axis. Conclusion Collectively, these findings contribute to establishing a well-defined adverse outcome pathway and identify novel therapeutic options for liver diseases associated with pollutants.
Collapse
Affiliation(s)
- Garam An
- Department of Biological Sciences, Institute of Basic Science, College of Science, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| | - Jisoo Song
- Department of Biological Sciences, Institute of Basic Science, College of Science, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| | - Wei Ying
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093 USA
| | - Whasun Lim
- Department of Biological Sciences, Institute of Basic Science, College of Science, Sungkyunkwan University, Suwon, 16419 Republic of Korea
| |
Collapse
|
10
|
Gan C, Yuan Y, Shen H, Gao J, Kong X, Che Z, Guo Y, Wang H, Dong E, Xiao J. Liver diseases: epidemiology, causes, trends and predictions. Signal Transduct Target Ther 2025; 10:33. [PMID: 39904973 PMCID: PMC11794951 DOI: 10.1038/s41392-024-02072-z] [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: 06/30/2024] [Revised: 10/06/2024] [Accepted: 11/12/2024] [Indexed: 02/06/2025] Open
Abstract
As a highly complex organ with digestive, endocrine, and immune-regulatory functions, the liver is pivotal in maintaining physiological homeostasis through its roles in metabolism, detoxification, and immune response. Various factors including viruses, alcohol, metabolites, toxins, and other pathogenic agents can compromise liver function, leading to acute or chronic injury that may progress to end-stage liver diseases. While sharing common features, liver diseases exhibit distinct pathophysiological, clinical, and therapeutic profiles. Currently, liver diseases contribute to approximately 2 million deaths globally each year, imposing significant economic and social burdens worldwide. However, there is no cure for many kinds of liver diseases, partly due to a lack of thorough understanding of the development of these liver diseases. Therefore, this review provides a comprehensive examination of the epidemiology and characteristics of liver diseases, covering a spectrum from acute and chronic conditions to end-stage manifestations. We also highlight the multifaceted mechanisms underlying the initiation and progression of liver diseases, spanning molecular and cellular levels to organ networks. Additionally, this review offers updates on innovative diagnostic techniques, current treatments, and potential therapeutic targets presently under clinical evaluation. Recent advances in understanding the pathogenesis of liver diseases hold critical implications and translational value for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Can Gan
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Yuan
- Aier Institute of Ophthalmology, Central South University, Changsha, China
| | - Haiyuan Shen
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jinhang Gao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangxin Kong
- Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Zhaodi Che
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yangkun Guo
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
| | - Erdan Dong
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Jia Xiao
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China.
- Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China.
| |
Collapse
|
11
|
Liu B, Liu W, Xu M, Zhao T, Zhou B, Zhou R, Zhu Z, Chen X, Bao Z, Wang K, Li H. Drug delivery systems based on mesoporous silica nanoparticles for the management of hepatic diseases. Acta Pharm Sin B 2025; 15:809-833. [PMID: 40177563 PMCID: PMC11959912 DOI: 10.1016/j.apsb.2024.12.015] [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: 08/14/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 04/05/2025] Open
Abstract
The liver performs multiple life-sustaining functions. Hepatic diseases, including hepatitis, cirrhosis, and hepatoma, pose significant health and economic burdens globally. Along with the advances in nanotechnology, mesoporous silica nanoparticles (MSNs) exhibiting diversiform size and shape, distinct morphological properties, and favorable physico-chemical features have become an ideal choice for drug delivery systems and inspire alternative thinking for the management of hepatic diseases. Initially, we introduce the physiological structure of the liver and highlight its intrinsic cell types and correlative functions. Next, we detail the synthesis methods and physicochemical properties of MSNs and their capacity for controlled drug loading and release. Particularly, we discuss the interactions between liver and MSNs with respect to the passive targeting mechanisms of MSNs within the liver by adjusting their particle size, pore diameter, surface charge, hydrophobicity/hydrophilicity, and surface functionalization. Subsequently, we emphasize the role of MSNs in regulating liver pathophysiology, exploring their value in addressing liver pathological states, such as tumors and inflammation, combined with multi-functional designs and intelligent modes to enhance drug targeting and minimize side effects. Lastly, we put forward the problems, challenges, opportunities, as well as clinical translational issues faced by MSNs in the management of liver diseases.
Collapse
Affiliation(s)
- Boyan Liu
- School of Pharmacy, China Medical University, Shenyang 110122, China
- China Medical University and Queen University of Belfast Joint College, China Medical University, Shenyang 110122, China
| | - Wenshi Liu
- Department of Organ Transplantation and Hepatobiliary, the First Hospital of China Medical University, Shenyang 110001, China
| | - Miao Xu
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Tongyi Zhao
- School of Pharmacy, China Medical University, Shenyang 110122, China
- China Medical University and Queen University of Belfast Joint College, China Medical University, Shenyang 110122, China
| | - Bingxin Zhou
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Ruilin Zhou
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Ze Zhu
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xuchun Chen
- Department of Organ Transplantation and Hepatobiliary, the First Hospital of China Medical University, Shenyang 110001, China
| | - Zhiye Bao
- Department of Organ Transplantation and Hepatobiliary, the First Hospital of China Medical University, Shenyang 110001, China
| | - Keke Wang
- Department of Pharmacy, the First Hospital of China Medical University, Shenyang 110001, China
| | - Heran Li
- School of Pharmacy, China Medical University, Shenyang 110122, China
- China Medical University and Queen University of Belfast Joint College, China Medical University, Shenyang 110122, China
| |
Collapse
|
12
|
Wang S, Huang Z, Nie S, Chen Y, Lei Y, Tu W, Luo M, Zhang ZG, Tian DA, Gong J, Liu M. Unveiling the interplay between hepatocyte SATB1 and innate immunity in autoimmune hepatitis. Int Immunopharmacol 2025; 144:113712. [PMID: 39626541 DOI: 10.1016/j.intimp.2024.113712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Investigating the function of SATB1 in hepatocytes is essential for developing therapeutic strategies for autoimmune hepatitis (AIH). Although SATB1 has been extensively studied in immune cells, its specific activity in hepatocytes within the context of AIH remains unclear. METHODS SATB1 expression in AIH hepatocytes was assessed by qRT-PCR, Western blotting, flow cytometry, and immunohistochemistry. In vivo modulation used RNA interference viruses and overexpression plasmids. SATB1's proinflammatory effects were analyzed with protein microarray, immunohistochemistry, and flow cytometry. Chemotactic effects on RAW264.7 macrophages were tested in vitro, with mechanisms explored by dual-luciferase assays and CUT&RUN qPCR. Liver injury was evaluated by histopathology and serum biochemistry. RESULTS SATB1 was significantly upregulated in hepatocytes of AIH patients and models, showing a stronger increase in hepatocytes than in CD45+ cells, and positively correlated with liver injury severity. In vivo RNAi-mediated SATB1 inhibition reduced liver inflammation, while SATB1 overexpression aggravated AIH progression. Both interference and overexpression experiments confirmed that SATB1 promotes liver injury by facilitating the infiltration of proinflammatory (Ly6Chigh) macrophage. In vitro, supernatant from SATB1-overexpressing hepatocytes enriched chemokine signaling pathways, leading to increased CCL2 expression and release, which attracted macrophages and drove their proinflammatory polarization. Mechanistically, SATB1 promoted CCL2 transcription by binding to its DNA and recruiting p300/CBP. CONCLUSIONS This study reveals that SATB1 is upregulated in hepatocytes in AIH. Elevated SATB1 levels in liver cells contribute to autoimmune hepatitis by increasing CCL2 expression, promoting the recruitment of inflammatory monocyte-derived macrophage, and reshaping the composition of the liver immune microenvironment.
Collapse
Affiliation(s)
- Shuhui Wang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zheng Huang
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Shangshu Nie
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Yu Chen
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Wei Tu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Min Luo
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400000, China
| | - Zhen-Gang Zhang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - De-An Tian
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Jin Gong
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| | - Mei Liu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| |
Collapse
|
13
|
Gibert-Ramos A, Andrés-Rozas M, Pastó R, Alfaro-Retamero P, Guixé-Muntet S, Gracia-Sancho J. Sinusoidal communication in chronic liver disease. Clin Mol Hepatol 2025; 31:32-55. [PMID: 39355871 PMCID: PMC11791556 DOI: 10.3350/cmh.2024.0734] [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: 08/29/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/03/2024] Open
Abstract
The liver sinusoid, mainly composed of liver sinusoidal endothelial cells, hepatic macrophages and hepatic stellate cells, shapes the hepatic vasculature and is key to maintaining liver homeostasis and function. During chronic liver disease (CLD), the function of sinusoidal cells is impaired, being directly involved in the progression of liver fibrosis, cirrhosis, and main clinical complications including portal hypertension and hepatocellular carcinoma. In addition to their roles in liver diseases pathobiology, sinusoidal cells' paracrine communication or cross-talk is being studied as a mechanism of disease but also as a remarkable target for treatment. The aim of this review is to gather current knowledge of intercellular signalling in the hepatic sinusoid during the progression of liver disease. We summarise studies developed in pre-clinical models of CLD, especially emphasizing those pathways characterized in human-based clinically relevant models. Finally, we describe pharmacological treatments targeting sinusoidal communication as promising options to treat CLD and its clinical complications.
Collapse
Affiliation(s)
- Albert Gibert-Ramos
- Liver Vascular Biology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - María Andrés-Rozas
- Liver Vascular Biology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Raül Pastó
- Liver Vascular Biology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Pablo Alfaro-Retamero
- Liver Vascular Biology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Sergi Guixé-Muntet
- Liver Vascular Biology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
14
|
Angendohr C, Missing L, Ehlting C, Wolf SD, Lang KS, Vucur M, Luedde T, Bode JG. Interleukin 1 β suppresses bile acid-induced BSEP expression via a CXCR2-dependent feedback mechanism. PLoS One 2024; 19:e0315243. [PMID: 39680527 DOI: 10.1371/journal.pone.0315243] [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: 09/13/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Inflammation-induced cholestasis is a common problem in septic patients and results from cytokine-mediated inhibition of bile acid export including impaired expression of the bile salt export pump (BSEP) with a consecutive increase in intracellular bile acids mediating cell damage. The present study focuses on the mechanisms by which interleukin 1 β (IL-1β), as a critical mediator of sepsis-induced cholestasis, controls the expression of BSEP in hepatocytes. Notably, the treatment of hepatocytes with IL-1β leads to the upregulation of a broad chemokine pattern. Thereby, the IL-1β -induced expression of in particular the CXCR2 ligands CXCL1 and 2 is further enhanced by bile acids, whereas the FXR-mediated upregulation of BSEP induced by bile acids is inhibited by IL-1β. In this context, it is interesting to note that inhibitor studies indicate that IL-1β mediates its inhibitory effects on bile acid-induced expression of BSEP indirectly via CXCR2 ligands. Consistently, inhibition of CXCR2 with the inhibitor SB225002 significantly attenuated of the inhibitory effect of IL-1β on BSEP expression. These data suggest that part of the cholestasis-inducing effect of IL-1β is mediated via a CXCR2-dependent feedback mechanism.
Collapse
Affiliation(s)
- Carolin Angendohr
- Faculty of Medicine & Düsseldorf University Hospital, Department of Gastroenterology, Hepatology and Infectious Disease, Heinrich-Heine-University, Düsseldorf, Germany
| | - Leah Missing
- Faculty of Medicine & Düsseldorf University Hospital, Department of Gastroenterology, Hepatology and Infectious Disease, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christian Ehlting
- Faculty of Medicine & Düsseldorf University Hospital, Department of Gastroenterology, Hepatology and Infectious Disease, Heinrich-Heine-University, Düsseldorf, Germany
| | - Stephanie D Wolf
- Faculty of Medicine & Düsseldorf University Hospital, Department of Gastroenterology, Hepatology and Infectious Disease, Heinrich-Heine-University, Düsseldorf, Germany
| | - Karl S Lang
- Department of Immunology, University of Essen, Essen, Germany
| | - Mihael Vucur
- Faculty of Medicine & Düsseldorf University Hospital, Department of Gastroenterology, Hepatology and Infectious Disease, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tom Luedde
- Faculty of Medicine & Düsseldorf University Hospital, Department of Gastroenterology, Hepatology and Infectious Disease, Heinrich-Heine-University, Düsseldorf, Germany
| | - Johannes G Bode
- Faculty of Medicine & Düsseldorf University Hospital, Department of Gastroenterology, Hepatology and Infectious Disease, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
15
|
Jang Y, Kim JY, Han SY, Park A, Baek SJ, Lee G, Kang J, Ryu H, Kim SH. Establishment of a chronic biliary disease mouse model with cholecystoduodenal anastomosis for intestinal microbiome preservation. World J Gastroenterol 2024; 30:4937-4946. [PMID: 39679313 PMCID: PMC11612716 DOI: 10.3748/wjg.v30.i46.4937] [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: 09/20/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Chronic biliary disease, including cholangitis and cholecystitis, is attributed to ascending infection by intestinal bacteria. Development of a mouse model for bile duct inflammation is imperative for the advancement of novel therapeutic approaches. Current models fail to replicate the harmful bacterial influx to the biliary tract observed in humans and spread of inflammation to the liver. Therefore, we aimed to establish an animal model of biliary disease that faithfully replicates the mechanisms of human diseases. AIM To establish a cholecystoduodenal anastomosis model capable of mimicking the mechanisms of ascending infection and inflammation observed in human biliary diseases. METHODS We established a mouse biliary disease model by directly connecting the gallbladder and duodenum, enabling ascending infection into the biliary tract without traversing the sphincter of Oddi. RESULTS In the cholecystoduodenal anastomosis mouse model, we observed impaired epithelial structure, wall thickening, and macrophage recruitment in the gallbladder. Despite the absence of postoperative antibiotics, we detected no changes in serum proinflammatory cytokine levels, indicating no systemic inflammation. Moreover, patency between the gallbladder and duodenum was confirmed via common bile duct ligation. Injection of patient-derived pathogenic bacteria into bile duct-ligated mice led to ascending infection, which significantly increased proinflammatory cytokine mRNA expression in the liver, duodenum, and ileum. These results indicate that our mouse model exhibited a direct connection between the gallbladder and duodenum, leading to ascending infection and closely mimicking the clinical features of biliary diseases observed in humans. CONCLUSION The cholecystoduodenal anastomosis mouse model is an effective chronic biliary disease model with significant relevance in the development of microbiome-based therapies for the prevention and treatment of biliary disease.
Collapse
Affiliation(s)
- Yunseon Jang
- Translational Immunology Institute, Chungnam National University School of Medicine, Daejeon 35015, South Korea
| | - Jung Yeon Kim
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, South Korea
| | - Song Yeon Han
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, South Korea
| | - Arum Park
- AtoGen Co., Ltd., Techno 1-ro, Daejeon 35015, South Korea
| | - So Jeong Baek
- AtoGen Co., Ltd., Techno 1-ro, Daejeon 35015, South Korea
| | - Gyurim Lee
- AtoGen Co., Ltd., Techno 1-ro, Daejeon 35015, South Korea
| | - Jihee Kang
- AtoGen Co., Ltd., Techno 1-ro, Daejeon 35015, South Korea
| | - Hyewon Ryu
- Translational Immunology Institute, Chungnam National University School of Medicine, Daejeon 35015, South Korea
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon 35015, South Korea
| | - Seok-Hwan Kim
- Department of Surgery, Chungnam National University Hospital, Daejeon 35015, South Korea
- Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon 35015, South Korea
| |
Collapse
|
16
|
Zhang G, Wu K, Jiang X, Gao Y, Ding D, Wang H, Yu C, Wang X, Jia N, Zhu L. The role of ferroptosis-related non-coding RNA in liver fibrosis. Front Cell Dev Biol 2024; 12:1517401. [PMID: 39717848 PMCID: PMC11663870 DOI: 10.3389/fcell.2024.1517401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024] Open
Abstract
Liver fibrosis represents a reversible pathophysiological process, caused by chronic inflammation stemming from hepatocyte damage. It delineates the initial stage in the progression of chronic liver disease. This pathological progression is characterized by the excessive accumulation of the extracellular matrix (ECM), which leads to significant structural disruption and ultimately impairs liver function. To date, no specific antifibrotic drugs have been developed, and advanced liver fibrosis remains largely incurable. Liver transplantation remains the sole efficacious intervention for advanced liver fibrosis; nevertheless, it is constrained by exorbitant costs and the risk of postoperative immune rejection, underscoring the imperative for novel therapeutic strategies. Ferroptosis, an emergent form of regulated cell death, has been identified as a pivotal regulatory mechanism in the development of liver fibrosis and is intricately linked with the progression of liver diseases. Recent investigations have elucidated that a diverse array of non-coding RNAs (ncRNAs), including microRNAs, long non-coding RNAs, and circular RNAs, are involved in the ferroptosis pathway, thereby modulating the progression of various diseases, including liver fibrosis. In recent years, the roles of ferroptosis and ferroptosis-related ncRNAs in liver fibrosis have attracted escalating scholarly attention. This paper elucidates the pathophysiology of liver fibrosis, explores the mechanisms underlying ferroptosis, and delineates the involvement of ncRNA-mediated ferroptosis pathways in the pathology of liver fibrosis. It aims to propose novel strategies for the prevention and therapeutic intervention of liver fibrosis.
Collapse
Affiliation(s)
- Guozhu Zhang
- Department of Emergency Medicine, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Kejia Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaobo Jiang
- Kunshan Zhenchuan Community Health Service Center, Kunshan, Jiangsu, China
| | - Yuan Gao
- Department of Hepato-Biliary-Pancreatic Surgery, The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Dong Ding
- Department of Hepato-Biliary-Pancreatic Surgery, The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Hao Wang
- Department of Emergency Medicine, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Chongyuan Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiaozhong Wang
- Department of General Surgery, Wujin Affiliated Hospital of Jiangsu University and the Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Naixin Jia
- Department of Hepatobiliary Surgery, Kunshan First People’s Hospital affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Li Zhu
- Department of Emergency Medicine, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
17
|
Mahajan R, Tyagi AK. Pharmacogenomic insights into tuberculosis treatment shows the NAT2 genetic variants linked to hepatotoxicity risk: a systematic review and meta-analysis. BMC Genom Data 2024; 25:103. [PMID: 39639188 PMCID: PMC11622454 DOI: 10.1186/s12863-024-01286-y] [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: 09/03/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Tuberculosis (TB) patients undergoing anti-tuberculosis treatment often face serious adverse drug reactions, such as hepatotoxicity. Genetic variants of the N-acetyltransferase 2 (NAT2) gene have been linked to an increased risk of these toxic events. OBJECTIVE This study aims to provide a comprehensive evaluation of the evidence linking NAT2 genetic variants to anti-tuberculosis drug-related hepatotoxicity (ATDH). METHOD A comprehensive review and meta-analysis was performed by accessing databases such as PubMed, Scopus, and Web of Science. A total of 24 articles were incorporated into the dataset. Meta-analyses were conducted to gather estimates of the association between the slow acetlylators (SA) genotype and ATDH. The studies were stratified by ethnicity, regimen, genotyping methods, criteria for liver toxicity, and dosage. Also, meta-analysis for the specific SA type that was most likely responsible for the ATDH was also conducted. RESULTS The included studies showed individuals with a slow NAT2 acetylator had a significantly greater risk of experiencing hepatotoxicity ATDH (odds ratio [OR] 2.52 (95% CI: 1.95-3.27; p value < 0.001) compared to individuals with other types of acetylator (i.e., rapid and immediate). Among individuals with slow acetylator NAT2*5/7, NAT2*5/6, and NAT2*6/6 genotypes, there is a greater likelihood of association compared to other variations. CONCLUSION Our meta-analysis confirms a significant association between slow NAT2 acetylator and increased hepatotoxicity risk. The findings from the present underscore the potential of pharmacogenomic testing to improve TB treatment outcomes. By identifying patients with the slow acetylator NAT2 genotype, healthcare providers can predict an increased risk of anti-tuberculosis drug-induced hepatotoxicity. This allows for personalized treatment strategies, such as adjusting drug dosages or selecting alternative therapies, to minimize adverse effects and optimize efficacy.
Collapse
Affiliation(s)
- Rashmi Mahajan
- Dr. Bhimrao Ramji Ambedkar Government Medical College, Kannauj, India
| | - Anuj Kumar Tyagi
- Dr. Bhimrao Ramji Ambedkar Government Medical College, Kannauj, India.
| |
Collapse
|
18
|
Zhang X, He B, Lu J, Bao Q, Wang J, Yang Y. The crucial roles and research advances of cGAS‑STING pathway in liver diseases. Ann Med 2024; 56:2394588. [PMID: 39183465 PMCID: PMC11348815 DOI: 10.1080/07853890.2024.2394588] [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: 06/04/2024] [Revised: 07/17/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024] Open
Abstract
Inflammation responses have identified as a key mediator of in various liver diseases with high morbidity and mortality. cGAS-STING signalling is essential in innate immunity since it triggers release of type I interferons and various of proinflammatory cytokines. The potential connection between cGAS-STING pathway and liver inflammatory diseases has recently been reported widely. In our review, the impact of cGAS-STING on liver inflammation and regulatory mechanism are summarized. Furthermore, many inhibitors of cGAS-STING signalling as promising agents to cure liver inflammation are also explored in detail. A comprehensive knowledge of molecular mechanisms of cGAS-STING signalling in liver inflammation is vital for exploring novel treatments and providing recommendations and perspectives for future utilization.
Collapse
Affiliation(s)
- Xiaoqian Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin He
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiongling Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yida Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
19
|
Besné-Eseverri I, Martín MÁ, Lobo G, Cano MP, Portillo MP, Trepiana J. Antioxidant and Anti-Inflammatory Effects of Opuntia Extracts on a Model of Diet-Induced Steatosis. Antioxidants (Basel) 2024; 13:1416. [PMID: 39594557 PMCID: PMC11591152 DOI: 10.3390/antiox13111416] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Oxidative stress and inflammation are widely recognised as factors that can initiate and facilitate the development of MAFLD. The aim of this study is to analyse the effect of low and high doses of Opuntia stricta var. dillenii peel extract (L-OD and H-OD, respectively) and Opuntia ficus-indica var. colorada pulp extract (L-OFI and H-OFI, respectively), which are rich in betalains and phenolic compounds, on oxidative stress, inflammation, DNA damage and apoptosis in rat livers with diet-induced steatosis. Steatotic diet led to increased final body and liver weight, serum transaminases, hepatic TG content, oxidative status and cell death. H-OFI treatment decreased serum AST levels, while L-OFI reduced hepatic TG accumulation. Oxidative stress was partially prevented with H-OD and H-OFI supplementation, and pro-inflammatory cytokines levels were especially improved with H-OFI treatment. Moreover, H-OFI appears to prevent DNA damage markers. Finally, H-OD and L-OFI supplementation down-regulated the apoptotic pathway. In conclusion, both H-OD and H-OFI supplementation were effective in regulating the progression to metabolic steatohepatitis, triggering different mechanisms of action.
Collapse
Affiliation(s)
- Irene Besné-Eseverri
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Centre, 01006 Vitoria-Gasteiz, Spain; (I.B.-E.); (M.P.P.)
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 28029 Madrid, Spain
| | - María Ángeles Martín
- Science and Food Technology and Nutrition Institute (ICTAN-CSIC), 28040 Madrid, Spain;
- CIBER Diabetes and Related Metabolic Diseases (CIBERdem), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Gloria Lobo
- Department of Crop Production in Tropical and Subtropical Areas, Instituto Canario de Investigaciones Agrarias (ICIA), 38297 Tenerife, Spain;
| | - M. Pilar Cano
- Laboratory of Phytochemistry and Plant Food Functionality, Biotechnology and Food Microbiology Department, Institute of Food Science Research (CIAL) (CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain;
| | - María P. Portillo
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Centre, 01006 Vitoria-Gasteiz, Spain; (I.B.-E.); (M.P.P.)
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 28029 Madrid, Spain
- BIOARABA Institute of Health, 01009 Vitoria-Gasteiz, Spain
| | - Jenifer Trepiana
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Centre, 01006 Vitoria-Gasteiz, Spain; (I.B.-E.); (M.P.P.)
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 28029 Madrid, Spain
- BIOARABA Institute of Health, 01009 Vitoria-Gasteiz, Spain
| |
Collapse
|
20
|
Patidar P, Hirani N, Bharti S, Baig MS. Key regulators of hepatic stellate cell activation in alcohol liver Disease: A comprehensive review. Int Immunopharmacol 2024; 141:112938. [PMID: 39163683 DOI: 10.1016/j.intimp.2024.112938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/27/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024]
Abstract
Alcoholic liver disease (ALD) is a broad category of disorders that begin with liver injury, lead to liver fibrosis, and ultimately conclude in alcohol-induced liver cirrhosis, the most chronic and irreversible liver damage. Liver fibrosis (LF) is a common pathological characteristic observed in most chronic liver inflammatory conditions that involve prolonged inflammation. In this review, we have summarized ethanol-mediated hepatic stellate cell (HSCs) activation and its role in liver fibrosis progression. We highlight important molecular mechanisms that are modulated by ethanol, play a role in the activation of HSCs and the progression of liver fibrosis and identifying potential targets to ameliorate liver fibrosis.
Collapse
Affiliation(s)
- Pramod Patidar
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Nik Hirani
- MRC Centre for Inflammation Research, Queen'sMedicalResearch Institute, University of Edinburgh, Edinburgh, EH164TJ, UK
| | - Shreya Bharti
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India.
| |
Collapse
|
21
|
Bu HF, Subramanian S, Chou PM, Liu F, Sun L, Geng H, Wang X, Liao J, Du C, Hu J, Tan SC, Nathan N, Yang GY, Tan XD. A novel mouse model of hepatocyte-specific apoptosis-induced myeloid cell-dominant sterile liver injury and repair response. Am J Physiol Gastrointest Liver Physiol 2024; 327:G499-G512. [PMID: 39104322 PMCID: PMC11482258 DOI: 10.1152/ajpgi.00005.2024] [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: 01/10/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 08/07/2024]
Abstract
Apoptosis, inflammation, and wound healing are critical pathophysiological events associated with various liver diseases. Currently, there is a lack of in vivo approaches to study hepatocyte apoptosis-induced liver injury and repair. To address this critical knowledge gap, we developed a unique genetically modified mouse model, namely, 3-Transgene (Tg) with inducible Hepatocyte-Specific Apoptosis Phenotype (3xTg-iHAP) in this study. The 3xTg-iHAP mice possess three transgenes including Alb-Cre, Rosa26-rtTA, and tetO-Fasl on a B6 background. These mice are phenotypically normal, viable, and fertile. After subcutaneous administration of a single dose of doxycycline (5 mg/kg, Dox) to 3xTg-iHAP mice, we observed a complete histological spectrum of sterile liver wound-healing responses: asymptomatic hepatocyte apoptosis at 8 h, necrotic liver injury and sterile inflammation at 48 h, followed by hepatocyte mitosis and regeneration within 7 days. During the injury phase, the mice exhibited an increase in the biomarkers of alanine aminotransferase (ALT), chemokine (C-X-C motif) ligand 1 (CXCL1), and IL-6 in peripheral blood, as well as α-smooth muscle actin (α-SMA) protein in liver tissues. Conversely, the mice displayed a decrease in these markers in the recovery phase. Remarkably, this model shows that the sterile liver injury following elevated hepatocyte apoptosis is associated with an increase in myeloid cells in the liver. Within 7 days post-Dox administration, the liver of Dox-treated 3xTg-iHAP mice displays a normal histological structure, indicating the completion of wound healing. Together, we established a novel mouse model of injury and regeneration induced by hepatocyte apoptosis. This tool provides a robust in vivo platform for studying the pathophysiology of sterile liver inflammation, regeneration, and new therapeutic interventions for liver diseases.NEW & NOTEWORTHY Bu et al. present a triple-transgenic mouse model, namely, 3xTg-iHAP mice that are engineered to explore hepatocyte apoptosis-triggered sterile liver injury and regeneration. This model demonstrates a full spectrum of liver wound-healing responses from asymptomatic apoptosis to injury, myeloid cell-dominant sterile inflammation, and repair after induction of hepatocyte-specific apoptosis. The robust nature of this model makes it an invaluable in vivo tool for studying sterile liver inflammation, regeneration, and new therapeutic strategies.
Collapse
Affiliation(s)
- Heng-Fu Bu
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| | - Saravanan Subramanian
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| | - Pauline M Chou
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Fangyi Liu
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Leyu Sun
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Hua Geng
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| | - Xiao Wang
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| | - Jie Liao
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Chao Du
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Joyce Hu
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Stephanie C Tan
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Nirmal Nathan
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Guang-Yu Yang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Xiao-Di Tan
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| |
Collapse
|
22
|
Haroun E, Lim SH, Dutta D. GBT1118, a Voxelotor Analog, Ameliorates Hepatopathy in Sickle Cell Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1581. [PMID: 39459368 PMCID: PMC11509622 DOI: 10.3390/medicina60101581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: In sickle cell disease (SCD), hepatopathy is a cumulative consequence of ischemia/reperfusion (I/R) injury from a vaso-occlusive crisis, tissue inflammation, and iron overload due to blood transfusion. Hepatopathy is a major contributing factor of shortened life span in SCD patients. We hypothesized that the voxelotor, a hemoglobin allosteric modifier, ameliorates sickle hepatopathy. Materials and Methods: Townes SCD mice and their controls were treated with either chow containing GBT1118, a voxelotor analog, or normal chow. We evaluated inflammation, fibrosis, apoptosis and ferroptosis in their livers using qPCR, ELISA, histology, and immunohistochemistry. Results: GBT1118 treatment resulted in reduced hemolysis, iron overload and inflammation in the liver of SCD mice. There were significant reductions in the liver enzyme levels and bile acids. Furthermore, GBT1118-treated mice exhibited reduced apoptosis, necrosis, and fibrosis. Increased ferroptosis as evident from elevated 4-hydroxynonenal (4-HNE) staining, malondialdehyde (MDA) levels, and expression of Ptgs2 and Slc7a11 mRNAs, were also significantly reduced after GBT1118 treatment. To explain the increased ferroptosis, we evaluated iron homeostasis markers in livers. SCD mice showed decreased expression of heme oxygenase-1, ferritin, hepcidin, and ferroportin mRNA levels. GBT1118 treatment significantly increased expressions of these genes. Conclusions: Our results suggest GBT1118 treatment in SCD confers the amelioration of sickle hepatopathy by reducing inflammation, fibrosis, apoptosis, iron overload and ferroptosis.
Collapse
Affiliation(s)
| | | | - Dibyendu Dutta
- Division of Hematology and Oncology, Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY 13210, USA; (E.H.); (S.H.L.)
| |
Collapse
|
23
|
Talubo NDD, Tsai PW, Tayo LL. Comprehensive RNA-Seq Gene Co-Expression Analysis Reveals Consistent Molecular Pathways in Hepatocellular Carcinoma across Diverse Risk Factors. BIOLOGY 2024; 13:765. [PMID: 39452074 PMCID: PMC11505157 DOI: 10.3390/biology13100765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/04/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024]
Abstract
Hepatocellular carcinoma (HCC) has the highest mortality rate and is the most frequent of liver cancers. The heterogeneity of HCC in its etiology and molecular expression increases the difficulty in identifying possible treatments. To elucidate the molecular mechanisms of HCC across grades, data from The Cancer Genome Atlas (TCGA) were used for gene co-expression analysis, categorizing each sample into its pre-existing risk factors. The R library BioNERO was used for preprocessing and gene co-expression network construction. For those modules most correlated with a grade, functional enrichments from different databases were then tested, which appeared to have relatively consistent patterns when grouped by G1/G2 and G3/G4. G1/G2 exhibited the involvement of pathways related to metabolism and the PI3K/Akt pathway, which regulates cell proliferation and related pathways, whereas G3/G4 showed the activation of cell adhesion genes and the p53 signaling pathway, which regulates apoptosis, cell cycle arrest, and similar processes. Module preservation analysis was then used with the no history dataset as the reference network, which found cell adhesion molecules and cell cycle genes to be preserved across all risk factors, suggesting they are imperative in the development of HCC regardless of potential etiology. Through hierarchical clustering, modules related to the cell cycle, cell adhesion, the immune system, and the ribosome were found to be consistently present across all risk factors, with distinct clusters linked to oxidative phosphorylation in viral HCC and pentose and glucuronate interconversions in non-viral HCC, underscoring their potential roles in cancer progression.
Collapse
Affiliation(s)
- Nicholas Dale D. Talubo
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila 1002, Philippines;
- School of Graduate Studies, Mapúa University, Manila 1002, Philippines
| | - Po-Wei Tsai
- Department of Food Science, National Taiwan Ocean University, Keelung 202, Taiwan;
| | - Lemmuel L. Tayo
- Department of Biology, School of Health Sciences, Mapúa University, Makati 1203, Philippines
| |
Collapse
|
24
|
Choi D, Gwon K, de Hoyos-Vega JM, Lee S, Nguyen KM, Gonzalez-Suarez AM, Stybayeva G, Revzin A. An Ultrathin Coating of Microcapsules Enhances the Function of Encapsulated Hepatocyte Spheroids. ACS APPLIED MATERIALS & INTERFACES 2024; 16:51411-51420. [PMID: 39269915 DOI: 10.1021/acsami.4c08329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Maintaining the differentiated phenotype and function of primary hepatocytes in vitro and in vivo represents a distinct challenge. Our paper describes microcapsules comprised of a bioactive polymer and overcoated with an ultrathin film as a means of maintaining the function of entrapped hepatocytes for at least two weeks. We previously demonstrated that heparin (Hep)-based microcapsules improved the function of entrapped primary hepatocytes by capturing and releasing cell-secreted inductive signals, including hepatocyte growth factor (HGF). Further enhancement of hepatic function could be gained by loading exogenous HGF into microcapsules. In this study, we demonstrate that an ultrathin coating of tannic acid (TA) further enhances endogenous HGF signaling for entrapped hepatocytes and increases by 2-fold the rate of uptake of exogenous HGF by Hep microcapsules. Hepatocytes in overcoated microcapsules exhibited better function and hepatic gene expression than in capsules without a TA coating. Our study showcases the potential application of ultrathin coatings to modulate the bioactivity of microcapsules and may enable the use of encapsulated hepatocytes for modeling drug toxicity or treating liver diseases.
Collapse
Affiliation(s)
- Daheui Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Kihak Gwon
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department of Biofibers and Biomaterials Science, Kyungpook National University, Daegu 41566, Republic of Korea
| | - José M de Hoyos-Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Seonhwa Lee
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Kianna M Nguyen
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Alan M Gonzalez-Suarez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| |
Collapse
|
25
|
Raz M, Milo T, Glass DS, Mayo A, Alon U. Endocrine gland size is proportional to its target tissue size. iScience 2024; 27:110625. [PMID: 39224518 PMCID: PMC11367476 DOI: 10.1016/j.isci.2024.110625] [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: 10/25/2023] [Revised: 06/26/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Endocrine glands secrete hormones into the circulation to target distant tissues and regulate their functions. The qualitative relationship between hormone-secreting organs and their target tissues is well established, but a quantitative approach is currently limited. Quantification is important, as it could allow us to study the endocrine system using engineering concepts of optimality and tradeoffs. In this study, we collected literature data on 24 human hormones secreted from dedicated endocrine cells. We find that the number of endocrine cells secreting a hormone is proportional to the number of its target cells. A single endocrine cell serves approximately 2,000 target cells, a relationship that spans 6 orders of magnitude of cell numbers. This suggests an economic principle of cells working near their maximal capacity, and glands that are no bigger than they need to be.
Collapse
Affiliation(s)
- Moriya Raz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tomer Milo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - David S. Glass
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
26
|
Faienza MF, Farella I, Khalil M, Portincasa P. Converging Pathways between Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) and Diabetes in Children. Int J Mol Sci 2024; 25:9924. [PMID: 39337412 PMCID: PMC11432101 DOI: 10.3390/ijms25189924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
In the past thirty years, childhood obesity rates have risen significantly worldwide, affecting over 340 million children in affluent nations. This surge is intricately tied to metabolic disorders, notably insulin resistance, type 2 diabetes mellitus (T2DM), and the continually evolving spectrum of metabolic-associated (dysfunction) steatotic liver disease (MASLD). This review underscores the alarming escalation of childhood obesity and delves comprehensively into the evolving and dynamic changes of nomenclature surrounding diverse conditions of hepatic steatosis, from the initial recognition of non-alcoholic fatty liver disease (NAFLD) to the progressive evolution into MASLD. Moreover, it emphasizes the crucial role of pediatric endocrinologists in thoroughly and accurately investigating MASLD onset in children with T2DM, where each condition influences and exacerbates the progression of the other. This review critically highlights the inadequacies of current screening strategies and diagnosis, stressing the need for a paradigm shift. A proposed solution involves the integration of hepatic magnetic resonance imaging assessment into the diagnostic arsenal for children showing insufficient glycemic control and weight loss post-T2DM diagnosis, thereby complementing conventional liver enzyme testing. This holistic approach aims to significantly enhance diagnostic precision, fostering improved outcomes in this vulnerable high-risk pediatric population.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Ilaria Farella
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (I.F.); (M.K.)
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (I.F.); (M.K.)
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy; (I.F.); (M.K.)
| |
Collapse
|
27
|
Rodrigues A, Alexandre-Pires G, Valério-Bolas A, Nunes T, Pereira da Fonseca I, Santos-Gomes G. Kupffer Cells and Hepatocytes: A Key Relation in the Context of Canine Leishmaniasis. Microorganisms 2024; 12:1887. [PMID: 39338560 PMCID: PMC11433711 DOI: 10.3390/microorganisms12091887] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/29/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
Human zoonotic visceral leishmaniasis (ZVL) and canine leishmaniasis (CanL) constitute a major public and veterinary health concern and are both caused by the infection with the protozoan parasite Leishmania infantum. One of the main target organs in CanL is the liver. This complex organ, composed of various highly specialized cell types, has garnered significant attention from the scientific community as a crucial player in innate immune functions. In the context of CanL, liver infection by parasites and the host immune response generated strongly influence the disease outcome. Thus, taking advantage of a co-culture system involving canine hepatocytes and L. infantum-infected autologous Kupffer cells (KCs), allowing cell-to-cell interaction, the current report aims to shed light on the hepatocyte-KCs immune interaction. The co-culture of infected KCs with hepatocytes revealed a vital role of these cells in the activation of a local immune response against L. infantum parasites. Although KCs alone can be immunologically silenced by L. infantum infection, the cell-to-cell interaction with hepatocytes in co-culture can lead to local immune activation. In co-culture it was observed gene expression increased the number of innate immune receptors, specifically cell membrane TLR2 and cytoplasmatic NOD1 along with high TNF-α generation. Altogether, these results suggest that the immune response generated in co-culture could induce the recruitment of other circulating cells to contain and contribute to the resolution of the infection in the liver. This work also enhances our understanding of the liver as a vital organ in innate immunity within the context of CanL.
Collapse
Affiliation(s)
- Armanda Rodrigues
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), 1349-008 Lisbon, Portugal
| | - Graça Alexandre-Pires
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1200-771 Lisbon, Portugal
| | - Ana Valério-Bolas
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), 1349-008 Lisbon, Portugal
| | - Telmo Nunes
- Microscopy Center, Faculty of Sciences, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Isabel Pereira da Fonseca
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1200-771 Lisbon, Portugal
| | - Gabriela Santos-Gomes
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), 1349-008 Lisbon, Portugal
| |
Collapse
|
28
|
Frojdenfal S, Zuchowska A. Advanced Liver-on-a-Chip Model for Evaluating Drug Metabolism and Hepatotoxicity. BIOSENSORS 2024; 14:435. [PMID: 39329810 PMCID: PMC11430604 DOI: 10.3390/bios14090435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024]
Abstract
The liver has many important functions, including the biotransformation of drugs and detoxification of the human organism. As such, it is also exposed to many harmful substances, which leads to disorders and diseases such as cirrhosis. For these reasons, it seems important to consider liver metabolism and the direct effects on the liver when evaluating the efficacy of new drugs. Accordingly, we have developed an advanced in vitro liver model using an organ-on-a-chip approach that replicates many of the morphological and functional features of the liver in vivo. The model we created can metabolize drugs, which we demonstrated using two widely used anticancer drugs, 5-fluorouracil (5FU) and capecitabine (CAP). In addition, to the best of our knowledge, we are the first who evaluate the direct effects of these drugs not only on the viability of liver model-building cells but on their functions, such as cytochrome P450 activity and albumin production. Our study brings new hope to properly evaluating drug efficacy at the in vitro level.
Collapse
Affiliation(s)
| | - Agnieszka Zuchowska
- Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warszawa, Poland;
| |
Collapse
|
29
|
Mata-Martínez E, Ramírez-Ledesma MG, Vázquez-Victorio G, Hernández-Muñoz R, Díaz-Muñoz M, Vázquez-Cuevas FG. Purinergic Signaling in Non-Parenchymal Liver Cells. Int J Mol Sci 2024; 25:9447. [PMID: 39273394 PMCID: PMC11394727 DOI: 10.3390/ijms25179447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Purinergic signaling has emerged as an important paracrine-autocrine intercellular system that regulates physiological and pathological processes in practically all organs of the body. Although this system has been thoroughly defined since the nineties, recent research has made substantial advances regarding its role in aspects of liver physiology. However, most studies have mainly targeted the entire organ, 70% of which is made up of parenchymal cells or hepatocytes. Because of its physiological role, the liver is exposed to toxic metabolites, such as xenobiotics, drugs, and fatty acids, as well as to pathogens such as viruses and bacteria. Under injury conditions, all cell types within the liver undergo adaptive changes. In this context, the concentration of extracellular ATP has the potential to increase dramatically. Indeed, this purinergic response has not been studied in sufficient detail in non-parenchymal liver cells. In the present review, we systematize the physiopathological adaptations related to the purinergic system in chronic liver diseases of non-parenchymal liver cells, such as hepatic stellate cells, Kupffer cells, sinusoidal endothelial cells, and cholangiocytes. The role played by non-parenchymal liver cells in these circumstances will undoubtedly be strategic in understanding the regenerative activities that support the viability of this organ under stressful conditions.
Collapse
Affiliation(s)
- Esperanza Mata-Martínez
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, Mexico City 04510, Mexico
| | - María Guadalupe Ramírez-Ledesma
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla #3001, Querétaro 76230, Mexico
| | - Genaro Vázquez-Victorio
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de México (UNAM), Circuito Exterior S/N, Ciudad Universitaria, Mexico City 04510, Mexico
| | - Rolando Hernández-Muñoz
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, Mexico City 04510, Mexico
| | - Mauricio Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla #3001, Querétaro 76230, Mexico
| | - Francisco G Vázquez-Cuevas
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla #3001, Querétaro 76230, Mexico
| |
Collapse
|
30
|
Geervliet E, Karkdijk E, Bansal R. Inhibition of intrahepatic monocyte recruitment by Cenicriviroc and extracellular matrix degradation by MMP1 synergistically attenuate liver inflammation and fibrogenesis in vivo. Sci Rep 2024; 14:16897. [PMID: 39043893 PMCID: PMC11266417 DOI: 10.1038/s41598-024-67926-6] [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: 06/06/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
The chemokine (CCL)-chemokine receptor (CCR2) interaction, importantly CCL2-CCR2, involved in the intrahepatic recruitment of monocytes upon liver injury promotes liver fibrosis. CCL2-CCR2 antagonism using Cenicriviroc (CVC) showed promising results in several preclinical studies. Unfortunately, CVC failed in phase III clinical trials due to lack of efficacy to treat liver fibrosis. Lack of efficacy could be attributed to the fact that macrophages are also involved in disease resolution by secreting matrix metalloproteinases (MMPs) to degrade extracellular matrix (ECM), thereby inhibiting hepatic stellate cells (HSCs) activation. HSCs are the key pathogenic cell types in liver fibrosis that secrete excessive amounts of ECM causing liver stiffening and liver dysfunction. Knowing the detrimental role of intrahepatic monocyte recruitment, ECM, and HSCs activation during liver injury, we hypothesize that combining CVC and MMP (MMP1) could reverse liver fibrosis. We evaluated the effects of CVC, MMP1 and CVC + MMP1 in vitro and in vivo in CCl4-induced liver injury mouse model. We observed that CVC + MMP1 inhibited macrophage migration, and TGF-β induced collagen-I expression in fibroblasts in vitro. In vivo, MMP1 + CVC significantly inhibited normalized liver weights, and improved liver function without any adverse effects. Moreover, MMP1 + CVC inhibited monocyte infiltration and liver inflammation as confirmed by F4/80 and CD11b staining, and TNFα gene expression. MMP1 + CVC also ameliorated liver fibrogenesis via inhibiting HSCs activation as assessed by collagen-I staining and collagen-I and α-SMA mRNA expression. In conclusion, we demonstrated that a combination therapeutic approach by combining CVC and MMP1 to inhibit intrahepatic monocyte recruitment and increasing collagen degradation respectively ameliorate liver inflammation and fibrosis.
Collapse
Affiliation(s)
- Eline Geervliet
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Carre 4419, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Esmee Karkdijk
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Carre 4419, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Ruchi Bansal
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Carre 4419, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands.
| |
Collapse
|
31
|
Akkız H, Gieseler RK, Canbay A. Liver Fibrosis: From Basic Science towards Clinical Progress, Focusing on the Central Role of Hepatic Stellate Cells. Int J Mol Sci 2024; 25:7873. [PMID: 39063116 PMCID: PMC11277292 DOI: 10.3390/ijms25147873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
The burden of chronic liver disease is globally increasing at an alarming rate. Chronic liver injury leads to liver inflammation and fibrosis (LF) as critical determinants of long-term outcomes such as cirrhosis, liver cancer, and mortality. LF is a wound-healing process characterized by excessive deposition of extracellular matrix (ECM) proteins due to the activation of hepatic stellate cells (HSCs). In the healthy liver, quiescent HSCs metabolize and store retinoids. Upon fibrogenic activation, quiescent HSCs transdifferentiate into myofibroblasts; lose their vitamin A; upregulate α-smooth muscle actin; and produce proinflammatory soluble mediators, collagens, and inhibitors of ECM degradation. Activated HSCs are the main effector cells during hepatic fibrogenesis. In addition, the accumulation and activation of profibrogenic macrophages in response to hepatocyte death play a critical role in the initiation of HSC activation and survival. The main source of myofibroblasts is resident HSCs. Activated HSCs migrate to the site of active fibrogenesis to initiate the formation of a fibrous scar. Single-cell technologies revealed that quiescent HSCs are highly homogenous, while activated HSCs/myofibroblasts are much more heterogeneous. The complex process of inflammation results from the response of various hepatic cells to hepatocellular death and inflammatory signals related to intrahepatic injury pathways or extrahepatic mediators. Inflammatory processes modulate fibrogenesis by activating HSCs and, in turn, drive immune mechanisms via cytokines and chemokines. Increasing evidence also suggests that cellular stress responses contribute to fibrogenesis. Recent data demonstrated that LF can revert even at advanced stages of cirrhosis if the underlying cause is eliminated, which inhibits the inflammatory and profibrogenic cells. However, despite numerous clinical studies on plausible drug candidates, an approved antifibrotic therapy still remains elusive. This state-of-the-art review presents cellular and molecular mechanisms involved in hepatic fibrogenesis and its resolution, as well as comprehensively discusses the drivers linking liver injury to chronic liver inflammation and LF.
Collapse
Affiliation(s)
- Hikmet Akkız
- Department of Gastroenterology and Hepatology, University of Bahçeşehir, Beşiktaş, Istanbul 34353, Turkey
| | - Robert K. Gieseler
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr University Bochum, In der Schornau 23–25, 44892 Bochum, Germany; (R.K.G.); (A.C.)
| | - Ali Canbay
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr University Bochum, In der Schornau 23–25, 44892 Bochum, Germany; (R.K.G.); (A.C.)
| |
Collapse
|
32
|
Gondal MN, Shah SUR, Chinnaiyan AM, Cieslik M. A systematic overview of single-cell transcriptomics databases, their use cases, and limitations. FRONTIERS IN BIOINFORMATICS 2024; 4:1417428. [PMID: 39040140 PMCID: PMC11260681 DOI: 10.3389/fbinf.2024.1417428] [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: 04/14/2024] [Accepted: 06/11/2024] [Indexed: 07/24/2024] Open
Abstract
Rapid advancements in high-throughput single-cell RNA-seq (scRNA-seq) technologies and experimental protocols have led to the generation of vast amounts of transcriptomic data that populates several online databases and repositories. Here, we systematically examined large-scale scRNA-seq databases, categorizing them based on their scope and purpose such as general, tissue-specific databases, disease-specific databases, cancer-focused databases, and cell type-focused databases. Next, we discuss the technical and methodological challenges associated with curating large-scale scRNA-seq databases, along with current computational solutions. We argue that understanding scRNA-seq databases, including their limitations and assumptions, is crucial for effectively utilizing this data to make robust discoveries and identify novel biological insights. Such platforms can help bridge the gap between computational and wet lab scientists through user-friendly web-based interfaces needed for democratizing access to single-cell data. These platforms would facilitate interdisciplinary research, enabling researchers from various disciplines to collaborate effectively. This review underscores the importance of leveraging computational approaches to unravel the complexities of single-cell data and offers a promising direction for future research in the field.
Collapse
Affiliation(s)
- Mahnoor N. Gondal
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Saad Ur Rehman Shah
- Gies College of Business, University of Illinois Business College, Champaign, MI, United States
| | - Arul M. Chinnaiyan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, United States
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
- Department of Urology, University of Michigan, Ann Arbor, MI, United States
- Howard Hughes Medical Institute, Ann Arbor, MI, United States
- University of Michigan Rogel Cancer Center, Ann Arbor, MI, United States
| | - Marcin Cieslik
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, United States
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
- University of Michigan Rogel Cancer Center, Ann Arbor, MI, United States
| |
Collapse
|
33
|
Caddeo A, Maurotti S, Kovooru L, Romeo S. 3D culture models to study pathophysiology of steatotic liver disease. Atherosclerosis 2024; 393:117544. [PMID: 38677899 DOI: 10.1016/j.atherosclerosis.2024.117544] [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: 11/21/2023] [Revised: 03/19/2024] [Accepted: 04/10/2024] [Indexed: 04/29/2024]
Abstract
Steatotic liver disease (SLD) refers to a spectrum of diseases caused by hepatic lipid accumulation. SLD has emerged as the leading cause of chronic liver disease worldwide. Despite this burden and many years, understanding the pathophysiology of this disease is challenging due to the inaccessibility to human liver specimens. Therefore, cell-based in vitro systems are widely used as models to investigate the pathophysiology of SLD. Culturing hepatic cells in monolayers causes the loss of their hepatocyte-specific phenotype and, consequently, tissue-specific function and architecture. Hence, three-dimensional (3D) culture models allow cells to mimic the in vivo microenvironment and spatial organization of the liver unit. The utilization of 3D in vitro models minimizes the drawbacks of two-dimensional (2D) cultures and aligns with the 3Rs principles to alleviate the number of in vivo experiments. This article provides an overview of liver 3D models highlighting advantages and limitations, and culminates by discussing their applications in pharmaceutical and biomedical research.
Collapse
Affiliation(s)
- Andrea Caddeo
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy.
| | - Samantha Maurotti
- Department of Clinical and Experimental Medicine, University Magna Graecia, Catanzaro, Italy
| | - Lohitesh Kovooru
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden; Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy.
| |
Collapse
|
34
|
Min K, Karuppannan SK, Tae G. The impact of matrix stiffness on hepatic cell function, liver fibrosis, and hepatocellular carcinoma-Based on quantitative data. BIOPHYSICS REVIEWS 2024; 5:021306. [PMID: 38846007 PMCID: PMC11151446 DOI: 10.1063/5.0197875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/14/2024] [Indexed: 06/09/2024]
Abstract
Over the past few decades, extensive research has explored the development of supportive scaffold materials for in vitro hepatic cell culture, to effectively mimic in vivo microenvironments. It is crucial for hepatic disease modeling, drug screening, and therapeutic evaluations, considering the ethical concerns and practical challenges associated with in vivo experiments. This review offers a comprehensive perspective on hepatic cell culture using bioscaffolds by encompassing all stages of hepatic diseases-from a healthy liver to fibrosis and hepatocellular carcinoma (HCC)-with a specific focus on matrix stiffness. This review begins by providing physiological and functional overviews of the liver. Subsequently, it explores hepatic cellular behaviors dependent on matrix stiffness from previous reports. For hepatic cell activities, softer matrices showed significant advantages over stiffer ones in terms of cell proliferation, migration, and hepatic functions. Conversely, stiffer matrices induced myofibroblastic activation of hepatic stellate cells, contributing to the further progression of fibrosis. Elevated matrix stiffness also correlates with HCC by increasing proliferation, epithelial-mesenchymal transition, metastasis, and drug resistance of HCC cells. In addition, we provide quantitative information on available data to offer valuable perspectives for refining the preparation and development of matrices for hepatic tissue engineering. We also suggest directions for further research on this topic.
Collapse
Affiliation(s)
- Kiyoon Min
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Sathish Kumar Karuppannan
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
35
|
Gondal MN, Shah SUR, Chinnaiyan AM, Cieslik M. A Systematic Overview of Single-Cell Transcriptomics Databases, their Use cases, and Limitations. ARXIV 2024:arXiv:2404.10545v1. [PMID: 38699169 PMCID: PMC11065044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Rapid advancements in high-throughput single-cell RNA-seq (scRNA-seq) technologies and experimental protocols have led to the generation of vast amounts of genomic data that populates several online databases and repositories. Here, we systematically examined large-scale scRNA-seq databases, categorizing them based on their scope and purpose such as general, tissue-specific databases, disease-specific databases, cancer-focused databases, and cell type-focused databases. Next, we discuss the technical and methodological challenges associated with curating large-scale scRNA-seq databases, along with current computational solutions. We argue that understanding scRNA-seq databases, including their limitations and assumptions, is crucial for effectively utilizing this data to make robust discoveries and identify novel biological insights. Furthermore, we propose that bridging the gap between computational and wet lab scientists through user-friendly web-based platforms is needed for democratizing access to single-cell data. These platforms would facilitate interdisciplinary research, enabling researchers from various disciplines to collaborate effectively. This review underscores the importance of leveraging computational approaches to unravel the complexities of single-cell data and offers a promising direction for future research in the field.
Collapse
Affiliation(s)
- Mahnoor N. Gondal
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI USA
| | - Saad Ur Rehman Shah
- Gies College of Business, University of Illinois Business College, Champaign, IL USA
| | - Arul M. Chinnaiyan
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI USA
- Department of Pathology, University of Michigan, Ann Arbor, MI USA
- Department of Urology, University of Michigan, Ann Arbor, MI USA
- Howard Hughes Medical Institute, Ann Arbor, MI USA
- University of Michigan Rogel Cancer Center, Ann Arbor, MI USA
| | - Marcin Cieslik
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI USA
- Department of Pathology, University of Michigan, Ann Arbor, MI USA
- University of Michigan Rogel Cancer Center, Ann Arbor, MI USA
| |
Collapse
|
36
|
Dare A, King SD, Chen SY. Surfactant protein A promotes western diet-induced hepatic steatosis and fibrosis in mice. Sci Rep 2024; 14:7464. [PMID: 38553537 PMCID: PMC10980756 DOI: 10.1038/s41598-024-58291-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) remains the most common cause of liver disease in the United States due to the increased incidence of metabolic dysfunction and obesity. Surfactant protein A (SPA) regulates macrophage function, strongly binds to lipids, and is implicated in renal and idiopathic pulmonary fibrosis (IPF). However, the role of SPA in lipid accumulation, inflammation, and hepatic fibrosis that characterize MASLD remains unknown. SPA deficient (SPA-/-) and age-matched wild-type (WT) control mice were fed a Western diet for 8 weeks to induce MASLD. Blood and liver samples were collected and used to analyze pathological features associated with MASLD. SPA expression was significantly upregulated in livers of mice with MASLD. SPA deficiency attenuated lipid accumulation along with downregulation of genes involved in fatty acid uptake and reduction of hepatic inflammation as evidenced by the diminished macrophage activation, decreased monocyte infiltration, and reduced production of inflammatory cytokines. Moreover, SPA-/- inhibited stellate cell activation, collagen deposit, and liver fibrosis. These results highlight the novel role of SPA in promoting fatty acid uptake into hepatocytes, causing excessive lipid accumulation, inflammation, and fibrosis implicated in the pathogenesis of MASLD.
Collapse
Affiliation(s)
- Ayobami Dare
- Department of Surgery, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO, 65212, USA
| | - Skylar D King
- Department of Surgery, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO, 65212, USA
| | - Shi-You Chen
- Department of Surgery, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO, 65212, USA.
- The Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA.
| |
Collapse
|
37
|
Lin Y, He L, Cai Y, Wang X, Wang S, Li F. The role of circadian clock in regulating cell functions: implications for diseases. MedComm (Beijing) 2024; 5:e504. [PMID: 38469551 PMCID: PMC10925886 DOI: 10.1002/mco2.504] [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: 07/19/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 03/13/2024] Open
Abstract
The circadian clock system orchestrates daily behavioral and physiological rhythms, facilitating adaptation to environmental and internal oscillations. Disruptions in circadian rhythms have been linked to increased susceptibility to various diseases and can exacerbate existing conditions. This review delves into the intricate regulation of diurnal gene expression and cell function by circadian clocks across diverse tissues. . Specifically, we explore the rhythmicity of gene expressions, behaviors, and functions in both immune and non-immune cells, elucidating the regulatory effects and mechanisms imposed by circadian clocks. A detailed discussion is centered on elucidating the complex functions of circadian clocks in regulating key cellular signaling pathways. We further review the circadian regulation in diverse diseases, with a focus on inflammatory diseases, cancers, and systemic diseases. By highlighting the intimate interplay between circadian clocks and diseases, especially through clock-controlled cell function, this review contributes to the development of novel disease intervention strategies. This enhanced understanding holds significant promise for the design of targeted therapies that can exploit the circadian regulation mechanisms for improved treatment efficacy.
Collapse
Affiliation(s)
- Yanke Lin
- Infectious Diseases InstituteGuangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
- Guangdong TCRCure Biopharma Technology Co., Ltd.GuangzhouChina
| | | | - Yuting Cai
- School of Pharmaceutical SciencesGuangzhou University of Chinese MedicineGuangzhouChina
| | - Xiaokang Wang
- Department of PharmacyShenzhen Longhua District Central HospitalShenzhenChina
| | - Shuai Wang
- School of Pharmaceutical SciencesGuangzhou University of Chinese MedicineGuangzhouChina
| | - Feng Li
- Infectious Diseases InstituteGuangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
38
|
Sun C, Zhu D, Zhu Q, He Z, Lou Y, Chen D. The significance of gut microbiota in the etiology of autoimmune hepatitis: a narrative review. Front Cell Infect Microbiol 2024; 14:1337223. [PMID: 38404291 PMCID: PMC10884129 DOI: 10.3389/fcimb.2024.1337223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 01/04/2024] [Indexed: 02/27/2024] Open
Abstract
Autoimmune hepatitis (AIH) is a chronic inflammatory disease of the liver that is mediated by autoimmunity and has complex pathogenesis. Its prevalence has increased globally. Since the liver is the first organ to be exposed to harmful substances, such as gut-derived intestinal microbiota and its metabolites, gut health is closely related to liver health, and the "liver-gut axis" allows abnormalities in the gut microbiota to influence the development of liver-related diseases such as AIH. Changes in the composition of the intestinal microbiota and its resultant disruption of the intestinal barrier and microbial transport are involved in multiple ways in the disruption of immune homeostasis and inflammation, thereby influencing the development of AIH. In terms of the mechanisms involved in immune, the gut microbiota or its metabolites, which is decreased in secondary bile acids, short-chain fatty acids (SCFAs), and polyamines, and increased in lipopolysaccharide (LPS), branched-chain amino acids (BCAA), tryptophan metabolite, amino acid, and bile acid, can disrupt immune homeostasis by activating various immune cells and immune-related signaling pathways, resulting in aberrant activation of the immune system. Clarifying this mechanism has significant clinical implications for the treatment of AIH with drugs that target intestinal microbiota and related signaling pathways. Therefore, this narrative review summarizes the progress in exploring the involvement of gut microbiota in the pathogenesis of AIH, with the aim of helping to improve the precise targeting of therapeutic treatments against AIH for the benefit of clinical AIH treatment.
Collapse
Affiliation(s)
- Chen Sun
- Clinical Research Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongzi Zhu
- Department of General Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Zhu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeping He
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yichao Lou
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Desheng Chen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
Zailaie SA, Khoja BB, Siddiqui JJ, Mawardi MH, Heaphy E, Aljagthmi A, Sergi CM. Investigating the Role of Non-Coding RNA in Non-Alcoholic Fatty Liver Disease. Noncoding RNA 2024; 10:10. [PMID: 38392965 PMCID: PMC10891858 DOI: 10.3390/ncrna10010010] [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: 12/12/2023] [Revised: 01/22/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Non-coding RNAs (ncRNAs) are RNA molecules that do not code for protein but play key roles in regulating cellular processes. NcRNAs globally affect gene expression in diverse physiological and pathological contexts. Functionally important ncRNAs act in chromatin modifications, in mRNA stabilization and translation, and in regulation of various signaling pathways. Non-alcoholic fatty liver disease (NAFLD) is a set of conditions caused by the accumulation of triacylglycerol in the liver. Studies of ncRNA in NAFLD are limited but have demonstrated that ncRNAs play a critical role in the pathogenesis of NAFLD. In this review, we summarize NAFLD's pathogenesis and clinical features, discuss current treatment options, and review the involvement of ncRNAs as regulatory molecules in NAFLD and its progression to non-alcoholic steatohepatitis (NASH). In addition, we highlight signaling pathways dysregulated in NAFLD and review their crosstalk with ncRNAs. Having a thorough understanding of the disease process's molecular mechanisms will facilitate development of highly effective diagnostic and therapeutic treatments. Such insights can also inform preventive strategies to minimize the disease's future development.
Collapse
Affiliation(s)
- Samar A. Zailaie
- Research Center, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia; (S.A.Z.); (B.B.K.); (E.H.); (A.A.)
| | - Basmah B. Khoja
- Research Center, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia; (S.A.Z.); (B.B.K.); (E.H.); (A.A.)
| | - Jumana J. Siddiqui
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Mohammad H. Mawardi
- Medicine Department, Gastroenterology Section, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia;
| | - Emily Heaphy
- Research Center, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia; (S.A.Z.); (B.B.K.); (E.H.); (A.A.)
| | - Amjad Aljagthmi
- Research Center, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia; (S.A.Z.); (B.B.K.); (E.H.); (A.A.)
| | - Consolato M. Sergi
- Children’s Hospital of Eastern Ontario (CHEO), University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|
40
|
Pecqueux M, Brückner F, Oehme F, Hempel S, Baenke F, Riediger C, Distler M, Weitz J, Kahlert C. Preoperative IL-8 levels as prognostic indicators of overall survival: an extended follow-up in a prospective cohort with colorectal liver metastases. BMC Cancer 2024; 24:90. [PMID: 38233759 PMCID: PMC10792859 DOI: 10.1186/s12885-023-11787-1] [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: 10/14/2023] [Accepted: 12/21/2023] [Indexed: 01/19/2024] Open
Abstract
INTRODUCTION CRC with liver metastases is a major contributor to cancer-related mortality. Despite advancements in liver resection techniques, patient survival remains a concern due to high recurrence rates. This study seeks to uncover prognostic biomarkers that predict overall survival in patients undergoing curative hepatic resection for CRC liver metastases. METHODS Prospectively collected serum samples from a cohort of 49 patients who received curative hepatic resection for CRC liver metastases were studied. The patients are part of a cohort, previously analyzed for perioperative complications (see methods). Various preoperative serum markers, clinical characteristics, and factors were analyzed. Univariate and multivariate Cox regression analyses were conducted to determine associations between these variables and disease-free survival as well as overall survival. RESULTS For disease-free survival, univariate analysis highlighted the correlation between poor outcomes and advanced primary tumor stage, high ASA score, and synchronous liver metastases. Multivariate analysis identified nodal-positive primary tumors and synchronous metastases as independent risk factors for disease-free survival. Regarding overall survival, univariate analysis demonstrated significant links between poor survival and high preoperative IL-8 levels, elevated neutrophil-lymphocyte ratio (NLR), and presence of metastases in other organs. Multivariate analysis confirmed preoperative IL-8 and having three or more liver metastases as independent risk factors for overall survival. The impact of IL-8 on survival was particularly noteworthy, surpassing the influence of established clinical factors. CONCLUSION This study establishes preoperative IL-8 levels as a potential prognostic biomarker for overall survival in patients undergoing curative liver resection for CRC liver metastases. This study underscores the importance of incorporating IL-8 and other biomarkers into clinical decision-making, facilitating improved patient stratification and tailored treatment approaches. Further research and validation studies are needed to solidify the clinical utility of IL-8 as a prognostic marker.
Collapse
Affiliation(s)
- Mathieu Pecqueux
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany.
| | - Frederik Brückner
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Florian Oehme
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Sebastian Hempel
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Franziska Baenke
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Carina Riediger
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Marius Distler
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Christoph Kahlert
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| |
Collapse
|
41
|
Wang Z, Liu N, Yang Y, Tu Z. The novel mechanism facilitating chronic hepatitis B infection: immunometabolism and epigenetic modification reprogramming. Front Immunol 2024; 15:1349867. [PMID: 38288308 PMCID: PMC10822934 DOI: 10.3389/fimmu.2024.1349867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024] Open
Abstract
Hepatitis B Virus (HBV) infections pose a global public health challenge. Despite extensive research on this disease, the intricate mechanisms underlying persistent HBV infection require further in-depth elucidation. Recent studies have revealed the pivotal roles of immunometabolism and epigenetic reprogramming in chronic HBV infection. Immunometabolism have identified as the process, which link cell metabolic status with innate immunity functions in response to HBV infection, ultimately contributing to the immune system's inability to resolve Chronic Hepatitis B (CHB). Within hepatocytes, HBV replication leads to a stable viral covalently closed circular DNA (cccDNA) minichromosome located in the nucleus, and epigenetic modifications in cccDNA enable persistence of infection. Additionally, the accumulation or depletion of metabolites not only directly affects the function and homeostasis of immune cells but also serves as a substrate for regulating epigenetic modifications, subsequently influencing the expression of antiviral immune genes and facilitating the occurrence of sustained HBV infection. The interaction between immunometabolism and epigenetic modifications has led to a new research field, known as metabolic epigenomics, which may form a mutually reinforcing relationship with CHB. Herein, we review the recent studies on immunometabolism and epigenetic reprogramming in CHB infection and discuss the potential mechanisms of persistent HBV infection. A deeper understanding of these mechanisms will offer novel insights and targets for intervention strategies against chronic HBV infection, thereby providing new hope for the treatment of related diseases.
Collapse
Affiliation(s)
- Zhengmin Wang
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Nan Liu
- Institute of Epigenetic Medicine, First Hospital of Jilin University, Changchun, China
| | - Yang Yang
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhengkun Tu
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin, China
- Institute of Liver Diseases, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
42
|
Schwarz C, Göring J, Grüttner C, Hilger I. Intravenous Injection of PEI-Decorated Iron Oxide Nanoparticles Impacts NF-kappaB Protein Expression in Immunologically Stressed Mice. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:3166. [PMID: 38133063 PMCID: PMC10745731 DOI: 10.3390/nano13243166] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
Nanoparticle-based formulations are considered valuable tools for diagnostic and treatment purposes. The surface decoration of nanoparticles with polyethyleneimine (PEI) is often used to enhance their targeting and functional properties. Here, we aimed at addressing the long-term fate in vivo and the potential "off-target" effects of PEI decorated iron oxide nanoparticles (PEI-MNPs) in individuals with low-grade and persistent systemic inflammation. For this purpose, we synthesized PEI-MNPs (core-shell method, PEI coating under high pressure homogenization). Further on, we induced a low-grade and persistent inflammation in mice through regular subcutaneous injection of pathogen-associated molecular patterns (PAMPs, from zymosan). PEI-MNPs were injected intravenously. Up to 7 weeks thereafter, the blood parameters were determined via automated fluorescence flow cytometry, animals were euthanized, and the organs analyzed for iron contents (atomic absorption spectrometry) and for expression of NF-κB associated proteins (p65, IκBα, p105/50, p100/52, COX-2, Bcl-2, SDS-PAGE and Western blotting). We observed that the PEI-MNPs had a diameter of 136 nm and a zeta-potential 56.9 mV. After injection in mice, the blood parameters were modified and the iron levels were increased in different organs. Moreover, the liver of animals showed an increased protein expression of canonical NF-κB signaling pathway members early after PEI-MNP application, whereas at the later post-observation time, members of the non-canonical signaling pathway were prominent. We conclude that the synergistic effect between PEI-MNPs and the low-grade and persistent inflammatory state is mainly due to the hepatocytes sensing infection (PAMPs), to immune responses resulting from the intracellular metabolism of the uptaken PEI-MNPs, or to hepatocyte and immune cell communications. Therefore, we suggest a careful assessment of the safety and toxicity of PEI-MNP-based carriers for gene therapy, chemotherapy, and other medical applications not only in healthy individuals but also in those suffering from chronic inflammation.
Collapse
Affiliation(s)
- Claudia Schwarz
- Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, D-07740 Jena, Germany; (C.S.); (J.G.)
| | - Julia Göring
- Experimental Radiology, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, D-07740 Jena, Germany; (C.S.); (J.G.)
| | - Cordula Grüttner
- Micromod Partikeltechnologie GmbH, Schillingallee 68, D-18057 Rostock, Germany;
| | - Ingrid Hilger
- Micromod Partikeltechnologie GmbH, Schillingallee 68, D-18057 Rostock, Germany;
| |
Collapse
|
43
|
Lu Y, Chen QM, An L. Semi-reference based cell type deconvolution with application to human metastatic cancers. NAR Genom Bioinform 2023; 5:lqad109. [PMID: 38143958 PMCID: PMC10748484 DOI: 10.1093/nargab/lqad109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/01/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023] Open
Abstract
Bulk RNA-seq experiments, commonly used to discern gene expression changes across conditions, often neglect critical cell type-specific information due to their focus on average transcript abundance. Recognizing cell type contribution is crucial to understanding phenotype and disease variations. The advent of single-cell RNA sequencing has allowed detailed examination of cellular heterogeneity; however, the cost and analytic caveat prohibits such sequencing for a large number of samples. We introduce a novel deconvolution approach, SECRET, that employs cell type-specific gene expression profiles from single-cell RNA-seq to accurately estimate cell type proportions from bulk RNA-seq data. Notably, SECRET can adapt to scenarios where the cell type present in the bulk data is unrepresented in the reference, thereby offering increased flexibility in reference selection. SECRET has demonstrated superior accuracy compared to existing methods using synthetic data and has identified unknown tissue-specific cell types in real human metastatic cancers. Its versatility makes it broadly applicable across various human cancer studies.
Collapse
Affiliation(s)
- Yingying Lu
- Interdisciplinary Program in Statistics and Data Science, University of Arizona, Tucson, AZ, USA
| | - Qin M Chen
- College of Pharmacy, University of Arizona, Tucson, AZ, USA
- Cancer Biology Program, University of Arizona, Tucson, AZ, USA
| | - Lingling An
- Interdisciplinary Program in Statistics and Data Science, University of Arizona, Tucson, AZ, USA
- Department of Biosystems Engineering, University of Arizona, Tucson, AZ, USA
- Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
44
|
Koksal AR, Ekmen N, Aydin Y, Nunez K, Sandow T, Delk M, Moehlen M, Thevenot P, Cohen A, Dash S. A Single-Step Immunocapture Assay to Quantify HCC Exosomes Using the Highly Sensitive Fluorescence Nanoparticle-Tracking Analysis. J Hepatocell Carcinoma 2023; 10:1935-1954. [PMID: 37936599 PMCID: PMC10627088 DOI: 10.2147/jhc.s423043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/07/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction Extracellular vesicles could serve as a non-invasive biomarker for early cancer detection. However, limited methods to quantitate cancer-derived vesicles in the native state remain a significant barrier to clinical translation. Aim This research aims to develop a rapid, one-step immunoaffinity approach to quantify HCC exosomes directly from a small serum volume. Methods HCC-derived exosomes in the serum were captured using fluorescent phycoerythrin (PE)-conjugated antibodies targeted to GPC3 and alpha-fetoprotein (AFP). Total and HCC-specific exosomes were then quantified in culture supernatant or patient-derived serums using fluorescence nanoparticle tracking analysis (F-NTA). The performance of HCC exosome quantification in the serum was compared with the tumor size determined by MRI. Results Initially we tested the detection limits of the F-NTA using synthetic fluorescent and non-fluorescent beads. The assay showed an acceptable sensitivity with a detection range of 104-108 particles/mL. Additionally, the combination of immunocapture followed by size-exclusion column purification allows the isolation of smaller-size EVs and quantification by F-NTA. Our assay demonstrated that HCC cell culture releases a significantly higher quantity of GPC3 or GPC3+AFP positive EVs (100-200 particles/cell) compared to non-HCC culture (10-40 particles/cell) (p<0.01 and p<0.05 respectively). The F-NTA enables absolute counting of HCC-specific exosomes in the clinical samples with preserved biological immunoreactivity. The performance of F-NTA was clinically validated in serum from patients ± cirrhosis and with confirmed HCC. F-NTA quantification data show selective enrichment of AFP and GPC3 positive EVs in HCC serum compared to malignancy-free cirrhosis (AUC values for GPC3, AFP, and GPC3/AFP were found 0.79, 0.71, and 0.72 respectively). The MRI-confirmed patient cohort indicated that there was a positive correlation between total tumor size and GPC3-positive exosome concentration (r:0.78 and p<0.001). Conclusion We developed an immunocapture assay that can be used for simultaneous isolation and quantification of HCC-derived exosomes from a small serum volume with high accuracy.
Collapse
Affiliation(s)
- Ali Riza Koksal
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Nergiz Ekmen
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Yucel Aydin
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Kelley Nunez
- Department of Gastroenterology and Hepatology, Institute of Translational Research, Ochsner Health, New Orleans, LA, USA
| | - Tyler Sandow
- Department of Radiology, Institute of Translational Research, Ochsner Health, New Orleans, LA, USA
| | - Molly Delk
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Martin Moehlen
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Paul Thevenot
- Department of Gastroenterology and Hepatology, Institute of Translational Research, Ochsner Health, New Orleans, LA, USA
| | - Ari Cohen
- Department of Gastroenterology and Hepatology, Institute of Translational Research, Ochsner Health, New Orleans, LA, USA
- Multi-Organ Transplant Institute, Ochsner Health, New Orleans, LA, USA
| | - Srikanta Dash
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| |
Collapse
|
45
|
Heo YJ, Lee N, Choi SE, Jeon JY, Han SJ, Kim DJ, Kang Y, Lee KW, Kim HJ. Amphiregulin Induces iNOS and COX-2 Expression through NF- κB and MAPK Signaling in Hepatic Inflammation. Mediators Inflamm 2023; 2023:2364121. [PMID: 37868614 PMCID: PMC10586434 DOI: 10.1155/2023/2364121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/09/2023] [Accepted: 09/16/2023] [Indexed: 10/24/2023] Open
Abstract
Background Inflammation is a major cause of hepatic tissue damage and accelerates the progression of nonalcoholic fatty liver disease (NAFLD). Amphiregulin (AREG), an epidermal growth factor receptor ligand, is associated with human liver cirrhosis and hepatocellular carcinoma. We aimed to investigate the effects of AREG on hepatic inflammation during NAFLD progression, in vivo and in vitro. Methods AREG gene expression was measured in the liver of mice fed a methionine choline-deficient (MCD) diet for 2 weeks. We evaluated inflammatory mediators and signaling pathways in HepG2 cells after stimulation with AREG. Nitric oxide (NO), prostaglandin E2 (PGE2), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) were analyzed using an enzyme-linked immunosorbent assay and western blotting. Nuclear transcription factor kappa-B (NF-κB) and mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38 mitogen-activated protein kinase, were analyzed using western blotting. Results Proinflammatory cytokines (interleukin (IL)-6, IL-1β, and IL-8) and immune cell recruitment (as indicated by L3T4, F4/80, and ly6G mRNA expression) increased, and expression of AREG increased in the liver of mice fed the MCD diet. AREG significantly increased the expression of IL-6 and IL-1β and the production of NO, PGE2, and IL-8 in HepG2 cells. It also activated the protein expression of iNOS and COX-2. AREG-activated NF-κB and MAPKs signaling, and together with NF-κB and MAPKs inhibitors, AREG significantly reduced the protein expression of iNOS and COX-2. Conclusion AREG plays a role in hepatic inflammation by increasing iNOS and COX-2 expression via NF-κB and MAPKs signaling.
Collapse
Affiliation(s)
- Yu Jung Heo
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 206, World cup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
- Institute of Medical Science, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Nami Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 206, World cup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
| | - Sung-E. Choi
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Ja Young Jeon
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 206, World cup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
| | - Seung Jin Han
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 206, World cup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
| | - Dae Jung Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 206, World cup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
| | - Yup Kang
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Kwan Woo Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 206, World cup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
| | - Hae Jin Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 206, World cup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
| |
Collapse
|
46
|
Zong X, Ding Q, Liu X, Liu Q, Song S, Yan X, Zhang Y. Preventive Effect of 6-shogaol on D-galactosamine Induced Hepatotoxicity Through NF-?B/MAPK Signaling Pathway in Rats. Physiol Res 2023; 72:445-454. [PMID: 37795887 PMCID: PMC10634558 DOI: 10.33549/physiolres.935092] [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: 03/03/2023] [Accepted: 04/19/2023] [Indexed: 01/05/2024] Open
Abstract
This analysis aims to see whether 6-shogaol could protect rats against D-galactosamine (D-GalN)-induced Hepatotoxicity. The Wistar rats were divided into four groups (n=6). Group 1 received a standard diet, Group 2 received an oral administration of 6-shogaol (20 mg/kg b.wt), Group 3 received an intraperitoneal injection of D-GalN (400 mg/kg b.wt) on 21st day, and Group 4 received an oral administration of 6-shogaol (20mg/kg b.wt) for 21 days and D-GalN (400 mg/kg b.wt) injection only on 21st day. The hepatic marker enzymes activity, lipid peroxidative markers level increased significantly and antioxidant activity/level significantly reduced in D-GalN-induced rats. 6-shogaol Pretreatment effectively improves the above changes in D-GalN-induced rats. Further, inflammatory marker expression and MAPK signaling molecules were downregulated by 6-shogaol. These findings showed that 6-shogaol exerts hepatoprotective effects via the enhanced antioxidant system and attenuated the inflammation and MAPK signaling pathway in D-GalN-induced rats.
Collapse
Affiliation(s)
- X Zong
- Department of Clinical Laboratory, Baoding First Central Hospital, Baoding, Hebei province, China.
| | | | | | | | | | | | | |
Collapse
|
47
|
Zhou S, Li H, Wang H, Wang R, Song W, Li D, Wei C, Guo Y, He X, Deng Y. Nickel Nanoparticles Induced Hepatotoxicity in Mice via Lipid-Metabolism-Dysfunction-Regulated Inflammatory Injury. Molecules 2023; 28:5757. [PMID: 37570729 PMCID: PMC10421287 DOI: 10.3390/molecules28155757] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Nickel nanoparticles (NiNPs) have wide applications in industry and biomedicine due to their unique characteristics. The liver is the major organ responsible for nutrient metabolism, exogenous substance detoxification and biotransformation of medicines containing nanoparticles. Hence, it is urgent to further understand the principles and potential mechanisms of hepatic effects on NiNPs administration. In this study, we explored the liver impacts in male C57/BL6 mice through intraperitoneal injection with NiNPs at doses of 10, 20 and 40 mg/kg/day for 7 and 28 days. The results showed that NiNPs treatment increased serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and induced pathological changes in liver tissues. Moreover, hepatic triglyceride (TG) content and lipid droplet deposition identified via de novo lipogenesis (DNL) progression were enhanced after NiNPs injection. Additionally, sustained NiNPs exposure induced a remarkable hepatic inflammatory response, significantly promoted endoplasmic reticulum stress (ER stress) sensors Ire1α, Perk and Atf6, and activated the occurrence of liver cell apoptosis. Overall, the research indicated that NiNPs exposure induced liver injury and disturbance of lipid metabolism. These findings revealed the public hazard from extreme exposure to NiNPs and provided new information on biological toxicity and biosafety evaluation.
Collapse
Affiliation(s)
- Shuang Zhou
- Beijing Institute of Medical Device Testing, Beijing Center for Testing and Research of Medical Biological Protective Equipment, Beijing 101111, China; (S.Z.)
- Beijing Institute of Technology, School of Life Science, Beijing 100081, China
| | - Hua Li
- Beijing Institute of Medical Device Testing, Beijing Center for Testing and Research of Medical Biological Protective Equipment, Beijing 101111, China; (S.Z.)
| | - Hui Wang
- Beijing Institute of Medical Device Testing, Beijing Center for Testing and Research of Medical Biological Protective Equipment, Beijing 101111, China; (S.Z.)
| | - Rui Wang
- Beijing Institute of Medical Device Testing, Beijing Center for Testing and Research of Medical Biological Protective Equipment, Beijing 101111, China; (S.Z.)
| | - Wei Song
- Beijing Institute of Medical Device Testing, Beijing Center for Testing and Research of Medical Biological Protective Equipment, Beijing 101111, China; (S.Z.)
| | - Da Li
- Beijing Institute of Medical Device Testing, Beijing Center for Testing and Research of Medical Biological Protective Equipment, Beijing 101111, China; (S.Z.)
| | - Changlei Wei
- Beijing Institute of Medical Device Testing, Beijing Center for Testing and Research of Medical Biological Protective Equipment, Beijing 101111, China; (S.Z.)
| | - Yu Guo
- Beijing Institute of Medical Device Testing, Beijing Center for Testing and Research of Medical Biological Protective Equipment, Beijing 101111, China; (S.Z.)
| | - Xueying He
- Beijing Institute of Medical Device Testing, Beijing Center for Testing and Research of Medical Biological Protective Equipment, Beijing 101111, China; (S.Z.)
| | - Yulin Deng
- Beijing Institute of Technology, School of Life Science, Beijing 100081, China
| |
Collapse
|
48
|
Yi Q, Yang J, Wu Y, Wang Y, Cao Q, Wen W. Immune microenvironment changes of liver cirrhosis: emerging role of mesenchymal stromal cells. Front Immunol 2023; 14:1204524. [PMID: 37539053 PMCID: PMC10395751 DOI: 10.3389/fimmu.2023.1204524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/21/2023] [Indexed: 08/05/2023] Open
Abstract
Cirrhosis is a progressive and diffuse liver disease characterized by liver tissue fibrosis and impaired liver function. This condition is brought about by several factors, including chronic hepatitis, hepatic steatosis, alcohol abuse, and other immunological injuries. The pathogenesis of liver cirrhosis is a complex process that involves the interaction of various immune cells and cytokines, which work together to create the hepatic homeostasis imbalance in the liver. Some studies have indicated that alterations in the immune microenvironment of liver cirrhosis are closely linked to the development and prognosis of the disease. The noteworthy function of mesenchymal stem cells and their paracrine secretion lies in their ability to promote the production of cytokines, which in turn enhance the self-repairing capabilities of tissues. The objective of this review is to provide a summary of the alterations in liver homeostasis and to discuss intercellular communication within the organ. Recent research on MSCs is yielding a blueprint for cell typing and biomarker immunoregulation. Hopefully, as MSCs researches continue to progress, novel therapeutic approaches will emerge to address cirrhosis.
Collapse
Affiliation(s)
- Qiuyun Yi
- National Center for Liver Cancer, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Jinxian Yang
- National Center for Liver Cancer, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ying Wu
- Department of Breast and Thyroid Surgery, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Ying Wang
- Department of Laboratory Diagnosis, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Qiqi Cao
- National Center for Liver Cancer, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wen Wen
- National Center for Liver Cancer, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Laboratory Diagnosis, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
49
|
Han DW, Xu K, Jin ZL, Xu YN, Li YH, Wang L, Cao Q, Kim KP, Ryu D, Hong K, Kim NH. Customized liver organoids as an advanced in vitro modeling and drug discovery platform for non-alcoholic fatty liver diseases. Int J Biol Sci 2023; 19:3595-3613. [PMID: 37497008 PMCID: PMC10367556 DOI: 10.7150/ijbs.85145] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/12/2023] [Indexed: 07/28/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and its progressive form non-alcoholic steatohepatitis (NASH) have presented a major and common health concern worldwide due to their increasing prevalence and progressive development of severe pathological conditions such as cirrhosis and liver cancer. Although a large number of drug candidates for the treatment of NASH have entered clinical trial testing, all have not been released to market due to their limited efficacy, and there remains no approved treatment for NASH available to this day. Recently, organoid technology that produces 3D multicellular aggregates with a liver tissue-like cytoarchitecture and improved functionality has been suggested as a novel platform for modeling the human-specific complex pathophysiology of NAFLD and NASH. In this review, we describe the cellular crosstalk between each cellular compartment in the liver during the pathogenesis of NAFLD and NASH. We also summarize the current state of liver organoid technology, describing the cellular diversity that could be recapitulated in liver organoids and proposing a future direction for liver organoid technology as an in vitro platform for disease modeling and drug discovery for NAFLD and NASH.
Collapse
Affiliation(s)
- Dong Wook Han
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
- Research and Development, Qingdao Haier Biotech Co. Ltd, Qingdao, China
- Guangdong ORGANOID Biotechnology Co. Ltd, Jiangmen, China
| | - KangHe Xu
- Department of Surgery, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Zhe-Long Jin
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
- Guangdong ORGANOID Biotechnology Co. Ltd, Jiangmen, China
| | - Yong-Nan Xu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
| | - Ying-Hua Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
| | - Lin Wang
- Research and Development, Qingdao Haier Biotech Co. Ltd, Qingdao, China
| | - Qilong Cao
- Research and Development, Qingdao Haier Biotech Co. Ltd, Qingdao, China
| | - Kee-Pyo Kim
- Department of Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - DongHee Ryu
- Department of Surgery, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, The institute of advanced regenerative science, Konkuk University, Seoul, Republic of Korea
| | - Nam-Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
- Research and Development, Qingdao Haier Biotech Co. Ltd, Qingdao, China
- Guangdong ORGANOID Biotechnology Co. Ltd, Jiangmen, China
| |
Collapse
|