1
|
Zhao G, Hu Y. Mechanistic insights into intrauterine adhesions. Semin Immunopathol 2024; 47:3. [PMID: 39613882 DOI: 10.1007/s00281-024-01030-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/14/2024] [Indexed: 12/01/2024]
Abstract
Intrauterine adhesions (IUA), also known as Asherman's syndrome, arise from damage to the basal layer of the endometrium, frequently caused by intrauterine interventions. This damage leads to nonregenerative healing of endometrium resulting in replacement by fibrous connective tissue, which bring about the adherence of opposing endometrium to render the uterine cavity and/or cervical canal partially or completely obliterated. IUA is a common cause of the refractory uterine infertility. Hysteroscopy is the gold standard for diagnosis of IUA. However, the method of accurately predicting the likelihood of achieving a live birth in the future remains established. Classical treatments have shown limited success, particularly in severe cases. Therefore, utilizing new research methods to deepen the understanding of the pathogenesis of IUA will facilitate the new treatment approaches to be found. In this article we briefly described the advances in the pathogenesis of IUA, with focus on inflammation and parenchymal cellular homeostasis disruption, defects in autophagy and the role of ferroptosis, and we also outlined the progress in IUA therapy.
Collapse
Affiliation(s)
- Guangfeng Zhao
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yali Hu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
2
|
Sui Y, Geng X, Wang Z, Zhang J, Yang Y, Meng Z. Targeting the regulation of iron homeostasis as a potential therapeutic strategy for nonalcoholic fatty liver disease. Metabolism 2024; 157:155953. [PMID: 38885833 DOI: 10.1016/j.metabol.2024.155953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/09/2024] [Accepted: 06/09/2024] [Indexed: 06/20/2024]
Abstract
With aging and the increasing incidence of obesity, nonalcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide. NAFLD mainly includes simple hepatic steatosis, nonalcoholic steatohepatitis (NASH), liver fibrosis and hepatocellular carcinoma (HCC). An imbalance in hepatic iron homeostasis is usually associated with the progression of NAFLD and induces iron overload, reactive oxygen species (ROS) production, and lipid peroxide accumulation, which leads to ferroptosis. Ferroptosis is a unique type of programmed cell death (PCD) that is characterized by iron dependence, ROS production and lipid peroxidation. The ferroptosis inhibition systems involved in NAFLD include the solute carrier family 7 member 11 (SLC7A11)/glutathione (GSH)/glutathione peroxidase 4 (GPX4) and ferroptosis suppressor protein 1 (FSP1)/coenzyme Q10 (CoQ10)/nicotinamide adenine dinucleotide phosphate (NADPH) regulatory axes. The main promotion system involved is the acyl-CoA synthetase long-chain family (ACSL4)/arachidonic lipoxygenase 15 (ALOX15) axis. In recent years, an increasing number of studies have focused on the multiple roles of iron homeostasis imbalance and ferroptosis in the progression of NAFLD. This review highlights the latest studies about iron homeostasis imbalance- and ferroptosis-associated NAFLD, mainly including the physiology and pathophysiology of hepatic iron metabolism, hepatic iron homeostasis imbalance during the development of NAFLD, and key regulatory molecules and roles of hepatic ferroptosis in NAFLD. This review aims to provide innovative therapeutic strategies for NAFLD.
Collapse
Affiliation(s)
- Yutong Sui
- Shenzhen Hospital, Southern Medical University, Shenzhen 518100, Guangdong, China
| | - Xue Geng
- Department of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Ziwei Wang
- Shenzhen Hospital, Southern Medical University, Shenzhen 518100, Guangdong, China
| | - Jing Zhang
- Shenzhen Hospital, Southern Medical University, Shenzhen 518100, Guangdong, China
| | - Yanqun Yang
- Shenzhen Hospital, Southern Medical University, Shenzhen 518100, Guangdong, China.
| | - Ziyu Meng
- NHC Key Laboratory of Hormones and Development, Tianjin Medical University Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China; Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300134, China.
| |
Collapse
|
3
|
Savic N, Markelic M, Stancic A, Velickovic K, Grigorov I, Vucetic M, Martinovic V, Gudelj A, Otasevic V. Sulforaphane prevents diabetes-induced hepatic ferroptosis by activating Nrf2 signaling axis. Biofactors 2024; 50:810-827. [PMID: 38299761 DOI: 10.1002/biof.2042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 12/25/2023] [Indexed: 02/02/2024]
Abstract
Recently, we characterized the ferroptotic phenotype in the liver of diabetic mice and revealed nuclear factor (erythroid-derived-2)-related factor 2 (Nrf2) inactivation as an integral part of hepatic injury. Here, we aim to investigate whether sulforaphane, an Nrf2 activator and antioxidant, prevents diabetes-induced hepatic ferroptosis and the mechanisms involved. Male C57BL/6 mice were divided into four groups: control (vehicle-treated), diabetic (streptozotocin-induced; 40 mg/kg, from Days 1 to 5), diabetic sulforaphane-treated (2.5 mg/kg from Days 1 to 42) and non-diabetic sulforaphane-treated group (2.5 mg/kg from Days 1 to 42). Results showed that diabetes-induced inactivation of Nrf2 and decreased expression of its downstream antiferroptotic molecules critical for antioxidative defense (catalase, superoxide dismutases, thioredoxin reductase), iron metabolism (ferritin heavy chain (FTH1), ferroportin 1), glutathione (GSH) synthesis (cystine-glutamate antiporter system, cystathionase, glutamate-cysteine ligase catalitic subunit, glutamate-cysteine ligase modifier subunit, glutathione synthetase), and GSH recycling - glutathione reductase (GR) were reversed/increased by sulforaphane treatment. In addition, we found that the ferroptotic phenotype in diabetic liver is associated with increased ferritinophagy and decreased FTH1 immunopositivity. The antiferroptotic effect of sulforaphane was further evidenced through the increased level of GSH, decreased accumulation of labile iron and lipid peroxides (4-hydroxy-2-nonenal, lipofuscin), decreased ferritinophagy and liver damage (decreased fibrosis, alanine aminotransferase, and aspartate aminotransferase). Finally, diabetes-induced increase in serum glucose and triglyceride level was significantly reduced by sulforaphane. Regardless of the fact that this study is limited by the use of one model of experimentally induced diabetes, the results obtained demonstrate for the first time that sulforaphane prevents diabetes-induced hepatic ferroptosis in vivo through the activation of Nrf2 signaling pathways. This nominates sulforaphane as a promising phytopharmaceutical for the prevention/alleviation of ferroptosis in diabetes-related pathologies.
Collapse
Affiliation(s)
- Nevena Savic
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milica Markelic
- Department of Cell and Tissue Biology, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Ana Stancic
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ksenija Velickovic
- Department of Cell and Tissue Biology, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Ilijana Grigorov
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milica Vucetic
- Medical Biology Department, Centre Scientifique de Monaco (CSM), Monaco, Monaco
| | - Vesna Martinovic
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Andjelija Gudelj
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Vesna Otasevic
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
4
|
Wang J, Du M, Meng L, He S, Zhu Y, Yang Y, Ren X, Huang Y, Sun S, Dong R, Zheng S, Chen G. NLRP3 inflammasome activation promotes liver inflammation and fibrosis in experimental biliary atresia. Dig Liver Dis 2024; 56:458-467. [PMID: 37635054 DOI: 10.1016/j.dld.2023.08.039] [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: 02/26/2023] [Revised: 07/06/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Biliary atresia (BA) is characterized by a progressive fibroinflammatory cholangiopathy in early infants with unknown etiology. Although innate immune disorder is involved in its mechanism, role of NLRP3 inflammasome in BA remains largely undefined. AIM To explore the role of NLRP3 inflammasome in BA. METHODS The expressions of NLRP3 inflammasome-related genes were determined in BA patients. Role of NLRP3 inflammasome was evaluated using MCC950 in experimental BA. Furthermore, gadolinium chloride, a macrophage scavenger, was applied to validate the inflammasome's cellular localization. Finally, the effects of NLRP3 inflammasome activation on liver fibrosis were explored in vivo and vitro in experimental BA. RESULTS The components of NLRP3 inflammasome were up-regulated in BA patients. Inflammasome-related genes showed positively correlated with liver inflammation and fibrosis in BA patients. In experimental BA, inflammasome-related genes were up-regulated, and their expressions were inhibited by MCC950, which promoted mice growth, protected liver function, alleviated obstructive jaundice, inhibited liver inflammation, and reduced serum IL-1β level. NLRP3 inflammasome was expressed in macrophages, and macrophage elimination exerted the same protective roles as MCC950 did in BA. Additionally, NLRP3 inflammasome activation promoted liver fibrosis in experimental BA. CONCLUSIONS NLRP3 inflammasome activation in macrophages promoted liver inflammation and fibrosis in experimental BA.
Collapse
Affiliation(s)
- Junfeng Wang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, PR China
| | - Min Du
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, PR China; Department of Pediatric Gastroenterology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610091, PR China
| | - Lingdu Meng
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, PR China
| | - Shiwei He
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, PR China
| | - Ye Zhu
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, PR China
| | - Yifan Yang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, PR China
| | - Xue Ren
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, PR China
| | - Yanlei Huang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, PR China
| | - Song Sun
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, PR China
| | - Rui Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, PR China
| | - Shan Zheng
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, PR China.
| | - Gong Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, PR China.
| |
Collapse
|
5
|
Jouve M, Carpentier R, Kraiem S, Legrand N, Sobolewski C. MiRNAs in Alcohol-Related Liver Diseases and Hepatocellular Carcinoma: A Step toward New Therapeutic Approaches? Cancers (Basel) 2023; 15:5557. [PMID: 38067261 PMCID: PMC10705678 DOI: 10.3390/cancers15235557] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 06/29/2024] Open
Abstract
Alcohol-related Liver Disease (ALD) is the primary cause of chronic liver disorders and hepatocellular carcinoma (HCC) development in developed countries and thus represents a major public health concern. Unfortunately, few therapeutic options are available for ALD and HCC, except liver transplantation or tumor resection for HCC. Deciphering the molecular mechanisms underlying the development of these diseases is therefore of major importance to identify early biomarkers and to design efficient therapeutic options. Increasing evidence indicate that epigenetic alterations play a central role in the development of ALD and HCC. Among them, microRNA importantly contribute to the development of this disease by controlling the expression of several genes involved in hepatic metabolism, inflammation, fibrosis, and carcinogenesis at the post-transcriptional level. In this review, we discuss the current knowledge about miRNAs' functions in the different stages of ALD and their role in the progression toward carcinogenesis. We highlight that each stage of ALD is associated with deregulated miRNAs involved in hepatic carcinogenesis, and thus represent HCC-priming miRNAs. By using in silico approaches, we have uncovered new miRNAs potentially involved in HCC. Finally, we discuss the therapeutic potential of targeting miRNAs for the treatment of these diseases.
Collapse
Affiliation(s)
- Mickaël Jouve
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Rodolphe Carpentier
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Sarra Kraiem
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Noémie Legrand
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Cyril Sobolewski
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| |
Collapse
|
6
|
Zheng K, Hao F, Medrano-Garcia S, Chen C, Guo F, Morán-Blanco L, Rodríguez-Perales S, Torres-Ruiz R, Peligros MI, Vaquero J, Bañares R, Gómez Del Moral M, Regueiro JR, Martínez-Naves E, Mohamed MR, Gallego-Durán R, Maya D, Ampuero J, Romero-Gómez M, Gilbert-Ramos A, Guixé-Muntet S, Fernández-Iglesias A, Gracia-Sancho J, Coll M, Graupera I, Ginès P, Ciudin A, Rivera-Esteban J, Pericàs JM, Frutos MD, Ramos Molina B, Herranz JM, Ávila MA, Nevzorova YA, Fernández-Malavé E, Cubero FJ. Neuroblastoma RAS viral oncogene homolog (N-RAS) deficiency aggravates liver injury and fibrosis. Cell Death Dis 2023; 14:514. [PMID: 37563155 PMCID: PMC10415403 DOI: 10.1038/s41419-023-06029-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/18/2023] [Accepted: 07/31/2023] [Indexed: 08/12/2023]
Abstract
Progressive hepatic damage and fibrosis are major features of chronic liver diseases of different etiology, yet the underlying molecular mechanisms remain to be fully defined. N-RAS, a member of the RAS family of small guanine nucleotide-binding proteins also encompassing the highly homologous H-RAS and K-RAS isoforms, was previously reported to modulate cell death and renal fibrosis; however, its role in liver damage and fibrogenesis remains unknown. Here, we approached this question by using N-RAS deficient (N-RAS-/-) mice and two experimental models of liver injury and fibrosis, namely carbon tetrachloride (CCl4) intoxication and bile duct ligation (BDL). In wild-type (N-RAS+/+) mice both hepatotoxic procedures augmented N-RAS expression in the liver. Compared to N-RAS+/+ counterparts, N-RAS-/- mice subjected to either CCl4 or BDL showed exacerbated liver injury and fibrosis, which was associated with enhanced hepatic stellate cell (HSC) activation and leukocyte infiltration in the damaged liver. At the molecular level, after CCl4 or BDL, N-RAS-/- livers exhibited augmented expression of necroptotic death markers along with JNK1/2 hyperactivation. In line with this, N-RAS ablation in a human hepatocytic cell line resulted in enhanced activation of JNK and necroptosis mediators in response to cell death stimuli. Of note, loss of hepatic N-RAS expression was characteristic of chronic liver disease patients with fibrosis. Collectively, our study unveils a novel role for N-RAS as a negative controller of the progression of liver injury and fibrogenesis, by critically downregulating signaling pathways leading to hepatocyte necroptosis. Furthermore, it suggests that N-RAS may be of potential clinical value as prognostic biomarker of progressive fibrotic liver damage, or as a novel therapeutic target for the treatment of chronic liver disease.
Collapse
Affiliation(s)
- Kang Zheng
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of Anesthesiology, Nanjing Pukou District Hospital of Chinese Medicine Central Laboratory affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Fengjie Hao
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Sandra Medrano-Garcia
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Chaobo Chen
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- Department of General Surgery, Wuxi Xishan People's Hospital, Wuxi, China
- Department of General Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Feifei Guo
- Department of Obstetrics and Gynaecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Laura Morán-Blanco
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
| | - Sandra Rodríguez-Perales
- Molecular Cytogenetics and Genome Editing Unit, Human Cancer Genetics Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Raúl Torres-Ruiz
- Molecular Cytogenetics and Genome Editing Unit, Human Cancer Genetics Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - María Isabel Peligros
- Servicio de Anatomía Patológica Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Javier Vaquero
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Rafael Bañares
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Manuel Gómez Del Moral
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of Cell Biology, Complutense University School of Medicine, Madrid, Spain
| | - José R Regueiro
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Eduardo Martínez-Naves
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | | | - Rocío Gallego-Durán
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Douglas Maya
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Javier Ampuero
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Manuel Romero-Gómez
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Albert Gilbert-Ramos
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute, Barcelona, Spain
| | - Sergi Guixé-Muntet
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute, Barcelona, Spain
| | - Anabel Fernández-Iglesias
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute, Barcelona, Spain
| | - Jordi Gracia-Sancho
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute, Barcelona, Spain
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Mar Coll
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Laboratorio de Plasticidad de Células Hepáticas y Reparación de Tejidos, Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Isabel Graupera
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Laboratorio de Plasticidad de Células Hepáticas y Reparación de Tejidos, Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Liver Unit, Hospital Clinic, Barcelona, Spain
| | - Pere Ginès
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Liver Unit, Hospital Clinic, Barcelona, Spain
| | - Andreea Ciudin
- Endocrinology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute for Research (VHIR), Barcelona, Spain
| | - Jesús Rivera-Esteban
- Liver Unit, Internal Medicine Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute for Research (VHIR), Barcelona, Spain
| | - Juan M Pericàs
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Liver Unit, Internal Medicine Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute for Research (VHIR), Barcelona, Spain
| | - María Dolores Frutos
- Department of General and Digestive System Surgery, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - Bruno Ramos Molina
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Laboratorio de Obesidad y Metabolismo, Instituto de Investigación Biomédica de Murcia (IMIB-Arrixaca), Murcia, Spain
| | - José María Herranz
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Hepatology Programme, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Matías A Ávila
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Hepatology Programme, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Yulia A Nevzorova
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Edgar Fernández-Malavé
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain.
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain.
| |
Collapse
|
7
|
Liu C, Li N, Peng M, Huang K, Fan D, Zhao Z, Huang X, Liu Y, Chen S, Li Z. Celastrol directly binds with VAMP7 and RAB7 to inhibit autophagy and induce apoptosis in preadipocytes. Front Pharmacol 2023; 14:1094584. [PMID: 36959859 PMCID: PMC10027750 DOI: 10.3389/fphar.2023.1094584] [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: 11/10/2022] [Accepted: 02/22/2023] [Indexed: 03/09/2023] Open
Abstract
Obesity is one of the most prevalent chronic metabolic diseases, and induction of apoptosis in preadipocytes and adipocytes is a potential strategy to treat obesity. Celastrol represents one of the most robust anti-obesity phytochemicals so far, yet its direct binding target remains elusive. Here, we determined that celastrol could induce apoptosis in preadipocytes via mitochondrial mediated pathway. Further study clarified that celastrol inhibited the fusion of autophagosome and lysosome to prohibit autophagy, leading to cell apoptosis. By conducting virtual screening and genetic manipulation, we verified that overexpression of VAMP7 and RAB7 could block the effects of celastrol on inhibiting autophagy and inducing apoptosis. The Surface Plasmon Resonance study confirmed the direct binding of celastrol with VAMP7 and RAB7. The functional study illustrated the inhibition of RAB7 GTPase activity after celastrol treatment. Moreover, celastrol induced comparable apoptosis in murine epididymal adipose tissue, human preadipocytes and adipocytes, but not in human hepatocytes. An inhibitory effect on differentiation of human primary visceral preadipocytes was also observed. In conclusion, celastrol exhibited inhibitory effect of autophagy via direct binding with VAMP7 and RAB7, leading to an increase in preadipocytes apoptosis. These results advance our understanding in the potential application of celastrol in treating obesity.
Collapse
Affiliation(s)
- Chenshu Liu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Na Li
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Meixiu Peng
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Kan Huang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Dongxiao Fan
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhengde Zhao
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiuyi Huang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yunchong Liu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Sifan Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, China
- *Correspondence: Sifan Chen, ; Zilun Li,
| | - Zilun Li
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- *Correspondence: Sifan Chen, ; Zilun Li,
| |
Collapse
|
8
|
Cho SS, Yang JH, Lee JH, Baek JS, Ku SK, Cho IJ, Kim KM, Ki SH. Ferroptosis contribute to hepatic stellate cell activation and liver fibrogenesis. Free Radic Biol Med 2022; 193:620-637. [PMID: 36370962 DOI: 10.1016/j.freeradbiomed.2022.11.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022]
Abstract
Ferroptosis is a widely known regulator of cell death in connection with the redox state as a consequence of the depletion of glutathione or accumulation of lipid peroxidation. Hepatic stellate cells (HSCs) play a pivotal role in the progression of hepatic fibrosis by increasing the production and secretion of the extracellular matrix. However, the role of ferroptosis in HSC activation and liver fibrogenesis has not been clearly elucidated. The ferroptosis inducer RAS-selective lethal 3 (RSL3) or erastin treatment in HSCs caused cell death. This effect was suppressed only after exposure to ferroptosis inhibitors. We observed induction of ferroptosis by RSL3 treatment in HSCs supported by decreased glutathione peroxidase 4, glutathione deficiency, reactive oxygen species generation, or lipid peroxidation. Interestingly, RSL3 treatment upregulated the expression of plasminogen activator inhibitor-1, a representative fibrogenic marker of HSCs. In addition, treatment with ferroptosis-inducing compounds increased c-JUN phosphorylation and activator protein 1 luciferase activity but did not alter Smad phosphorylation and Smad-binding element luciferase activity. Chronic administration of iron dextran to mice causes ferroptosis of liver in vivo. The expression of fibrosis markers, such as alpha-smooth muscle actin and plasminogen activator inhibitor-1, was increased in the livers of mice with iron accumulation. Hepatic injury accompanying liver fibrosis was observed based on the levels of alanine aminotransferase, aspartate aminotransferase, and hematoxylin and eosin staining. Furthermore, we found that both isolated primary hepatocyte and HSCs undergo ferroptosis. Consistently, cirrhotic liver tissue of patients indicated glutathione peroxidase 4 downregulation in fibrotic region. In conclusion, our results suggest that ferroptosis contribute to HSC activation and the progression of hepatic fibrosis.
Collapse
Affiliation(s)
- Sam Seok Cho
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Ji Hye Yang
- College of Korean Medicine, Dongshin University, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Ji Hyun Lee
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Jin Sol Baek
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan, Gyeongsangbuk-do, 38610, Republic of Korea
| | - Il Je Cho
- College of Korean Medicine, Daegu Haany University, Gyeongsan, Gyeongsangbuk-do, 38610, Republic of Korea
| | - Kyu Min Kim
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea; Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju, 61452, Republic of Korea.
| | - Sung Hwan Ki
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
9
|
Liu P, Mao Y, Xie Y, Wei J, Yao J. Stem cells for treatment of liver fibrosis/cirrhosis: clinical progress and therapeutic potential. Stem Cell Res Ther 2022; 13:356. [PMID: 35883127 PMCID: PMC9327386 DOI: 10.1186/s13287-022-03041-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023] Open
Abstract
Cost-effective treatment strategies for liver fibrosis or cirrhosis are limited. Many clinical trials of stem cells for liver disease shown that stem cells might be a potential therapeutic approach. This review will summarize the published clinical trials of stem cells for the treatment of liver fibrosis/cirrhosis and provide the latest overview of various cell sources, cell doses, and delivery methods. We also describe the limitations and strengths of various stem cells in clinical applications. Furthermore, to clarify how stem cells play a therapeutic role in liver fibrosis, we discuss the molecular mechanisms of stem cells for treatment of liver fibrosis, including liver regeneration, immunoregulation, resistance to injury, myofibroblast repression, and extracellular matrix degradation. We provide a perspective for the prospects of future clinical implementation of stem cells.
Collapse
Affiliation(s)
- Pinyan Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yongcui Mao
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ye Xie
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Jiayun Wei
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Jia Yao
- The First Clinical Medical College of Lanzhou University, Lanzhou, China. .,Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China.
| |
Collapse
|
10
|
Wei Y, Bingyu W, Lei Y, Xingxing Y. The antifibrotic role of natural killer cells in liver fibrosis. Exp Biol Med (Maywood) 2022; 247:1235-1243. [PMID: 35475367 PMCID: PMC9379607 DOI: 10.1177/15353702221092672] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Liver fibrosis is the common pathological change of chronic liver diseases characterized by increased deposition of extracellular matrix and reduced matrix degradation. In response to liver injury caused by a variety of pathogenic agents, such as virus and alcohol, hepatic stellate cells (HSCs) are differentiated into myofibroblast-like cells and produce excessive collagens, thus resulting in fibrogenesis. Natural killer (NK) cells are the essential innate immune cells in the liver and generally control fibrosis by killing activated HSCs. This review briefly describes the fibrogenesis process and the phenotypic features of hepatic NK cells. Besides, it focuses on the antifibrotic mechanisms of NK cells and explores the potential of activating NK cells as a therapeutic strategy for the disease.
Collapse
Affiliation(s)
- Yuan Wei
- Department of Hepatology, The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha 410000, China
| | - Wang Bingyu
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin 150001, China
| | - Yang Lei
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin 150001, China
| | - Yuan Xingxing
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin 150001, China,Yuan Xingxing.
| |
Collapse
|
11
|
Shan S, Liu Z, Wang S, Liu Z, Huang Z, Yang Y, Zhang C, Song F. Drp1-mediated mitochondrial fission promotes carbon tetrachloride-induced hepatic fibrogenesis in mice. Toxicol Res (Camb) 2022; 11:486-497. [PMID: 35782650 DOI: 10.1093/toxres/tfac027] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/18/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Mitochondrial dynamics is essential for the maintenance of healthy mitochondrial network. Emerging evidence suggests that mitochondrial dysfunction is closely linked to the pathogenesis of hepatic fibrogenesis following chronic liver injury. However, the role of dynamin-related protein 1 (Drp1)-mediated mitochondrial fission in the context of liver fibrosis remains unclear.
Methods and Results
In this study, C57BL/6 mice were used to establish a model of liver fibrosis via oral gavage with CCl4 treatment for 8 weeks. Furthermore, mitochondrial fission intervention experiments were achieved by the mitochondrial division inhibitor 1 (Mdivi-1). The results demonstrated that chronic CCl4 exposure resulted in severe hepatic fibrogenesis and mitochondrial damage. By contrast, pharmacological inhibition of mitochondrial division by Mdivi-1 substantially reduced the changes of mitochondrial dynamics and finally prevented the deposition of extracellular matrix proteins. Mechanistically, excessive mitochondrial fission may activate hepatic stellate cells through RIPK1-MLKL-dependent hepatocyte death, which ultimately promotes liver fibrosis.
Conclusion
Our study imply that inhibiting Drp1-mediated mitochondrial fission attenuates CCl4-induced liver fibrosis and may serve as a therapeutic target for retarding progression of chronic liver disease.
Collapse
Affiliation(s)
- Shulin Shan
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Zhidan Liu
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Shuai Wang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Zhaoxiong Liu
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Zhengcheng Huang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Yiyu Yang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Cuiqin Zhang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Fuyong Song
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| |
Collapse
|
12
|
Neuman MG, Seitz HK, Teschke R, Malnick S, Johnson-Davis KL, Cohen LB, German A, Hohmann N, Moreira B, Moussa G, Opris M. Molecular, Viral and Clinical Features of Alcohol- and Non-Alcohol-Induced Liver Injury. Curr Issues Mol Biol 2022; 44:1294-1315. [PMID: 35723310 PMCID: PMC8947098 DOI: 10.3390/cimb44030087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/06/2022] [Accepted: 03/14/2022] [Indexed: 01/08/2023] Open
Abstract
Hepatic cells are sensitive to internal and external signals. Ethanol is one of the oldest and most widely used drugs in the world. The focus on the mechanistic engine of the alcohol-induced injury has been in the liver, which is responsible for the pathways of alcohol metabolism. Ethanol undergoes a phase I type of reaction, mainly catalyzed by the cytoplasmic enzyme, alcohol dehydrogenase (ADH), and by the microsomal ethanol-oxidizing system (MEOS). Reactive oxygen species (ROS) generated by cytochrome (CYP) 2E1 activity and MEOS contribute to ethanol-induced toxicity. We aimed to: (1) Describe the cellular, pathophysiological and clinical effects of alcohol misuse on the liver; (2) Select the biomarkers and analytical methods utilized by the clinical laboratory to assess alcohol exposure; (3) Provide therapeutic ideas to prevent/reduce alcohol-induced liver injury; (4) Provide up-to-date knowledge regarding the Corona virus and its affect on the liver; (5) Link rare diseases with alcohol consumption. The current review contributes to risk identification of patients with alcoholic, as well as non-alcoholic, liver disease and metabolic syndrome. Additional prevalence of ethnic, genetic, and viral vulnerabilities are presented.
Collapse
Affiliation(s)
- Manuela G. Neuman
- In Vitro Drug Safety and Biotechnology and the Department of Pharmacology and Toxicology, Temerity Faculty of Medicine, University of Toronto, Toronto, ON M5G 1L5, Canada; (G.M.); (M.O.)
| | - Helmut K. Seitz
- Centre of Liver and Alcohol Diseases, Ethianum Clinic and Department of Clinical Pharmacology and Pharmacoepidemiology, Faculty of Medicine, University of Heidelberg, 69115 Heidelberg, Germany; (H.K.S.); (N.H.); (B.M.)
| | - Rolf Teschke
- Department of Internal Medicine II, Division of Gastroenterology and Hepatology, Klinikum Hanau, Hanau, Academic Teaching Hospital of the Medical Faculty, Goethe University Frankfurt/Main, 60323 Frankfurt, Germany;
| | - Stephen Malnick
- Department of Internal Medicine C. Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Rehovot 76100, Israel; (S.M.); (A.G.)
| | - Kamisha L. Johnson-Davis
- Department of Pathology, University of Utah Health Sciences Centre and Division of Toxicology, ARUP Institute for Clinical and Experimental Pathology, Salt Lake City, UT 84115, USA;
| | - Lawrence B. Cohen
- Division of Gastroenterology, Sunnybrook Health Sciences Centre and Department of Medicine, Temerity Faculty of Medicine, University of Toronto, Toronto, ON M4N 3N5, Canada;
| | - Anit German
- Department of Internal Medicine C. Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Rehovot 76100, Israel; (S.M.); (A.G.)
| | - Nicolas Hohmann
- Centre of Liver and Alcohol Diseases, Ethianum Clinic and Department of Clinical Pharmacology and Pharmacoepidemiology, Faculty of Medicine, University of Heidelberg, 69115 Heidelberg, Germany; (H.K.S.); (N.H.); (B.M.)
| | - Bernhardo Moreira
- Centre of Liver and Alcohol Diseases, Ethianum Clinic and Department of Clinical Pharmacology and Pharmacoepidemiology, Faculty of Medicine, University of Heidelberg, 69115 Heidelberg, Germany; (H.K.S.); (N.H.); (B.M.)
| | - George Moussa
- In Vitro Drug Safety and Biotechnology and the Department of Pharmacology and Toxicology, Temerity Faculty of Medicine, University of Toronto, Toronto, ON M5G 1L5, Canada; (G.M.); (M.O.)
| | - Mihai Opris
- In Vitro Drug Safety and Biotechnology and the Department of Pharmacology and Toxicology, Temerity Faculty of Medicine, University of Toronto, Toronto, ON M5G 1L5, Canada; (G.M.); (M.O.)
- Family Medicine Clinic CAR, 010362 Bucharest, Romania
| |
Collapse
|
13
|
Rodríguez-Antonio I, López-Sánchez GN, Uribe M, Chávez-Tapia NC, Nuño-Lámbarri N. Role of the inflammasome, gasdermin D, and pyroptosis in non-alcoholic fatty liver disease. J Gastroenterol Hepatol 2021; 36:2720-2727. [PMID: 34050551 DOI: 10.1111/jgh.15561] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/17/2021] [Accepted: 05/26/2021] [Indexed: 12/25/2022]
Abstract
Pyroptosis is a type of programmed cell death mediated by a multiprotein complex called the inflammasome through the pro-inflammatory activity of gasdermin D. This study aimed to recognize the final biological product that leads to pore formation in the cell membrane, lysis, pro-inflammatory cytokines release, and the establishment of an immune response. An exhaustive search engine investigation of an elevated immune response can induce a sustained inflammation that directly links this mechanism to non-alcoholic fatty liver disease and its progression to non-alcoholic steatohepatitis. Clinical studies and systematic reviews suggest that gasdermin D is a critical molecule between the immune response and the disease manifestation, which could be considered a therapeutic target for highly prevalent diseases characterized by presenting perpetuated inflammatory processes. Both basic and clinical research show evidence on the expression and regulation of the inflammasome-gasdermin D-pyroptosis trinomial for the progression of non-alcoholic fatty liver disease to non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
| | | | - Misael Uribe
- Obesity and Digestive Diseases Unit, Médica Sur Clinic Foundation, Mexico City, Mexico
| | - Norberto C Chávez-Tapia
- Traslational Research Unit, Médica Sur Clinic Foundation, Mexico City, Mexico.,Obesity and Digestive Diseases Unit, Médica Sur Clinic Foundation, Mexico City, Mexico
| | | |
Collapse
|
14
|
Neuman MG, Mueller J, Mueller S. Non-invasive Biomarkers of Liver Inflammation and Cell Death in Response to Alcohol Detoxification. Front Physiol 2021; 12:678118. [PMID: 34305638 PMCID: PMC8292967 DOI: 10.3389/fphys.2021.678118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Introduction Alcohol-related liver disease (ALD) represents the most common liver disease worldwide, however, the underlying molecular mechanisms are still poorly understood. Namely centrilobular inflammation and programmed cell death are characteristic to ALD and it remains to be elucidated why they persist despite the absence of alcohol. Aims To study the effects of alcohol withdrawal in a cohort of heavy drinkers and the role of cirrhosis by using non-invasive biomarkers such as cytokines, apoptotic and angiogenic markers. Methods Caspase 3-cleaved M30, M65, cytokines (IL-6, IL-8), tumor necrosis factor alpha (TNF-α), transforming growth factor (TGF-β) and vascular endothelial growth factor (VEGF) were measured in 114 heavy drinkers. The role of alcohol detoxification was investigated in 45 patients. The liver histology was available in 23 patients. Fibrosis stage and steatosis were assessed by measuring liver stiffness (LS) and controlled attenuation parameter (CAP) in all patients using transient elastography (FibroScan, Echosens, Paris). Mean observation interval between the measurements was 5.7 ± 1.4 days (mean + -SD). Results Patients consumed a mean of 204 ± 148 g/day alcohol with a heavy drinking duration of 15.3 ± 11.0 years. Mean LS was 20.7 ± 24.4 kPa and mean CAP was 303 ± 51 dB/m. Fibrosis distribution was F0-38.1%, F1-2-31%, F3-7.1 and F4-23.9%. Apoptotic markers M30 and M65 were almost five times above normal. In contrast, TNF- α a, IL-8 and VEGF were only slightly elevated. Patients with manifest liver cirrhosis (F4) had significantly higher levels of M30, M65, IL-6 and IL-8. Histology features such as hepatocyte ballooning, Mallory-Denk bodies, inflammation and fibrosis were all significantly associated with elevated LS, and serum levels of TNF-alpha, M30 and M65 but not with CAP and other cytokines. During alcohol detoxification, LS, transaminases, TGF- β, IL-6, IL-8 and VEGF decreased significantly. In contrast, no significant changes were observed for M30, M65 and TNF- α and M30 even increased during detoxification in non-cirrhotic patients. Profibrogenic cytokine TGF-beta and pro-angiogenic cytokine VEGF showed a delayed decrease in patients with manifest cirrhosis. Conclusion Patients with alcohol-related cirrhosis have a pronounced apoptotic activity and a distinct inflammatory response that only partly improves after 1 week of alcohol detoxification. Alcohol withdrawal may represent an important approach to better dissect the underlying mechanisms in the setting of alcohol metabolism.
Collapse
Affiliation(s)
- Manuela G Neuman
- In Vitro Drug Safety and Biotechnology, Department of Pharmacology and Toxicology, Temerity Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Johannes Mueller
- Center for Alcohol Research, University of Heidelberg, Heidelberg, Germany
| | - Sebastian Mueller
- Center for Alcohol Research, University of Heidelberg, Heidelberg, Germany.,Department of Internal Medicine, Salem Medical Center, Heidelberg, Germany
| |
Collapse
|
15
|
Su S, Xiang X, Lin L, Xiong Y, Ma H, Yuan G, Zhao J, Zhang Z, Liu S, Nie D, Tang G. Cell death PET/CT imaging of rat hepatic fibrosis with 18F-labeled small molecule tracer. Nucl Med Biol 2021; 98-99:76-83. [PMID: 34062322 DOI: 10.1016/j.nucmedbio.2021.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/16/2021] [Accepted: 04/15/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE To evaluate the potential feasibility of Al[18F]F-1,4,7-triazacyclononane-1,4,7-triaceticacid (NOTA)-tripolyethylene glycol (PEG3)-Duramycin (Al[18F]F-NOTA-PEG3-Duramycin) positron emission tomography (PET) for imaging of rat hepatic fibrosis. PROCEDURES Hepatic fibrosis rat models were injected with thioacetamide (TAA), control rats received saline (n = 12 per group). Rats in the two groups underwent PET imaging using Al[18F]F-NOTA-PEG3-Duramycin and [18F]FDG at multiple time points (2, 4, 6, and 8 weeks after TAA or saline treatment). Between-group differences in the apoptosis rate, fibrotic activity, and liver uptake of Al[18F]F-NOTA-PEG3-Duramycin or [18F]FDG were assessed using Student's t-test. Imaging results were cross-validated using histopathology detection and Pearson's correlation test was used to assess the association relationships between radioactive uptake value and quantified histopathological data. RESULTS Compared with control group at multiple time points, each TAA group showed a higher radioactive liver uptake of Al[18F]F-NOTA-PEG3-Duramycin (each P < 0.05). Furthermore, the increase in the liver uptake of Al[18F]F-NOTA-PEG3-Duramycin was proportional to the progression of fibrosis (R2 = 0.8846, P < 0.001) and apoptosis rate (R2 = 0.9208, P < 0.001) in the TAA group. Meanwhile, there were also between-group differences in [18F]FDG uptake in each phase (P < 0.05), however, no relationship between [18F]FDG uptake and the fibrotic activity was observed. CONCLUSIONS Al[18F]F-NOTA-PEG3-Duramycin PET/CT could be applied to monitor the progression of liver fibrosis, whereas [18F]FDG PET/CT could not. Implications of this work for noninvasive diagnosis of liver fibrosis, assessment of fibrotic activity, and evaluation of antifibrotic therapy are expected.
Collapse
Affiliation(s)
- Shu Su
- Department of Radiology and Nuclear Medicine, Sun Yat-sen University, Guangzhou 510080, China; The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, ,Department of Medical Imaging, China.
| | - Xianhong Xiang
- Department of Interventional Radiology, Sun Yat-sen University, Guangzhou 510080, China; The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, ,Department of Medical Imaging, China.
| | - Liping Lin
- Department of Radiology and Nuclear Medicine, Sun Yat-sen University, Guangzhou 510080, China; The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, ,Department of Medical Imaging, China
| | - Ying Xiong
- Department of Radiology and Nuclear Medicine, Sun Yat-sen University, Guangzhou 510080, China; The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, ,Department of Medical Imaging, China
| | - Hui Ma
- Department of Radiology and Nuclear Medicine, Sun Yat-sen University, Guangzhou 510080, China; The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, ,Department of Medical Imaging, China
| | - Gongjun Yuan
- Department of Radiology and Nuclear Medicine, Sun Yat-sen University, Guangzhou 510080, China; The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, ,Department of Medical Imaging, China
| | - Jing Zhao
- Department of Radiology and Nuclear Medicine, Sun Yat-sen University, Guangzhou 510080, China; The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, ,Department of Medical Imaging, China
| | - Zhanwen Zhang
- Department of Radiology and Nuclear Medicine, Sun Yat-sen University, Guangzhou 510080, China; The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, ,Department of Medical Imaging, China
| | - Shaoyu Liu
- Department of Radiology and Nuclear Medicine, Sun Yat-sen University, Guangzhou 510080, China; The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, ,Department of Medical Imaging, China
| | - Dahong Nie
- Department of Radiation Oncology, Sun Yat-sen University, Guangzhou 510080, China; The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, ,Department of Medical Imaging, China.
| | - Ganghua Tang
- Department of Radiology and Nuclear Medicine, Sun Yat-sen University, Guangzhou 510080, China; The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Nanfang PET Center, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, ,Department of Medical Imaging, China.
| |
Collapse
|
16
|
Fuzheng Huayu Capsule Attenuates Hepatic Fibrosis by Inhibiting Activation of Hepatic Stellate Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3468791. [PMID: 32454856 PMCID: PMC7243025 DOI: 10.1155/2020/3468791] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 03/02/2020] [Indexed: 01/27/2023]
Abstract
Aim To investigate the mechanisms of Fuzheng Huayu (FZHY) Capsule in the treatment of hepatitis B (HBV)- associated fibrosis, HBV patients were divided into two groups, 50 cases were in the nucleotide analogues (NAs) group, while additional 50 cases were in the NAs + FZHY group. Methods We assessed the curative effects of antifibrosis through liver function, FibroScan test, and liver biopsy and detected the ratio of lymphocyte subsets by flow cytometry. Peripheral blood lymphocyte and CD8+T, CD4+T, and natural killer cell subsets collected from patients were cocultured with LX-2 cells. Activation of LX-2 cells, production of the extracellular matrix, apoptosis, and proliferation of LX-2 cells were determined. Chronic liver injury models were established by ConA treatment. Results It is evident that FZHY treatment significantly increased the percentage of NK cells, the rate of death, and apoptosis of LX-2 cells and decreased the FibroScan liver stiffness measurement value. The expressions of α-SMA and procollagen type I mRNA in LX-2 cells of the FZHY treatment group as downregulated when they were cocultured with lymphocytes compared to those from the NAs group. The proliferation of LX-2 cells in the FZHY treatment group was inhibited compared to that in the NAs group. In a mouse model of hepatic fibrosis, PBLs and IHLs from ConA exposure plus FZHY treatment inhibited the ability of JS-1 cells to express α-SMA. Conclusions FZHY Capsule improved the disordered cellular immunity and postponed liver fibrosis possibly through inhibiting the interaction between lymphocyte and hepatic stellate cells.
Collapse
|
17
|
Xie RJ, Hu XX, Zheng L, Cai S, Chen YS, Yang Y, Yang T, Han B, Yang Q. Calpain-2 activity promotes aberrant endoplasmic reticulum stress-related apoptosis in hepatocytes. World J Gastroenterol 2020; 26:1450-1462. [PMID: 32308346 PMCID: PMC7152521 DOI: 10.3748/wjg.v26.i13.1450] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/20/2020] [Accepted: 03/09/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Calpain-2 is a Ca2+-dependent cysteine protease, and high calpain-2 activity can enhance apoptosis mediated by multiple triggers.
AIM To investigate whether calpain-2 can modulate aberrant endoplasmic reticulum (ER) stress-related apoptosis in rat hepatocyte BRL-3A cells.
METHODS BRL-3A cells were treated with varying doses of dithiothreitol (DTT), and their viability and apoptosis were quantified by 3-[4, 5-dimethyl-2-thiazolyl]-2, 5-diphenyl-2-H-tetrazolium bromide and flow cytometry. The expression of ER stress- and apoptosis-related proteins was detected by Western blot analysis. The protease activity of calpain-2 was determined using a fluorescent substrate, N-succinyl-Leu-Leu-Val-Tyr-AMC. Intracellular Ca2+ content, and ER and calpain-2 co-localization were characterized by fluorescent microscopy. The impact of calpain-2 silencing by specific small interfering RNA on caspase-12 activation and apoptosis of BRL-3A cells was quantified.
RESULTS DTT exhibited dose-dependent cytotoxicity against BRL-3A cells and treatment with 2 mmol/L DTT triggered BRL-3A cell apoptosis. DTT treatment significantly upregulated 78 kDa glucose-regulated protein, activating transcription factor 4, C/EBP-homologous protein expression by >2-fold, and enhanced PRKR-like ER kinase phosphorylation, caspase-12 and caspase-3 cleavage in BRL-3A cells in a trend of time-dependence. DTT treatment also significantly increased intracellular Ca2+ content, calpain-2 expression, and activity by >2-fold in BRL-3A cells. Furthermore, immunofluorescence revealed that DTT treatment promoted the ER accumulation of calpain-2. Moreover, calpain-2 silencing to decrease calpain-2 expression by 85% significantly mitigated DTT-enhanced calpain-2 expression, caspase-12 cleavage, and apoptosis in BRL-3A cells.
CONCLUSION The data indicated that Ca2+-dependent calpain-2 activity promoted the aberrant ER stress-related apoptosis of rat hepatocytes by activating caspase-12 in the ER.
Collapse
Affiliation(s)
- Ru-Jia Xie
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Xiao-Xia Hu
- Department of Physiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Lu Zheng
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Shuang Cai
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Yu-Si Chen
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Yi Yang
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Ting Yang
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Bing Han
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Qin Yang
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| |
Collapse
|
18
|
Wu J, Lin S, Wan B, Velani B, Zhu Y. Pyroptosis in Liver Disease: New Insights into Disease Mechanisms. Aging Dis 2019; 10:1094-1108. [PMID: 31595205 PMCID: PMC6764727 DOI: 10.14336/ad.2019.0116] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/16/2019] [Indexed: 12/12/2022] Open
Abstract
There has been increasing interest in pyroptosis as a novel form of pro-inflammatory programmed cell death. The mechanism of pyroptosis is significantly different from other forms of cell death in its morphological and biochemical features. Pyroptosis is characterized by the activation of two different types of caspase enzymes-caspase-1 and caspase-4/5/11, and by the occurrence of a proinflammatory cytokine cascade and an immune response. Pyroptosis participates in the immune defense mechanisms against intracellular bacterial infections. On the other hand, excessive inflammasome activation can induce sterile inflammation and eventually cause some diseases, such as acute or chronic hepatitis and liver fibrosis. The mechanism and biological significance of this novel form of cell death in different liver diseases will be evaluated in this review. Specifically, we will focus on the role of pyroptosis in alcoholic and non-alcoholic fatty liver disease, as well as in liver failure. Finally, the therapeutic implications of pyroptosis in liver diseases will be discussed.
Collapse
Affiliation(s)
- Jiali Wu
- Liver research center of the First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| | - Su Lin
- Liver research center of the First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| | - Bo Wan
- Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, United Kingdom
| | - Bharat Velani
- Basildon and Thurrock University Hospitals NHS Foundation Trust, Nethermayne, Basildon, Essex SS16 5NL, United Kingdom
| | - Yueyong Zhu
- Liver research center of the First Affiliated Hospital of Fujian Medical University, Fujian 350005, China
| |
Collapse
|
19
|
A. Assirey E, M. Wagih H, N. Mahran H. Phoenix dactylifera L. Extract Diminished Apoptotic Effect in Cirrhotic Liver of a Rat Model. INT J PHARMACOL 2018. [DOI: 10.3923/ijp.2019.92.101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
20
|
Tardelli LP, Breda L, Marques LF, Gomes Carvalho Lima NC, Furtado de Camargo T, Scherer BR, Moreira NF, Dias JF, Dalia RA, Thomazini BF, Corezolla do Amaral ME, Alves AA. High lipid and low carbohydrate content diet, immediately after weaning, causes hepatic injury, systemic oxidative stress and diminishment of lipids in white adipose tissue. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2018. [DOI: 10.1016/j.jnim.2018.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
21
|
Cong M, Jia J, Kisseleva T, Brenner DA. The Liver's Response to Injury. ZAKIM AND BOYER'S HEPATOLOGY 2018:77-83.e5. [DOI: 10.1016/b978-0-323-37591-7.00005-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
22
|
Mueller S, Nahon P, Rausch V, Peccerella T, Silva I, Yagmur E, Straub BK, Lackner C, Seitz HK, Rufat P, Sutton A, Bantel H, Longerich T. Caspase-cleaved keratin-18 fragments increase during alcohol withdrawal and predict liver-related death in patients with alcoholic liver disease. Hepatology 2017; 66:96-107. [PMID: 28170108 DOI: 10.1002/hep.29099] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 01/30/2017] [Indexed: 12/24/2022]
Abstract
UNLABELLED Noninvasive assessment of disease activity in patients with nonalcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD) is still unsettled, but essential for the evaluation of disease progression. We here studied the association of total (M65) and caspase-cleaved (M30) serum keratin-18 fragments (n = 204) with histological parameters (n = 106) in heavy drinkers primarily admitted for alcohol withdrawal before and after alcohol detoxification. An age-, sex-, and fibrosis-stage matched NAFLD cohort (n = 30) was used for comparison. The prognostic value of M30 and M65 levels were assessed in an additional prospectively followed-up cohort of 230 patients with alcoholic cirrhosis (AC) using competing risk analyses. Among the histological parameters, both M30/65 correlated significantly and better than any other serum marker with apoptosis and liver damage, such as ballooning (r = 0.65; P < 0.001), followed by lobular inflammation (0.48; P < 0.001), steatosis (0.46; P < 0.001), but less with fibrosis (0.24; P < 0.001). Area under the receiver operating characteristics curves to detect ballooning, steatosis, or steatohepatitis (SH) were slightly better for M30 (P < 0.005). Optimal M30 cut-off values for mild and severe ballooning were 330 and 420 U/L, and 290 and 330 U/L for SH grades 1 and 2. No significant differences of M30/65 were found between the matched NAFLD and ALD cohort. In contrast to aspartate-amino-transferase and M65, M30 levels increased significantly from 391 to 518 U/L during alcohol detoxification. Moreover, levels of M30 and M65 predicted non-hepatocellular carcinoma liver-related mortality in patients with AC during a mean observation interval of 67.2 months. CONCLUSION Our data suggest M30 as highly specific marker of liver apoptosis both in ALD and NAFLD. In addition, hepatocellular apoptosis, as determined by M30 levels, occurs during alcohol withdrawal, and survival data point toward a novel underestimated role of apoptosis in patients with ALD. (Hepatology 2017;66:96-107).
Collapse
Affiliation(s)
- Sebastian Mueller
- Salem Medical Center and Center for Alcohol Research, University of Heidelberg, Heidelberg, Germany
| | - Pierre Nahon
- AP-HP, Hôpital Jean Verdier, Service d'Hépatologie, Bondy, France.,Université Paris 13, Sorbonne Paris Cité, "Equipe labellisée Ligue Contre le Cancer", F-93206, Saint-Denis, France.,Inserm, UMR-1162, "Génomique fonctionnelle des tumeur solides", Paris, France
| | - Vanessa Rausch
- Salem Medical Center and Center for Alcohol Research, University of Heidelberg, Heidelberg, Germany
| | - Tessa Peccerella
- Salem Medical Center and Center for Alcohol Research, University of Heidelberg, Heidelberg, Germany
| | - Ines Silva
- Salem Medical Center and Center for Alcohol Research, University of Heidelberg, Heidelberg, Germany
| | - Eray Yagmur
- Laboratory Diagnostics Center, RWTH-University Hospital Aachen, Aachen and Medical Care Center, Dr. Stein and colleagues, Mönchengladbach, Germany
| | - Beate K Straub
- Institute for Pathology, Universities of Mainz and Heidelberg, Germany
| | - Carolin Lackner
- Institute for Pathology, Medical University Graz, Graz, Austria
| | - Helmut K Seitz
- Salem Medical Center and Center for Alcohol Research, University of Heidelberg, Heidelberg, Germany
| | - Pierre Rufat
- APHP, Biostatistics, Pitié-Salepêtrière Hospital, Paris, France
| | - Angela Sutton
- CRB (liver disease biobank) Groupe Hospitalier Paris Seine-Saint-Denis BB-0033-00027, Paris, France.,APHP, Biochemistry Unit, Jean Verdier Hospital, Bondy, France.,INSERM U1148, and Paris 13 University, Bobigny, France
| | - Heike Bantel
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Thomas Longerich
- Institute of Pathology, RWTH-University Hospital Aachen, Aachen, Germany
| |
Collapse
|
23
|
Sarkar U, Ravindra KC, Large E, Young CL, Rivera-Burgos D, Yu J, Cirit M, Hughes DJ, Wishnok JS, Lauffenburger DA, Griffith LG, Tannenbaum SR. Integrated Assessment of Diclofenac Biotransformation, Pharmacokinetics, and Omics-Based Toxicity in a Three-Dimensional Human Liver-Immunocompetent Coculture System. Drug Metab Dispos 2017; 45:855-866. [PMID: 28450578 PMCID: PMC5469400 DOI: 10.1124/dmd.116.074005] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 04/21/2017] [Indexed: 12/13/2022] Open
Abstract
In vitro hepatocyte culture systems have inherent limitations in capturing known human drug toxicities that arise from complex immune responses. Therefore, we established and characterized a liver immunocompetent coculture model and evaluated diclofenac (DCF) metabolic profiles, in vitro-in vivo clearance correlations, toxicological responses, and acute phase responses using liquid chromatography-tandem mass spectrometry. DCF biotransformation was assessed after 48 hours of culture, and the major phase I and II metabolites were similar to the in vivo DCF metabolism profile in humans. Further characterization of secreted bile acids in the medium revealed that a glycine-conjugated bile acid was a sensitive marker of dose-dependent toxicity in this three-dimensional liver microphysiological system. Protein markers were significantly elevated in the culture medium at high micromolar doses of DCF, which were also observed previously for acute drug-induced toxicity in humans. In this immunocompetent model, lipopolysaccharide treatment evoked an inflammatory response that resulted in a marked increase in the overall number of acute phase proteins. Kupffer cell-mediated cytokine release recapitulated an in vivo proinflammatory response exemplified by a cohort of 11 cytokines that were differentially regulated after lipopolysaccharide induction, including interleukin (IL)-1β, IL-1Ra, IL-6, IL-8, IP-10, tumor necrosis factor-α, RANTES (regulated on activation normal T cell expressed and secreted), granulocyte colony-stimulating factor, macrophage colony-stimulating factor, macrophage inflammatory protein-1β, and IL-5. In summary, our findings indicate that three-dimensional liver microphysiological systems may serve as preclinical investigational platforms from the perspective of the discovery of a set of clinically relevant biomarkers including potential reactive metabolites, endogenous bile acids, excreted proteins, and cytokines to predict early drug-induced liver toxicity in humans.
Collapse
Affiliation(s)
- Ujjal Sarkar
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Kodihalli C Ravindra
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Emma Large
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Carissa L Young
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Dinelia Rivera-Burgos
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Jiajie Yu
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Murat Cirit
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - David J Hughes
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - John S Wishnok
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Douglas A Lauffenburger
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Linda G Griffith
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Steven R Tannenbaum
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| |
Collapse
|
24
|
Valdecantos MP, Pardo V, Ruiz L, Castro-Sánchez L, Lanzón B, Fernández-Millán E, García-Monzón C, Arroba AI, González-Rodríguez Á, Escrivá F, Álvarez C, Rupérez FJ, Barbas C, Konkar A, Naylor J, Hornigold D, Santos AD, Bednarek M, Grimsby J, Rondinone CM, Valverde ÁM. A novel glucagon-like peptide 1/glucagon receptor dual agonist improves steatohepatitis and liver regeneration in mice. Hepatology 2017; 65:950-968. [PMID: 27880981 DOI: 10.1002/hep.28962] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 10/11/2016] [Accepted: 11/20/2016] [Indexed: 12/14/2022]
Abstract
UNLABELLED Because nonalcoholic steatohepatitis (NASH) is associated with impaired liver regeneration, we investigated the effects of G49, a dual glucagon-like peptide-1/glucagon receptor agonist, on NASH and hepatic regeneration. C57Bl/6 mice fed chow or a methionine and choline-deficient (MCD) diet for 1 week were divided into 4 groups: control (chow diet), MCD diet, chow diet plus G49, and M+G49 (MCD diet plus G49). Mice fed a high-fat diet (HFD) for 10 weeks were divided into groups: HFD and H+G49 (HFD plus G49). Following 2 (MCD groups) or 3 (HFD groups) weeks of treatment with G49, partial hepatectomy (PH) was performed, and all mice were maintained on the same treatment schedule for 2 additional weeks. Analysis of liver function, hepatic regeneration, and comprehensive genomic and metabolic profiling were conducted. NASH was ameliorated in the M+G49 group, manifested by reduced inflammation, steatosis, oxidative stress, and apoptosis and increased mitochondrial biogenesis. G49 treatment was also associated with replenishment of intrahepatic glucose due to enhanced gluconeogenesis and reduced glucose use through the pentose phosphate cycle and oxidative metabolism. Following PH, G49 treatment increased survival, restored the cytokine-mediated priming phase, and enhanced the proliferative capacity and hepatic regeneration ratio in mice on the MCD diet. NASH markers remained decreased in M+G49 mice after PH, and glucose use was shifted to the pentose phosphate cycle and oxidative metabolism. G49 administered immediately after PH was also effective at alleviating the pathological changes induced by the MCD diet. Benefits in terms of liver regeneration were also found in mice fed HFD and treated with G49. CONCLUSION Dual-acting glucagon-like peptide-1/glucagon receptor agonists such as G49 represent a novel therapeutic approach for patients with NASH and particularly those requiring PH. (Hepatology 2017;65:950-968).
Collapse
Affiliation(s)
- M Pilar Valdecantos
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Virginia Pardo
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Ruiz
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Borja Lanzón
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo CEU, Campus Monteprincipe, Madrid, Spain
| | - Elisa Fernández-Millán
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Carmelo García-Monzón
- Liver Research Unit, Instituto de Investigación Sanitaria Princesa, University Hospital Santa Cristina, CIBERehd, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana I Arroba
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Águeda González-Rodríguez
- Liver Research Unit, Instituto de Investigación Sanitaria Princesa, University Hospital Santa Cristina, CIBERehd, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Fernando Escrivá
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Carmen Álvarez
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Francisco J Rupérez
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo CEU, Campus Monteprincipe, Madrid, Spain
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo CEU, Campus Monteprincipe, Madrid, Spain
| | | | | | | | | | | | | | | | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
25
|
Awan SJ, Baig MT, Yaqub F, Tayyeb A, Ali G. In vitro differentiated hepatic oval-like cells enhance hepatic regeneration in CCl 4 -induced hepatic injury. Cell Biol Int 2016; 41:51-61. [PMID: 27805290 DOI: 10.1002/cbin.10699] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 10/29/2016] [Indexed: 02/06/2023]
Abstract
Hepatic oval cells are likely to be activated during advanced stage of liver fibrosis to reconstruct damaged hepatic tissue. However, their scarcity, difficulties in isolation, and in vitro expansion hampered their transplantation in fibrotic liver. This study was aimed to investigate the repair potential of in vitro differentiated hepatic oval-like cells in CCl4 -induced liver fibrosis. BMSCs and oval cells were isolated and characterized from C57BL/6 GFP+ mice. BMSCs were differentiated into oval cells by preconditioning with HGF, EGF, SCF, and LIF and analyzed for the oval cells-specific genes. Efficiency of oval cells to reduce hepatocyte injury was studied by determining cell viability, release of LDH, and biochemical tests in a co-culture system. Further, in vivo repair potential of differentiated oval cells was determined in CCl4 -induced fibrotic model by gene expression analysis, biochemical tests, mason trichrome, and Sirius red staining. Differentiated oval cells expressed hepatic oval cells-specific markers AFP, ALB, CK8, CK18, CK19. These differentiated cells when co-cultured with injured hepatocytes showed significant hepato-protection as measured by reduction in apoptosis, LDH release, and improvement in liver functions. Transplantation of differentiated oval cells like cells in fibrotic livers exhibited enhanced homing, reduced liver fibrosis, and improved liver functions by augmenting hepatic microenvironment by improved liver functions. This preconditioning strategy to differentiate BMSCs into oval cell leads to improved survival and homing of transplanted cells. In addition, reduction in fibrosis and functional improvement in mice with CCl4 -induced liver fibrosis was achieved.
Collapse
Affiliation(s)
- Sana Javaid Awan
- National Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan.,Institute of Molecular Biology and Biotechnology, University of Lahore, Lahore, Pakistan
| | - Maria Tayyab Baig
- National Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Faiza Yaqub
- National Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Asima Tayyeb
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| | - Gibran Ali
- National Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
26
|
Ku NO, Strnad P, Bantel H, Omary MB. Keratins: Biomarkers and modulators of apoptotic and necrotic cell death in the liver. Hepatology 2016; 64:966-76. [PMID: 26853542 PMCID: PMC4977204 DOI: 10.1002/hep.28493] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 01/28/2016] [Accepted: 02/03/2016] [Indexed: 12/14/2022]
Abstract
UNLABELLED Keratins, formerly known as cytokeratins, are the major epithelial-specific subgroup of intermediate filament proteins. Adult hepatocytes express keratin polypeptides 8 and 18 (K8/K18), whereas cholangiocytes express K8/K18 and keratins 7 and 19 (K7/K19). Keratins function primarily to protect hepatocytes from apoptosis and necrosis, which was revealed using several genetic mouse models. This cytoprotective function was further clarified by the identification of natural human keratin variants that are normally silent, but become pathogenic by predisposing their carriers to apoptosis during acute or chronic liver injury mediated by toxins, virus infection, or metabolic stress. During apoptosis, caspases cleave K18 and K19 at conserved aspartates (human K18/K19: (235) Val-Glu-Val-Asp(↓) ) and K18 at a unique aspartate (human K18: (394) Asp-Ala-Leu-Asp(↓) ), with the latter exposed epitope becoming recognized by the M30 antibody in blood and tissues. Additional K18-containing protein backbone epitopes are detected using the M6 and M5 (termed M65) antibodies. Intact K18 and its associated fragments, which are released into blood during apoptosis and necrosis in various diseases, have been analyzed by enzyme-linked immunosorbent assay using the M30/M65 antibodies or their signal ratios. Furthermore, M30/M65 levels have been used as diagnostic and prognostic biomarkers in acute and chronic liver diseases, including nonalcoholic steatohepatitis and acute liver failure. Other keratin biomarkers include K8/K18/K19-related tissue polypeptide antigen, K18-related tissue polypeptide-specific antigen, and K19-related CYFRA-21-1, which have been evaluated mostly in patients with epithelial tumors. CONCLUSION Keratins and their fragments are released into blood during liver and other epithelial tissue injury. The epithelial specificity of K18/K19, epitope unmasking upon caspase digestion, keratin abundance, and relative keratin stability render them useful biomarkers for hepatocyte and cholangiocyte apoptosis and necrosis. However, the precise biochemical nature and release mechanism of circulating keratins remain unknown. (Hepatology 2016;64:966-976).
Collapse
Affiliation(s)
- Nam-On Ku
- Department of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul 120-749, Korea
| | - Pavel Strnad
- Department of Internal Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Heike Bantel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - M. Bishr Omary
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 7744 Medical Science II, 1301 E. Catherine Street, Ann Arbor, MI 48109-5622,Department of Medicine, University of Michigan Medical School, 7744 Medical Science II, 1301 E. Catherine Street, Ann Arbor, MI 48109-5622; and VA Ann Arbor Health Care System, Ann Arbor MI, 48105
| |
Collapse
|
27
|
Diagnostic performance of T lymphocyte subpopulations in assessment of liver fibrosis stages in hepatitis C virus patients: simple noninvasive score. Eur J Gastroenterol Hepatol 2016; 28:931-9. [PMID: 27159125 DOI: 10.1097/meg.0000000000000656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND/AIMS Evaluation of liver fibrosis in patients infected with hepatitis C virus is highly useful for the diagnosis of the disease as well as therapeutic decision. Our aim was to develop and validate a simple noninvasive score for liver fibrosis staging in chronic hepatitis C (CHC) patients and compare its performance against three published simple noninvasive indexes. MATERIALS AND METHODS CHC patients were divided into two groups: an estimated group (n=70) and a validated group (n=52). Liver fibrosis was tested in biopsies using the Metavair score system. CD4 and CD8 count/percentage were assayed by fluorescence-activated cell sorting analysis. RESULTS The multivariate discriminant analysis selects a function on the basis of absolute values of five biochemical markers: immune fibrosis index (IFI); score=3.07+3.06×CD4/CD8+0.02×α-fetoprotein (U/l)-0.07×alanine aminotransferase ratio-0.005×platelet count (10/l)-1.4×albumin (g/dl). The IFI score produced areas under curve of 0.949, 0.947, and 0.806 for differentiation of all patient categories [significant fibrosis (F2-F4), advanced fibrosis (F3-F4), and cirrhosis (F4)]. CONCLUSION The IFI score, a novel noninvasive test, can be used easily for the prediction of liver fibrosis stage in CHC patients. Our score was more efficient than aspartate aminotransferase to platelet ratio index, fibrosis index, and fibroQ and more suitable for use in Egyptian hepatitis C virus patients.
Collapse
|
28
|
β-Amyrin, a pentacyclic triterpene, exhibits anti-fibrotic, anti-inflammatory, and anti-apoptotic effects on dimethyl nitrosamine–induced hepatic fibrosis in male rats. Hum Exp Toxicol 2016; 36:113-122. [DOI: 10.1177/0960327116638727] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Hepatic fibrosis is a leading cause of morbidity and mortality worldwide. Attenuation of fibrogenic process can significantly lower the mortality rate. However, pharmaceutical intervention at fibrogenesis stage remains a major task in medicine. So there is a need for a natural compound to treat hepatic fibrosis. This study was outlined to investigate the anti-fibrotic effect of β-amyrin in dimethylnitrosamine (DMN)-induced hepatic fibrosis male rats. Serum liver function markers (aspartate transaminase, alanine transaminase, alkaline phosphatase and lactate dehydrogenase), oxidative stress markers (malondialdehyde, superoxide dismutase, catalase, glutathione peroxidase, glutathione reduced content and vitamin C), tissue inflammatory marker (tumor necrosis factor α (TNF-α)), apoptosis marker (caspase 3) and fibrolytic marker (tissue inhibitor of metalloproteinase 1 (TIMP-1)) were evaluated before and after β-amyrin treatment in DMN-induced rat. β-Amyrin treatment attenuated the altered levels of the serum enzyme markers produced by DMN and caused a subsequent recovery toward normalization. Oxidative stress markers and TNF-α levels were reduced significantly ( p < 0.001) as well as proteins’ (caspase-3 and TIMP-1) expression was reduced in β-amyrin –treated DMN rats. By virtue of β-amyrin properties of inhibiting oxidative stress, apoptosis, inflammation, and fibrogenesis, it might act as an ideal anti-inflammatory and anti-fibrogenic agent to halt the progression of liver fibrosis to chronicity.
Collapse
|
29
|
Sorafenib induces autophagic cell death and apoptosis in hepatic stellate cell through the JNK and Akt signaling pathways. Anticancer Drugs 2016; 27:192-203. [PMID: 26629768 DOI: 10.1097/cad.0000000000000316] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Increasing hepatic stellate cell (HSC) death is an attractive approach for limiting liver fibrosis. We investigated the molecular mechanisms underlying the effects of sorafenib on HSCs. LX2 cells were incubated with sorafenib and a variety of inhibitors of apoptosis, autophagy, and necrosis. Electron microscopy was used to observe autophagosomes. Inhibitors and siRNA were used to examine the role of the Akt/mTOR/p70S6K and JNK pathways. Ultrastructural analysis revealed that rat HSCs treated with 5 μmol/l sorafenib accumulated residual digested material and empty or autophagic vacuoles. Incubating LX2 cells with lysosomal protease inhibitors increased the accumulation of LC3-II, indicating that sorafenib enhances autophagic flux in HSCs. Autophagy may precede apoptosis. Lower concentrations of sorafenib and a shorter treatment time resulted in the dominance of autophagic cell death over apoptosis. Further analysis showed that Beclin 1 is inactivated by the caspases induced by sorafenib during apoptosis. Inhibition of autophagy in LX2 cells using 3-methyladenine treatment or siRNA-mediated knockdown of Atg5 resulted in a marked increase in apoptosis. Finally, sorafenib induced programmed cell death by attenuation and activation of Akt/mTOR/p70S6K and JNK signaling. Sorafenib-induced cell death is mediated by both autophagy and apoptosis. Elucidation of the signaling pathways activated by sorafenib could potentially lead to novel antifibrosis therapies for chronic liver diseases.
Collapse
|
30
|
Xie RJ, Han B, Yang T, Yang Q. Activation of Caspase12, a key molecule in endoplasmic reticulum stress related apoptosis pathway, induces apoptosis of hepatocytes in rats with hepatic fibrosis. Shijie Huaren Xiaohua Zazhi 2016; 24:2470-2477. [DOI: 10.11569/wcjd.v24.i16.2470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the change of activity of Caspase12, a key signaling molecule in endoplasmic reticulum stress related apoptosis pathway, and to explore its role in the apoptosis of hepatocytes in rats with hepatic fibrosis.
METHODS: Wistar rats were randomly divided into a 4 wk control group, an 8 wk control group, a 4 wk hepatic fibrosis group and an 8 wk hepatic fibrosis group. Rats of hepatic fibrosis groups received subcutaneous injections of 40% CCl4 twice a week at doses of 0.3 mL/100 g. Expression of GRP78, GRP94 and Caspase12 genes in hepatic tissue was detected by RT-PCR. The expression of GRP78, GRP94, procaspase12 and activated Caspase12 proteins was determined by Western blot. The apoptosis of hepatocytes was evaluated by TUNEL assay.
RESULTS: Expression of GRP78, GRP94 and Caspase12 mRNAs in the 4 wk hepatic fibrosis group was significantly higher than that in the 4 wk control group. In the 8 wk hepatic fibrosis group, expression of GRP78, GRP94 and Caspase12 mRNAs was also elevated obviously. Western blot analysis revealed that expression of GRP78, GRP94, procaspase12 and activated Caspase12 proteins was increased obviously in the liver in the 4 wk hepatic group. Compared with the 8 wk control group, expression of GRP78, GRP94, procaspase12 and activated Caspase12 proteins was elevated obviously in the 8 wk hepatic group. The apoptosis of hepatocytes in the 4 wk and 8 wk hepatic fibrosis groups was significantly higher than that in the control groups.
CONCLUSION: Activation of Caspase12, a key signaling molecule in endoplasmic reticulum stress related apoptosis pathway, may mediate the apoptosis of hepatocytes in rats with hepatic fibrosis.
Collapse
|
31
|
Yu SJ, Bae S, Kang JS, Yoon JH, Cho EJ, Lee JH, Kim YJ, Lee WJ, Kim CY, Lee HS. Hepatoprotective effect of vitamin C on lithocholic acid-induced cholestatic liver injury in Gulo(−/−) mice. Eur J Pharmacol 2015; 762:247-55. [DOI: 10.1016/j.ejphar.2015.06.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 06/01/2015] [Accepted: 06/02/2015] [Indexed: 12/21/2022]
|
32
|
He W, Shi F, Zhou ZW, Li B, Zhang K, Zhang X, Ouyang C, Zhou SF, Zhu X. A bioinformatic and mechanistic study elicits the antifibrotic effect of ursolic acid through the attenuation of oxidative stress with the involvement of ERK, PI3K/Akt, and p38 MAPK signaling pathways in human hepatic stellate cells and rat liver. Drug Des Devel Ther 2015; 9:3989-4104. [PMID: 26347199 PMCID: PMC4529259 DOI: 10.2147/dddt.s85426] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
NADPH oxidases (NOXs) are a predominant mediator of redox homeostasis in hepatic stellate cells (HSCs), and oxidative stress plays an important role in the pathogenesis of liver fibrosis. Ursolic acid (UA) is a pentacyclic triterpenoid with various pharmacological activities, but the molecular targets and underlying mechanisms for its antifibrotic effect in the liver remain elusive. This study aimed to computationally predict the molecular interactome and mechanistically investigate the antifibrotic effect of UA on oxidative stress, with a focus on NOX4 activity and cross-linked signaling pathways in human HSCs and rat liver. Drug-drug interaction via chemical-protein interactome tool, a server that can predict drug-drug interaction via chemical-protein interactome, was used to predict the molecular targets of UA, and Database for Annotation, Visualization, and Integrated Discovery was employed to analyze the signaling pathways of the predicted targets of UA. The bioinformatic data showed that there were 611 molecular proteins possibly interacting with UA and that there were over 49 functional clusters responding to UA. The subsequential benchmarking data showed that UA significantly reduced the accumulation of type I collagen in HSCs in rat liver, increased the expression level of MMP-1, but decreased the expression level of TIMP-1 in HSC-T6 cells. UA also remarkably reduced the gene expression level of type I collagen in HSC-T6 cells. Furthermore, UA remarkably attenuated oxidative stress via negative regulation of NOX4 activity and expression in HSC-T6 cells. The employment of specific chemical inhibitors, SB203580, LY294002, PD98059, and AG490, demonstrated the involvement of ERK, PI3K/Akt, and p38 MAPK signaling pathways in the regulatory effect of UA on NOX4 activity and expression. Collectively, the antifibrotic effect of UA is partially due to the oxidative stress attenuating effect through manipulating NOX4 activity and expression. The results suggest that UA may act as a promising antifibrotic agent. More studies are warranted to evaluate the safety and efficacy of UA in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Wenhua He
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Feng Shi
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Zhi-Wei Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Bimin Li
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Kunhe Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Xinhua Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Canhui Ouyang
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Xuan Zhu
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| |
Collapse
|
33
|
Abstract
Inflammation contributes to the pathogenesis of most acute and chronic liver diseases. Inflammasomes are multiprotein complexes that can sense danger signals from damaged cells and pathogens and assemble to mediate caspase-1 activation, which proteolytically activates the cytokines IL-1β and IL-18. In contrast to other inflammatory responses, inflammasome activation uniquely requires two signals to induce inflammation, therefore setting an increased threshold. IL-1β, generated upon caspase-1 activation, provides positive feed-forward stimulation for inflammatory cytokines, thereby amplifying inflammation. Inflammasome activation has been studied in different human and experimental liver diseases and has been identified as a major contributor to hepatocyte damage, immune cell activation and amplification of liver inflammation. In this Review, we discuss the different types of inflammasomes, their activation and biological functions in the context of liver injury and disease progression. Specifically, we focus on the triggers of inflammasome activation in alcoholic steatohepatitis and NASH, chronic HCV infection, ischaemia-reperfusion injury and paracetamol-induced liver injury. The application and translation of these discoveries into therapies promises novel approaches in the treatment of inflammation in liver disease.
Collapse
Affiliation(s)
- Gyongyi Szabo
- Department of Medicine, University of Massachusetts Medical School, LRB 215, 364 Plantation Street, Worcester, MA 01605, USA
| | - Jan Petrasek
- Department of Medicine, University of Massachusetts Medical School, LRB 215, 364 Plantation Street, Worcester, MA 01605, USA
| |
Collapse
|
34
|
Sekiguchi T, Umemura T, Fujimori N, Shibata S, Ichikawa Y, Kimura T, Joshita S, Komatsu M, Matsumoto A, Tanaka E, Ota M. Serum cell death biomarkers for prediction of liver fibrosis and poor prognosis in primary biliary cirrhosis. PLoS One 2015; 10:e0131658. [PMID: 26110613 PMCID: PMC4482393 DOI: 10.1371/journal.pone.0131658] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 06/04/2015] [Indexed: 12/23/2022] Open
Abstract
The development of simple, noninvasive markers of liver fibrosis is urgently needed for primary biliary cirrhosis (PBC). This study examined the ability of several serum biomarkers of cell death to estimate fibrosis and prognosis in PBC. A cohort of 130 patients with biopsy-proven PBC and 90 healthy subjects were enrolled. We assessed the utility of the M30 ELISA, which detects caspase-cleaved cytokeratin-18 (CK-18) fragments and is representative of apoptotic cell death, as well as the M65 and newly developed M65 Epideath (M65ED) ELISAs, which detect total CK-18 as indicators of overall cell death, in predicting clinically relevant fibrosis stage. All 3 cell death biomarkers were significantly higher in patients with PBC than in healthy controls and were significantly correlated with fibrosis stage. The areas under the receiver operating characteristic curve for the M65 and M65ED assays for differentiation among significant fibrosis, severe fibrosis, and cirrhosis were 0.66 and 0.76, 0.66 and 0.73, and 0.74 and 0.82, respectively. In multivariate analysis, high M65ED (hazard ratio 6.13; 95% confidence interval 1.18-31.69; P = 0.031) and severe fibrosis (hazard ratio 7.45; 95% confidence interval 1.82-30.51; P = 0.005) were independently associated with liver-related death, transplantation, or decompensation. High serum M65ED was also significantly associated with poor outcome in PBC (log-rank test; P = 0.001). Noninvasive cell death biomarkers appear to be clinically useful in predicting fibrosis in PBC. Moreover, the M65ED assay may represent a new surrogate marker of adverse disease outcome.
Collapse
Affiliation(s)
- Tomohiro Sekiguchi
- Department of Medicine, Division of Hepatology and Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takeji Umemura
- Department of Medicine, Division of Hepatology and Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
- * E-mail:
| | - Naoyuki Fujimori
- Department of Medicine, Division of Hepatology and Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Soichiro Shibata
- Department of Medicine, Division of Hepatology and Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yuki Ichikawa
- Department of Medicine, Division of Hepatology and Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takefumi Kimura
- Department of Medicine, Division of Hepatology and Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Satoru Joshita
- Department of Medicine, Division of Hepatology and Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Michiharu Komatsu
- Department of Medicine, Division of Hepatology and Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Akihiro Matsumoto
- Department of Medicine, Division of Hepatology and Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Eiji Tanaka
- Department of Medicine, Division of Hepatology and Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Masao Ota
- Department of Legal Medicine, Shinshu University Hospital, Matsumoto, Japan
| |
Collapse
|
35
|
Sun C, Sun H, Zhang C, Tian Z. NK cell receptor imbalance and NK cell dysfunction in HBV infection and hepatocellular carcinoma. Cell Mol Immunol 2015; 12:292-302. [PMID: 25308752 PMCID: PMC4654321 DOI: 10.1038/cmi.2014.91] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/25/2014] [Accepted: 08/26/2014] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is currently the third leading cause of cancer mortality and a common poor-prognosis malignancy due to postoperative recurrence and metastasis. There is a significant correlation between chronic hepatitis B virus (HBV) infection and hepatocarcinogenesis. As the first line of host defense against viral infections and tumors, natural killer (NK) cells express a large number of immune recognition receptors (NK receptors (NKRs)) to recognize ligands on hepatocytes, liver sinusoidal endothelial cells, stellate cells and Kupffer cells, which maintain the balance between immune response and immune tolerance of NK cells. Unfortunately, the percentage and absolute number of liver NK cells decrease significantly during the development and progression of HCC. The abnormal expression of NK cell receptors and dysfunction of liver NK cells contribute to the progression of chronic HBV infection and HCC and are significantly associated with poor prognosis for liver cancer. In this review, we focus on the role of NK cell receptors in anti-tumor immune responses in HCC, particularly HBV-related HCC. We discuss specifically how tumor cells evade attack from NK cells and how emerging understanding of NKRs may aid the development of novel treatments for HCC. Novel mono- and combination therapeutic strategies that target the NK cell receptor-ligand system may potentially lead to successful and effective immunotherapy in HCC.Cellular & Molecular Immunology advance online publication, 6 October 2014; doi:10.1038/cmi.2014.91.
Collapse
Affiliation(s)
- Cheng Sun
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Haoyu Sun
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Cai Zhang
- Institute of Immunopharmacology and Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Zhigang Tian
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, China
| |
Collapse
|
36
|
Fernández-Álvarez S, Juan VGD, Zubiete-Franco I, Barbier-Torres L, Lahoz A, Parés A, Luka Z, Wagner C, Lu SC, Mato JM, Martínez-Chantar ML, Beraza N. TRAIL-producing NK cells contribute to liver injury and related fibrogenesis in the context of GNMT deficiency. J Transl Med 2015; 95:223-36. [PMID: 25531568 PMCID: PMC4310762 DOI: 10.1038/labinvest.2014.151] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 10/16/2014] [Accepted: 10/27/2014] [Indexed: 12/21/2022] Open
Abstract
Glycine-N-methyltransferase (GNMT) is essential to preserve liver homeostasis. Cirrhotic patients show low expression of GNMT that is absent in hepatocellular carcinoma (HCC) samples. Accordingly, GNMT deficiency in mice leads to steatohepatitis, fibrosis, cirrhosis, and HCC. Lack of GNMT triggers NK cell activation in GNMT(-/-) mice and depletion of TRAIL significantly attenuates acute liver injury and inflammation in these animals. Chronic inflammation leads to fibrogenesis, further contributing to the progression of chronic liver injury regardless of the etiology. The aim of our study is to elucidate the implication of TRAIL-producing NK cells in the progression of chronic liver injury and fibrogenesis. For this we generated double TRAIL(-/-)/GNMT(-/-) mice in which we found that TRAIL deficiency efficiently protected the liver against chronic liver injury and fibrogenesis in the context of GNMT deficiency. Next, to better delineate the implication of TRAIL-producing NK cells during fibrogenesis we performed bile duct ligation (BDL) to GNMT(-/-) and TRAIL(-/-)/GNMT(-/-) mice. In GNMT(-/-) mice, exacerbated fibrogenic response after BDL concurred with NK1.1(+) cell activation. Importantly, specific inhibition of TRAIL-producing NK cells efficiently protected GNMT(-/-) mice from BDL-induced liver injury and fibrogenesis. Finally, TRAIL(-/-)/GNMT(-/-) mice showed significantly less fibrosis after BDL than GNMT(-/-) mice further underlining the relevance of the TRAIL/DR5 axis in mediating liver injury and fibrogenesis in GNMT(-/-) mice. Finally, in vivo silencing of DR5 efficiently protected GNMT(-/-) mice from BDL-liver injury and fibrogenesis, overall underscoring the key role of the TRAIL/DR5 axis in promoting fibrogenesis in the context of absence of GNMT. Overall, our work demonstrates that TRAIL-producing NK cells actively contribute to liver injury and further fibrogenesis in the pathological context of GNMT deficiency, a molecular scenario characteristic of chronic human liver disease.
Collapse
Affiliation(s)
- Sara Fernández-Álvarez
- Department of Metabolomics, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Virginia Gutiérrez-de Juan
- Department of Metabolomics, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Imanol Zubiete-Franco
- Department of Metabolomics, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Lucia Barbier-Torres
- Department of Metabolomics, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Agustín Lahoz
- Unidad de Hepatología, Instituto de Investigación Sanitaria La Fé, Valencia, Spain
| | - Albert Parés
- Liver Unit, Hospital Clínic, IDIBAPS, CIBERehd, University of Barcelona, Barcelona, Spain
| | - Zigmund Luka
- Department of Biochemistry, Vanderbilt University school of medicine, Nashville, TN, USA
| | - Conrad Wagner
- Department of Biochemistry, Vanderbilt University school of medicine, Nashville, TN, USA
| | - Shelly C. Lu
- Division of Gastrointestinal and Liver Diseases, USC Research Center for Liver Diseases, Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - José M. Mato
- Department of Metabolomics, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - María Luz Martínez-Chantar
- Department of Metabolomics, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain,Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), P. O. BOX 644, E-48080 Bilbao, Spain
| | - Naiara Beraza
- Department of Metabolomics, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain,Ikerbasque, Basque Foundation for Science, 48011, Bilbao, Spain,To whom correspondence should be addressed. Corresponding Author: Naiara Beraza, PhD, Department of Metabolomics, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160-Derio, Bizkaia, Spain, , Tel. 0034/944061301; Fax 0034/944061304
| |
Collapse
|
37
|
Duval F, Moreno-Cuevas JE, González-Garza MT, Rodríguez-Montalvo C, Cruz-Vega DE. Liver fibrosis and protection mechanisms action of medicinal plants targeting apoptosis of hepatocytes and hepatic stellate cells. Adv Pharmacol Sci 2014; 2014:373295. [PMID: 25505905 PMCID: PMC4258378 DOI: 10.1155/2014/373295] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 10/29/2014] [Indexed: 12/13/2022] Open
Abstract
Following chronic liver injury, hepatocytes undergo apoptosis leading to activation of hepatic stellate cells (HSC). Consequently, activated HSC proliferate and produce excessive extracellular matrix, responsible for the scar formation. The pandemic trend of obesity, combined with the high incidence of alcohol intake and viral hepatitis infections, highlights the urgent need to find accessible antifibrotic therapies. Treatment strategies should take into account the versatility of its pathogenesis and act on all the cell lines involved to reduce liver fibrosis. Medicinal plants are achieving popularity as antifibrotic agents, supported by their safety, cost-effectiveness, and versatility. This review will describe the role of hepatocytes and HSC in the pathogenesis of liver fibrosis and detail the mechanisms of modulation of apoptosis of both cell lines by twelve known hepatoprotective plants in order to reduce liver fibrosis.
Collapse
Affiliation(s)
- Florent Duval
- Catedra de Terapia Celular, Escuela de Medicina, Tecnológico de Monterrey, Avenida Morones Prieto 3000 Pte., 64710 Monterrey, NL, Mexico
| | - Jorge E. Moreno-Cuevas
- Catedra de Terapia Celular, Escuela de Medicina, Tecnológico de Monterrey, Avenida Morones Prieto 3000 Pte., 64710 Monterrey, NL, Mexico
| | - Maria Teresa González-Garza
- Catedra de Terapia Celular, Escuela de Medicina, Tecnológico de Monterrey, Avenida Morones Prieto 3000 Pte., 64710 Monterrey, NL, Mexico
| | - Carlos Rodríguez-Montalvo
- Centro de Enfermedades Hepáticas-Digestivas y Nutrición, Hospital San José, Avenida Morones Prieto 3000, 64710 Monterrey, NL, Mexico
| | - Delia Elva Cruz-Vega
- Catedra de Terapia Celular, Escuela de Medicina, Tecnológico de Monterrey, Avenida Morones Prieto 3000 Pte., 64710 Monterrey, NL, Mexico
| |
Collapse
|
38
|
Kim J, Yoon SP, Toews ML, Imig JD, Hwang SH, Hammock BD, Padanilam BJ. Pharmacological inhibition of soluble epoxide hydrolase prevents renal interstitial fibrogenesis in obstructive nephropathy. Am J Physiol Renal Physiol 2014; 308:F131-9. [PMID: 25377915 DOI: 10.1152/ajprenal.00531.2014] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Treating chronic kidney disease (CKD) has been challenging because of its pathogenic complexity. Epoxyeicosatrienoic acids (EETs) are cytochrome P-450-dependent derivatives of arachidonic acid with antihypertensive, anti-inflammatory, and profibrinolytic functions. We recently reported that genetic ablation of soluble epoxide hydrolase (sEH), an enzyme that converts EETs to less active dihydroxyeicosatrienoic acids, prevents renal tubulointerstitial fibrosis and inflammation in experimental mouse models of CKD. Here, we tested the hypothesis that pharmacological inhibition of sEH after unilateral ureteral obstruction (UUO) would attenuate tubulointerstitial fibrosis and inflammation in mouse kidneys and may provide a novel approach to manage the progression of CKD. Inhibition of sEH enhanced levels of EET regioisomers and abolished tubulointerstitial fibrosis, as demonstrated by reduced collagen deposition and myofibroblast formation after UUO. The inflammatory response was also attenuated, as demonstrated by decreased influx of neutrophils and macrophages and decreased expression of inflammatory cytokines keratinocyte chemoattractant, macrophage inflammatory protein-2, monocyte chemotactic protein-1, TNF-α, and ICAM-1 in kidneys after UUO. UUO upregulated transforming growth factor-β1/Smad3 signaling and induced NF-κB activation, oxidative stress, tubular injury, and apoptosis; in contrast, it downregulated antifibrotic factors, including peroxisome proliferator-activated receptor (PPAR) isoforms, especially PPAR-γ. sEH inhibition mitigated the aforementioned malevolent effects in UUO kidneys. These data demonstrate that pharmacological inhibition of sEH promotes anti-inflammatory and fibroprotective effects in UUO kidneys by preventing tubular injury, downregulation of NF-κB, transforming growth factor-β1/Smad3, and inflammatory signaling pathways, and activation of PPAR isoforms. Our data suggest the potential use of sEH inhibitors in treating fibrogenesis in the UUO model of CKD.
Collapse
Affiliation(s)
- Jinu Kim
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; Department of Anatomy, Jeju National University School of Medicine, Jeju, Republic of Korea; Department of Biomedicine and Drug Development, Jeju National University, Jeju, Republic of Korea
| | - Sang Pil Yoon
- Department of Anatomy, Jeju National University School of Medicine, Jeju, Republic of Korea
| | - Myron L Toews
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - John D Imig
- Department of Pharmacology and Toxicology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sung Hee Hwang
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, California; and
| | - Bruce D Hammock
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, California; and
| | - Babu J Padanilam
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; Department of Internal Medicine, Section of Nephrology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
39
|
Kim J, Imig JD, Yang J, Hammock BD, Padanilam BJ. Inhibition of soluble epoxide hydrolase prevents renal interstitial fibrosis and inflammation. Am J Physiol Renal Physiol 2014; 307:F971-80. [PMID: 25164080 PMCID: PMC4200297 DOI: 10.1152/ajprenal.00256.2014] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 08/15/2014] [Indexed: 11/22/2022] Open
Abstract
The pathophysiological events that lead to renal interstitial fibrogenesis are incompletely understood. Epoxyeicosatrienoic acid (EET), an arachidonic acid metabolite, has anti-inflammatory and profibrinolytic functions. Soluble epoxide hydrolase (sEH) converts EET to less active dihydroxyeicosatrienoic acid. Here, we tested the hypothesis that sEH deficiency would prevent tubulointerstitial fibrosis and inflammation induced by unilateral ureteral obstruction (UUO) in mouse kidneys. The loss of sEH enhanced levels of EET regioisomers and abolished tubulointerstitial fibrosis as demonstrated by reduced collagen deposition and myofibroblast formation at 3 and 10 days after UUO. The inflammatory response was prevented as demonstrated by decreased influx of neutrophil and macrophage, expression of inflammatory cytokines, and chemotactic factors in sEH-deficient UUO kidneys. Pharmacological inhibition of sEH also prevented inflammation and fibrosis after UUO. Next, we delved into the molecular mechanisms piloting the beneficial effects of sEH deficiency in renal fibrosis. UUO upregulated profibrotic factors associated with transforming growth factor (TGF)-β1/Smad3 signaling, oxidative stress, and NF-κB activation, and downregulated antifibrotic factors including peroxisome proliferator-activated receptor (PPAR) isoforms, especially PPARγ, but the loss of sEH prevented these adverse effects in UUO kidneys. Furthermore, administration of PPAR antagonists enhanced myofibroblast formation and activation of Smad3 and NF-κB p65, effects that were prevented by sEH deficiency in UUO kidneys. These data demonstrate that loss of sEH promotes anti-inflammatory and fibroprotective effects in UUO kidneys via activation of PPAR isoforms and downregulation of NF-κB, TGF-β1/Smad3, and inflammatory signaling pathways. Our data suggest the potential use of sEH inhibitors in treating fibrotic diseases.
Collapse
Affiliation(s)
- Jinu Kim
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; Department of Anatomy, Jeju National University School of Medicine, Jeju, Republic of Korea; Department of Biomedicine and Drug Development, Jeju National University, Jeju, Republic of Korea
| | - John D Imig
- Department of Pharmacology and Toxicology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jun Yang
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, California; and
| | - Bruce D Hammock
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, California; and
| | - Babu J Padanilam
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; Department of Internal Medicine, Section of Nephrology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
40
|
Elpek G&O. Cellular and molecular mechanisms in the pathogenesis of liver fibrosis: An update. World J Gastroenterol 2014; 20:7260-7276. [PMID: 24966597 PMCID: PMC4064072 DOI: 10.3748/wjg.v20.i23.7260] [Citation(s) in RCA: 285] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 02/08/2014] [Accepted: 05/23/2014] [Indexed: 02/06/2023] Open
Abstract
There have been considerable recent advances towards a better understanding of the complex cellular and molecular network underlying liver fibrogenesis. Recent data indicate that the termination of fibrogenic processes and the restoration of deficient fibrolytic pathways may allow the reversal of advanced fibrosis and even cirrhosis. Therefore, efforts have been made to better clarify the cellular and molecular mechanisms that are involved in liver fibrosis. Activation of hepatic stellate cells (HSCs) remains a central event in fibrosis, complemented by other sources of matrix-producing cells, including portal fibroblasts, fibrocytes and bone marrow-derived myofibroblasts. These cells converge in a complex interaction with neighboring cells to provoke scarring in response to persistent injury. Defining the interaction of different cell types, revealing the effects of cytokines on these cells and characterizing the regulatory mechanisms that control gene expression in activated HSCs will enable the discovery of new therapeutic targets. Moreover, the characterization of different pathways associated with different etiologies aid in the development of disease-specific therapies. This article outlines recent advances regarding the cellular and molecular mechanisms involved in liver fibrosis that may be translated into future therapies. The pathogenesis of liver fibrosis associated with alcoholic liver disease, non-alcoholic fatty liver disease and viral hepatitis are also discussed to emphasize the various mechanisms involved in liver fibrosis.
Collapse
|
41
|
Hu XX, Wang Y, Wang JXY. Handan Ganle inhibits PI3K/Akt signaling pathway in liver fibrosis in rats. Shijie Huaren Xiaohua Zazhi 2014; 22:1915-1920. [DOI: 10.11569/wcjd.v22.i14.1915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To observe the change of the PI3K/Akt signaling pathway in CCl4 induced liver fibrosis and to explore the effect of Handan Ganle on this signaling pathway in hepatic fibrosis in rats.
METHODS: Thirty SD rats were randomly divided into a normal control group, a liver fibrosis group and a Handan Ganle treated group. The rats of the liver fibrosis group and Handan Ganle treated group were treated by hypodermic injection of 40% CCl4 at 0.3 mL/100 g body weight to induce hepatic fibrosis. Then, the rats in the Handan Ganle group were treated with 1.0 g/kg Handan Ganle once daily for 8 weeks. The expression of Akt1 and phospho-Akt1 was detected by immunohistochemistry and Western blot, and the apoptosis of HSCs was determined by TUNEL assay.
RESULTS: Compared with the normal control group, the expression of Akt1 (2.73 ± 0.52 vs 9.60 ± 2.28, P < 0.01) and phospho-Akt1 (0.92 ± 0.40 vs 6.51 ± 1.39, P < 0.01) in the liver fibrosis group was increased. Handan Ganle treatment decreased the levels of Akt1 (9.60 ± 2.28 vs 5.36 ± 1.59, P < 0.01) and phospho-Akt1 (6.51 ± 1.39 vs 2.08 ± 0.85, P < 0.01) but increased the apoptosis of HSCs (1.07 ± 0.32 vs 4.24 ± 0.86, P < 0.01).
CONCLUSION: The PI3K/Akt signaling pathway may play an important role in CCl4 induced liver fibrosis. Handan Ganle can suppress this signaling pathway and increase the apoptosis of HSCs, which might be related with its anti-hepatic fibrosis activity.
Collapse
|
42
|
Deng G, Huang XJ, Luo HW, Huang FZ, Liu XY, Wang YH. Amelioration of carbon tetrachloride-induced cirrhosis and portal hypertension in rat using adenoviral gene transfer of Akt. World J Gastroenterol 2013; 19:7778-7787. [PMID: 24431897 PMCID: PMC3837279 DOI: 10.3748/wjg.v19.i43.7778] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 08/28/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether a virus constitutively expressing active Akt is useful to prevent cirrhosis induced by carbon tetrachloride (CCl4).
METHODS: Using cre-loxp technique, we created an Ad-myr-HA-Akt virus, in which Akt is labeled by a HA tag and its expression is driven by myr promoter. Further, through measuring enzyme levels and histological structure, we determined the efficacy of this Ad-myr-HA-Akt virus in inhibiting the development of cirrhosis induced by CCl4 in rats. Lastly, using western blotting, we examined the expression levels and/or phosphorylation status of Akt, apoptotic mediators, endothelial nitric oxide synthase (eNOS), and markers for hepatic stellate cells activation to understand the underlying mechanisms of protective role of this virus.
RESULTS: The Ad-myr-HA-Akt virus was confirmed using polymerase chain reaction amplification of inserted Akt gene and sequencing for full length of inserted fragment, which was consistent with the sequence reported in the GenBank. The concentrations of Ad-myr-HA-Akt and adenoviral enhanced green fluorescent protein (Ad-EGFP) virus used in the current study were 5.5 × 1011 vp/mL. The portal vein diameter, peak velocity of blood flow, portal blood flow and congestion index were significantly increased in untreated, saline and Ad-EGFP cirrhosis groups when compared to normal control after the virus was introduced to animal through tail veil injection. In contrast, these parameters in the Akt cirrhosis group were comparable to normal control group. Compared to the normal control, the liver function (Alanine aminotransferase, Aspartate aminotransferase and Albumin) was significantly impaired in the untreated, saline and Ad-EGFP cirrhosis groups. The Akt cirrhosis group showed significant improvement of liver function when compared to the untreated, saline and Ad-EGFP cirrhosis groups. The Hyp level and portal vein pressure in Akt cirrhosis groups were also significantly lower than other cirrhosis groups. The results of HE and Van Gieson staining indicated that Akt group has better preservation of histological structure and less fibrosis than other cirrhosis groups. The percentage of apoptotic cell was greatly less in Akt cirrhosis group than in other cirrhosis groups. Akt group showed positive HA tag and an increased level of phosphorylated Akt as well as decreased levels of Fas. In contrast, Caspase-3 and Caspase-9 levels in Akt group were significantly lower than other cirrhosis groups. Noticeable decrease of DR5 and α-SMA and increase of phosphorylated eNOS were observed in the Akt group when compared to other cirrhosis groups. The NO level in liver was significantly higher in Akt group than other cirrhosis groups, which was consistent with the level of phosphorylated eNOS in these groups.
CONCLUSION: This study suggest that Ad-myr-HA-Akt virus is a useful tool to prevent CCl4-induced cirrhosis in rat model and Akt pathway may be a therapeutic target for human cirrhosis.
Collapse
|
43
|
Abstract
Because of its unique function and anatomical location, the liver is exposed to a multitude of toxins and xenobiotics, including medications and alcohol, as well as to infection by hepatotropic viruses, and therefore, is highly susceptible to tissue injury. Cell death in the liver occurs mainly by apoptosis or necrosis, with apoptosis also being the physiologic route to eliminate damaged or infected cells and to maintain tissue homeostasis. Liver cells, especially hepatocytes and cholangiocytes, are particularly susceptible to death receptor-mediated apoptosis, given the ubiquitous expression of the death receptors in the organ. In a quite unique way, death receptor-induced apoptosis in these cells is mediated by both mitochondrial and lysosomal permeabilization. Signaling between the endoplasmic reticulum and the mitochondria promotes hepatocyte apoptosis in response to excessive free fatty acid generation during the metabolic syndrome. These cell death pathways are partially regulated by microRNAs. Necrosis in the liver is generally associated with acute injury (i.e., ischemia/reperfusion injury) and has been long considered an unregulated process. Recently, a new form of "programmed" necrosis (named necroptosis) has been described: the role of necroptosis in the liver has yet to be explored. However, the minimal expression of a key player in this process in the liver suggests this form of cell death may be uncommon in liver diseases. Because apoptosis is a key feature of so many diseases of the liver, therapeutic modulation of liver cell death holds promise. An updated overview of these concepts is given in this article.
Collapse
Affiliation(s)
- Maria Eugenia Guicciardi
- 1Division of Gastroenterology and Hepatology, College of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | |
Collapse
|
44
|
Gobejishvili L, Barve S, Breitkopf-Heinlein K, Li Y, Zhang J, Avila DV, Dooley S, McClain CJ. Rolipram attenuates bile duct ligation-induced liver injury in rats: a potential pathogenic role of PDE4. J Pharmacol Exp Ther 2013; 347:80-90. [PMID: 23887098 PMCID: PMC3781411 DOI: 10.1124/jpet.113.204933] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 07/18/2013] [Indexed: 12/19/2022] Open
Abstract
Anti-inflammatory and antifibrotic effects of the broad spectrum phosphodiesterase (PDE) inhibitor pentoxifylline have suggested an important role for cyclic nucleotides in the pathogenesis of hepatic fibrosis; however, studies examining the role of specific PDEs are lacking. Endotoxemia and Toll-like receptor 4 (TLR4)-mediated inflammatory and profibrotic signaling play a major role in the development of hepatic fibrosis. Because cAMP-specific PDE4 critically regulates lipopolysaccharide (LPS)-TLR4-induced inflammatory cytokine expression, its pathogenic role in bile duct ligation-induced hepatic injury and fibrogenesis in Sprague-Dawley rats was examined. Initiation of cholestatic liver injury and fibrosis was accompanied by a significant induction of PDE4A, B, and D expression and activity. Treatment with the PDE4-specific inhibitor rolipram significantly decreased liver PDE4 activity, hepatic inflammatory and profibrotic cytokine expression, injury, and fibrosis. At the cellular level, in relevance to endotoxemia and inflammatory cytokine production, PDE4B was observed to play a major regulatory role in the LPS-inducible tumor necrosis factor (TNF) production by isolated Kupffer cells. Moreover, PDE4 expression was also involved in the in vitro activation and transdifferentiation of isolated hepatic stellate cells (HSCs). Particularly, PDE4A, B, and D upregulation preceded induction of the HSC activation marker α-smooth muscle actin (α-SMA). In vitro treatment of HSCs with rolipram effectively attenuated α-SMA, collagen expression, and accompanying morphologic changes. Overall, these data strongly suggest that upregulation of PDE4 expression during cholestatic liver injury plays a potential pathogenic role in the development of inflammation, injury, and fibrosis.
Collapse
Affiliation(s)
- Leila Gobejishvili
- Department of Internal Medicine (L.G., S.B., J.Z., D.V.A., C.J.M.) and Department of Pharmacology and Toxicology (S.B., D.V.A., C.J.M.), University of Louisville, Louisville, Kentucky; Department of Surgery, University of Louisville Medical Center, Louisville, Kentucky (Y.L.); VA Medical Center, Louisville, Kentucky (C.J.M.); University of Louisville Alcohol Research Center Louisville, Kentucky (L.G., S.B., J.Z., D.V.A., C.J.M.); and Department of Medicine II, Section Molecular Hepatology-Alcohol Associated Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (K.B.-H., S.D.)
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Aldaba-Muruato LR, Moreno MG, Shibayama M, Tsutsumi V, Muriel P. Allopurinol reverses liver damage induced by chronic carbon tetrachloride treatment by decreasing oxidative stress, TGF-β production and NF-κB nuclear translocation. Pharmacology 2013; 92:138-49. [PMID: 24008378 DOI: 10.1159/000339078] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 04/13/2012] [Indexed: 12/22/2022]
Abstract
Allopurinol is an inhibitor of xanthine oxidase. The aim of this work was to evaluate the efficacy of allopurinol to reverse the experimental cirrhosis induced by CCl4. Rats received CCl4 for 8 weeks, and immediately after allopurinol was administered for 4 weeks more. Allopurinol reversed all markers of liver damage and oxidative stress to normal values, restoring the metabolic capacity of the liver. Chronic injury by CCl4 induced significant overexpression of profibrogenic cytokine TGF-β, while allopurinol decreased this production and consequently decreased the collagen content. Moreover, allopurinol is capable of partially inhibiting NF-κB. These findings suggest that allopurinol is capable of reversing the cirrhosis induced by CCl4, modulating oxidative stress, TGF-β expression and NF-κB nuclear translocation.
Collapse
|
46
|
Abstract
Fibrosis is an intrinsic response to chronic injury, maintaining organ integrity when extensive necrosis or apoptosis occurs. With protracted damage, fibrosis can progress toward excessive scarring and organ failure, as in liver cirrhosis. To date, antifibrotic treatment of fibrosis represents an unconquered area for drug development, with enormous potential but also high risks. Preclinical research has yielded numerous targets for antifibrotic agents, some of which have entered early-phase clinical studies, but progress has been hampered due to the relative lack of sensitive and specific biomarkers to measure fibrosis progression or reversal. Here we focus on antifibrotic approaches for liver that address specific cell types and functional units that orchestrate fibrotic wound healing responses and have a sound preclinical database or antifibrotic activity in early clinical trials. We also touch upon relevant clinical study endpoints, optimal study design, and developments in fibrosis imaging and biomarkers.
Collapse
Affiliation(s)
- Detlef Schuppan
- Institute of Molecular and Translational Medicine and Department of Medicine I, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany.
| | | |
Collapse
|
47
|
Feuth T, Arends JE, Fransen JH, Nanlohy NM, van Erpecum KJ, Siersema PD, Hoepelman AIM, van Baarle D. Complementary role of HCV and HIV in T-cell activation and exhaustion in HIV/HCV coinfection. PLoS One 2013; 8:e59302. [PMID: 23555014 PMCID: PMC3598709 DOI: 10.1371/journal.pone.0059302] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 02/13/2013] [Indexed: 11/18/2022] Open
Abstract
Objectives To investigate whether T-cell activation and exhaustion is linked to HCV- and HIV disease parameters in HIV/HCV infected individuals, we studied T-cell characteristics in HIV/HCV coinfected patients and controls. Methods 14 HIV/HCV coinfected, 19 HCV monoinfected, 10 HIV monoinfected patients and 15 healthy controls were included in this cross-sectional study. Differences in expression of activation and exhaustion markers (HLA-DR, CD38, PD-1, Tim-3 and Fas) and phenotypic markers on CD4+ and CD8+ T-cells were analysed by flow cytometry and were related to HCV disease parameters (HCV-viremia, ALT and liver fibrosis). Results Frequencies of activated CD4+ and CD8+ T-cells were higher in HIV/HCV-coinfected compared to healthy controls and HCV or HIV mono-infected individuals. Coinfected patients also showed high expression of the exhaustion marker PD-1 and death receptor Fas. In contrast, the exhaustion marker Tim-3 was only elevated in HIV-monoinfected patients. T-cell activation and exhaustion were correlated with HCV-RNA, suggesting that viral antigen influences T-cell activation and exhaustion. Interestingly, increased percentages of effector CD8+ T-cells were found in patients with severe (F3–F4) liver fibrosis compared to those with no to minimal fibrosis (F0–F2). Conclusions HIV/HCV coinfected patients display a high level of T-cell activation and exhaustion in the peripheral blood. Our data suggest that T-cell activation and exhaustion are influenced by the level of HCV viremia. Furthermore, high percentages of cytotoxic/effector CD8+ T-cells are associated with liver fibrosis in both HCV monoinfected and HIV/HCV coinfected patients.
Collapse
Affiliation(s)
- Thijs Feuth
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Joop E. Arends
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Justin H. Fransen
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
- Department of Immunology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Nening M. Nanlohy
- Department of Immunology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Karel J. van Erpecum
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Peter D. Siersema
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Andy I. M. Hoepelman
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Debbie van Baarle
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
- Department of Immunology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
48
|
Nilotinib induces apoptosis and autophagic cell death of activated hepatic stellate cells via inhibition of histone deacetylases. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1992-2003. [PMID: 23499874 DOI: 10.1016/j.bbamcr.2013.02.033] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 02/07/2013] [Accepted: 02/27/2013] [Indexed: 12/21/2022]
Abstract
Increasing hepatic stellate cell (HSC) death is a very attractive approach for limiting liver fibrosis. Tyrosine kinase inhibitors have been shown to have anti-fibrotic properties, but the mechanisms are poorly understood. Here, we identified the mechanism of action of the second-generation tyrosine kinase inhibitor nilotinib in inducing HSC death. Human HSC line (LX-2) and rat HSCs were treated with nilotinib and its predecessor, imatinib, in the absence or presence of various blockers, known to interfere with death signaling pathways. Nilotinib, but not imatinib, induced progressive cell death of activated, but not quiescent, HSCs in a dose-dependent manner. Activated HSCs died through apoptosis, as denoted by increased DNA fragmentation and caspase activation, and through autophagy, as indicated by the accumulation of autophagic markers, light chain (LC)3A-II and LC3B-II. Although inhibition of caspases with Z-VAD-FMK suppressed nilotinib-induced HSCs' apoptosis, there was no increase in HSCs' survival, because autophagy was exacerbated. However, blocking the mitochondrial permeability transition pore (mPTP) opening with cyclosporin A completely abolished both apoptosis and autophagy due to nilotinib. Moreover, nilotinib treatment decreased the protein expression of histone deacetylases 1, 2 and 4. Interestingly, pretreament with C646, a selective p300/CBP histone acetyl transferase inhibitor, resulted in diverting nilotinib-induced apoptosis and autophagy towards necrosis. In conclusion, the identification of mPTP as a target of nilotinib in activated HSCs suggests coordination with histone deacetylases inhibition to induce apoptosis and autophagy. Thus, our study provides novel insights into the anti-fibrotic effects of nilotinib.
Collapse
|
49
|
Wang K, Lin B. Pathophysiological Significance of Hepatic Apoptosis. ISRN HEPATOLOGY 2012; 2013:740149. [PMID: 27335822 PMCID: PMC4890876 DOI: 10.1155/2013/740149] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Accepted: 12/13/2012] [Indexed: 12/19/2022]
Abstract
Apoptosis is a classical pathological feature in liver diseases caused by various etiological factors such as drugs, viruses, alcohol, and cholestasis. Hepatic apoptosis and its deleterious effects exacerbate liver function as well as involvement in fibrosis/cirrhosis and carcinogenesis. An imbalance between apoptotic and antiapoptotic capabilities is a prominent characteristic of liver injury. The regulation of apoptosis and antiapoptosis can be a pivotal step in the treatment of liver diseases.
Collapse
Affiliation(s)
- Kewei Wang
- Departments of Surgery and Pediatrics, University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
| | - Bingliang Lin
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
50
|
Alisi A, Arciello M, Petrini S, Conti B, Missale G, Balsano C. Focal adhesion kinase (FAK) mediates the induction of pro-oncogenic and fibrogenic phenotypes in hepatitis C virus (HCV)-infected cells. PLoS One 2012; 7:e44147. [PMID: 22937161 PMCID: PMC3429423 DOI: 10.1371/journal.pone.0044147] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 08/01/2012] [Indexed: 01/18/2023] Open
Abstract
Hepatitis C Virus (HCV) infection is one of the most common etiological factors involved in fibrosis development and its progression to hepatocellular carcinoma (HCC). The pivotal role of hepatic stellate cells (HCSs) and extracellular matrix (ECM) in fibrogenesis is now certainly accepted, while the network of molecular interactions connecting HCV is emerging as a master regulator of several biological processes including proliferation, inflammation, cytoskeleton and ECM remodeling. In this study, the effects of HCV proteins expression on liver cancer cells, both pro-invasive and pro-fibrogenic phenotypes were explored. As a model of HCV infection, we used permissive Huh7.5.1 hepatoma cells infected with JFH1-derived ccHCV. Conditioned medium from these cells was used to stimulate LX-2 cells, a line of HSCs. We found that the HCV infection of Huh7.5.1 cells decreased adhesion, increased migration and caused the delocalization of alpha-actinin from plasma membrane to cytoplasm and increased expression levels of paxillin. The treatment of LX-2 cells, with conditioned medium from HCV-infected Huh7.5.1 cells, caused an increase in cell proliferation, expression of alpha-smooth muscle actin, hyaluronic acid release and apoptosis rate measured as cleaved poly ADP-ribose polymerase (PARP). These effects were accompanied in Huh7.5.1 cells by an HCV-dependent increasing of FAK activation that physically interacts with phosphorylated paxillin and alpha-actinin, and a rising of tumor necrosis factor alpha production/release. Silencing of FAK by siRNA reverted all effects of HCV infection, both those directed on Huh7.5.1 cells, and those indirect effects on the LX-2 cells. Moreover and interestingly, FAK inhibition enhances apoptosis in HCV-conditioned LX-2 cells. In conclusion, our findings demonstrate that HCV, through FAK activation, may promote cytoskeletal reorganization and a pro-oncogenic phenotype in hepatocyte-like cells, and a fibrogenic phenotype in HSCs.
Collapse
Affiliation(s)
- Anna Alisi
- Liver Research Unit, Confocal Microscopy Facility of “Bambino Gesù” Children’s Hospital, IRCCS, Rome, Italy
- * E-mail: (AA); (CB)
| | - Mario Arciello
- Laboratory of Molecular Virology and Oncology, A. Cesalpino Foundation, University of Rome “La Sapienza”, Rome, Italy
- Department of Internal Medicine, University of L’Aquila, L’Aquila, Italy
| | - Stefania Petrini
- Confocal Microscopy Facility of “Bambino Gesù” Children’s Hospital, IRCCS, Rome, Italy
| | - Beatrice Conti
- Laboratory of Molecular Virology and Oncology, A. Cesalpino Foundation, University of Rome “La Sapienza”, Rome, Italy
- Department of Internal Medicine, University of L’Aquila, L’Aquila, Italy
| | - Gabriele Missale
- Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero – Universitaria di Parma, Parma, Italy
| | - Clara Balsano
- Laboratory of Molecular Virology and Oncology, A. Cesalpino Foundation, University of Rome “La Sapienza”, Rome, Italy
- IBPM (Institute of Biology and Molecular Pathology), Rome, Italy
- * E-mail: (AA); (CB)
| |
Collapse
|