1
|
Nabekura T, Matsuo S, Shibuya A. Concanavalin-A-Induced Acute Liver Injury in Mice. Curr Protoc 2024; 4:e1117. [PMID: 39126326 DOI: 10.1002/cpz1.1117] [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] [Indexed: 08/12/2024]
Abstract
Acute liver injury is a life-threatening disease. Although immune responses are involved in the development and exacerbation of acute liver injury, the cellular and molecular mechanisms are not fully understood. Intravenous administration of the plant lectin concanavalin A (ConA) is widely used as a model of acute liver injury. ConA triggers T cell activation and cytokine production by crosslinking glycoproteins, including the T cell receptor, leading to the infiltration of myeloid cells into the liver and the subsequent amplification of inflammation in the liver. Thus, the pathogenesis of ConA-induced acute liver injury is considered a model of immune-mediated acute liver injury or autoimmune hepatitis in humans. However, the severity of the liver injury and the analyses of immune cells and non-hematopoietic cells in the liver following ConA injection are significantly influenced by the experimental conditions. This article outlines protocols for ConA-induced acute liver injury in mice and evaluation methods for liver injury, immune cells, and non-hematopoietic cells in the liver. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Induction of acute liver injury by ConA injection Basic Protocol 2: Evaluation of inflammatory cytokines in mouse plasma Basic Protocol 3: Preparation of liver sections and histological analysis of liver injury Basic Protocol 4: Preparation of liver immune cells Basic Protocol 5: Preparation of hepatocytes, endothelial cells, and hepatic stellate cells Basic Protocol 6: Flow cytometry of immune and non-hematopoietic liver cells Basic Protocol 7: Flow cytometric sorting of endothelial cells and hepatic stellate cells Basic Protocol 8: Quantitative reverse transcription polymerase chain reaction.
Collapse
Affiliation(s)
- Tsukasa Nabekura
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Aichi, Japan
| | - Soichi Matsuo
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Doctoral Program in Medical Science, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Advanced Medical Technologies, National Cerebral and Vascular Cancer Center Research Institute, Suita, Osaka, Japan
| | - Akira Shibuya
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
2
|
Azuma I, Mizuno T, Morita K, Suzuki Y, Kusuhara H. Investigation of the usefulness of liver-specific deconvolution method by establishing a liver benchmark dataset. NAR Genom Bioinform 2024; 6:lqad111. [PMID: 38187088 PMCID: PMC10768887 DOI: 10.1093/nargab/lqad111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/31/2023] [Accepted: 12/16/2023] [Indexed: 01/09/2024] Open
Abstract
Immune responses in the liver are related to the development and progression of liver failure, and precise prediction of their behavior is important. Deconvolution is a methodology for estimating the immune cell proportions from the transcriptome, and it is mainly applied to blood-derived samples and tumor tissues. However, the influence of tissue-specific modeling on the estimation results has rarely been investigated. Here, we constructed a system to evaluate the performance of the deconvolution method on liver transcriptome data. We prepared seven mouse liver injury models using small-molecule compounds and established a benchmark dataset with corresponding liver bulk RNA-Seq and immune cell proportions. RNA-Seq expression for nine leukocyte subsets and four liver-associated cell types were obtained from the Gene Expression Omnibus to provide a reference. We found that the combination of reference cell sets affects the estimation results of reference-based deconvolution methods and established a liver-specific deconvolution by optimizing the reference cell set for each cell to be estimated. We applied this model to independent datasets and showed that liver-specific modeling is highly extrapolatable. We expect that this approach will enable sophisticated estimation from rich tissue data accumulated in public databases and to obtain information on aggregated immune cell trafficking.
Collapse
Affiliation(s)
- Iori Azuma
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
| | - Tadahaya Mizuno
- To whom correspondence should be addressed. Tel: +81 3 5841 4771; Fax: +81 3 5841 4766;
| | - Katsuhisa Morita
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
| | - Yutaka Suzuki
- Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8561, Japan
| | - Hiroyuki Kusuhara
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
| |
Collapse
|
3
|
Haikal A, Galala AA, Elshal M, Amen Y, Gohar AA. Bioactivity of Eriocephalus africanus essential oil against concanavalin A-induced hepatitis via suppressing immune cell infiltration, inhibiting TNF-α/NF-κB and IFN-γ/STAT1 signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117000. [PMID: 37544345 DOI: 10.1016/j.jep.2023.117000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 08/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Eriocephalus africanus infusion is used as a diuretic and a diaphoretic and is also used in the treatment of gastrointestinal disorders and gynaecological conditions, inflammation and dermal disorders, asthma, coughs, fevers, and painful ailments. The plant has been used traditionally as a medication to cure inflammation and skin problems. AIM OF THE STUDY Studying E. africanus essential oil (EAEO) as a potential hepatoprotective measure against concanavalin (Con) A-induced hepatitis in mice and investigating its underlying mechanism. MATERIALS AND METHODS Hydro-distilled oil of the fresh plant aerial shoots is subjected to GC/MS analysis. Autoimmune hepatitis (AIH) was induced in mice by intravenous injection of Con A (15 mg/kg). EAEO was administered orally before Con A injection to test its hepatoprotective activity. RESULTS GC/MS analysis revealed the presence of 22 compounds representing 99.43% of the oil components. The monoterpene artemisia ketone (41.02%) and the sesquiterpene juniper camphor (14.17%) are the major components. The in vivo study showed that the oil suppressed Con A-induced neutrophil and CD4+T cell infiltration into the liver, restored hepatic redox balance, inhibited Con A-induced elevation of tumor necrosis factor-alpha (TNF-α), interleukin (IL-6), and interferon-gamma (IFN-γ) hepatic levels which were correlated with its ability to suppress nuclear factor kappa B (NF-κB) and Signal Transducer and Activator of Transcription (STAT1) activation in the liver. CONCLUSION EAEO showed hepatoprotective potential against Con A-induced hepatitis in mice collectively through selective anti-oxidant, anti-inflammatory, and anti-necrotic effects.
Collapse
Affiliation(s)
- Abdullah Haikal
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Amal A Galala
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Mahmoud Elshal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Yhiya Amen
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Ahmed A Gohar
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
4
|
Matsuo S, Nabekura T, Matsuda K, Shibuya K, Shibuya A. DNAM-1 Immunoreceptor Protects Mice from Concanavalin A-Induced Acute Liver Injury by Reducing Neutrophil Infiltration. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:954-963. [PMID: 37522739 DOI: 10.4049/jimmunol.2200705] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 07/13/2023] [Indexed: 08/01/2023]
Abstract
DNAX accessory molecule-1 (DNAM-1; CD226) is an activating immunoreceptor on T cells and NK cells. The interaction of DNAM-1 with its ligand CD155 expressed on hematopoietic and nonhematopoietic cells plays an important role in innate and adaptive immune responses. In this study, we investigated the role of the DNAM-1-CD155 axis in the pathogenesis of T cell-mediated Con A-induced acute liver injury. Unexpectedly, DNAM-1-deficient (Cd226-/-) mice exhibited more severe acute liver injury and higher concentrations of IL-6 and TNF-α than did wild-type (WT) mice after Con A injection. We found that a larger number of neutrophils infiltrated into the liver of Cd226-/- mice compared with WT mice after Con A injection. Depletion of neutrophils ameliorated liver injury and decreased IL-6 and TNF-α in Cd226-/- mice after Con A injection, suggesting that neutrophils exacerbate the liver injury in Cd226-/- mice. Hepatocytes produced more significant amounts of CXCL1, a chemoattractant for neutrophils, in Cd226-/- mice than in WT mice after Con A injection. In the coculture of hepatocytes with liver lymphocytes, either DNAM-1 deficiency in liver lymphocytes or CD155 deficiency in hepatocytes promoted CXCL1 production by hepatocytes. These results suggest that the interaction of DNAM-1 with CD155 inhibits CXCL1 production by hepatocytes, leading to ameliorating acute liver injury.
Collapse
Affiliation(s)
- Soichi Matsuo
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Doctoral Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Tsukasa Nabekura
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kenshiro Matsuda
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuko Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
5
|
Inoue A, Chiba S, Eto S, Taniguchi T, Yanai H. Potential of HMGB-inhibitory oligodeoxynucleotide ISM ODN to neutrophil recruitment in mouse model of hepatitis. Genes Cells 2023; 28:202-210. [PMID: 36550748 DOI: 10.1111/gtc.13002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/25/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
High-mobility group box 1 (HMGB1) is a nucleotide-binding chromatin protein that has also been characterized as a prototypical damage-associate molecular pattern. It triggers inflammatory responses upon release from damaged or dying cells. In fact, HMGB1 has been linked to the induction of many inflammatory diseases through immune cell activation including neutrophil recruitment. In this study, we examined the impact of HMGB1-binding inhibitory oligodeoxynucleotide (ISM ODN) on the development of hepatitis using a murine model of the disease. Our results indicate that ISM ODN effectively suppresses pathological features of hepatitis, including neutrophil accumulation. This study therefore may offer clinical insight into the treatment of hepatitis and possibly other inflammatory diseases.
Collapse
Affiliation(s)
- Asuka Inoue
- Department of Inflammology, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| | - Shiho Chiba
- Department of Inflammology, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| | - Shotaro Eto
- Department of Inflammology, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| | - Tadatsugu Taniguchi
- Department of Inflammology, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| | - Hideyuki Yanai
- Department of Inflammology, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Chen CL, Tseng PC, Satria RD, Nguyen TT, Tsai CC, Lin CF. Role of Glycogen Synthase Kinase-3 in Interferon-γ-Mediated Immune Hepatitis. Int J Mol Sci 2022; 23:ijms23094669. [PMID: 35563060 PMCID: PMC9101719 DOI: 10.3390/ijms23094669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/16/2022] [Accepted: 04/20/2022] [Indexed: 12/04/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK-3), a serine/threonine kinase, is a vital glycogen synthase regulator controlling glycogen synthesis, glucose metabolism, and insulin signaling. GSK-3 is widely expressed in different types of cells, and its abundant roles in cellular bioregulation have been speculated. Abnormal GSK-3 activation and inactivation may affect its original bioactivity. Moreover, active and inactive GSK-3 can regulate several cytosolic factors and modulate their diverse cellular functional roles. Studies in experimental liver disease models have illustrated the possible pathological role of GSK-3 in facilitating acute hepatic injury. Pharmacologically targeting GSK-3 is therefore suggested as a therapeutic strategy for liver protection. Furthermore, while the signaling transduction of GSK-3 facilitates proinflammatory interferon (IFN)-γ in vitro and in vivo, the blockade of GSK-3 can be protective, as shown by an IFN-γ-induced immune hepatitis model. In this study, we explored the possible regulation of GSK-3 and the potential relevance of GSK-3 blockade in IFN-γ-mediated immune hepatitis.
Collapse
Affiliation(s)
- Chia-Ling Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Po-Chun Tseng
- Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei 110, Taiwan;
| | - Rahmat Dani Satria
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (R.D.S.); (T.T.N.)
- Department of Clinical Pathology and Laboratory Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Clinical Laboratory Installation, Dr. Sardjito Central General Hospital, Yogyakarta 55281, Indonesia
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Thi Thuy Nguyen
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (R.D.S.); (T.T.N.)
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Oncology, Hue University of Medicine and Pharmacy, Hue University, Hue City 530000, Vietnam
| | - Cheng-Chieh Tsai
- Department of Nursing, Chung Hwa University of Medical Technology, Tainan 703, Taiwan
- Department of Long Term Care Management, Chung Hwa University of Medical Technology, Tainan 703, Taiwan
- Correspondence: (C.-C.T.); (C.-F.L.)
| | - Chiou-Feng Lin
- Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei 110, Taiwan;
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (R.D.S.); (T.T.N.)
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: (C.-C.T.); (C.-F.L.)
| |
Collapse
|
7
|
Ilyinskii PO, Roy CJ, LePrevost J, Rizzo GL, Kishimoto TK. Enhancement of the Tolerogenic Phenotype in the Liver by ImmTOR Nanoparticles. Front Immunol 2021; 12:637469. [PMID: 34113339 PMCID: PMC8186318 DOI: 10.3389/fimmu.2021.637469] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
ImmTOR biodegradable nanoparticles encapsulating rapamycin have been shown to induce a durable tolerogenic immune response to co-administered biologics and gene therapy vectors. Prior mechanism of action studies have demonstrated selective biodistribution of ImmTOR to the spleen and liver following intravenous (IV) administration. In the spleen, ImmTOR has been shown to induce tolerogenic dendritic cells and antigen-specific regulatory T cells and inhibit antigen-specific B cell activation. Splenectomy of mice resulted in partial but incomplete abrogation of the tolerogenic immune response induced by ImmTOR. Here we investigated the ability of ImmTOR to enhance the tolerogenic environment in the liver. All the major resident populations of liver cells, including liver sinusoidal endothelial cells (LSECs), Kupffer cells (KC), stellate cells (SC), and hepatocytes, actively took up fluorescent-labeled ImmTOR particles, which resulted in downregulation of MHC class II and co-stimulatory molecules and upregulation of the PD-L1 checkpoint molecule. The LSEC, known to play an important role in hepatic tolerance induction, emerged as a key target cell for ImmTOR. LSEC isolated from ImmTOR treated mice inhibited antigen-specific activation of ovalbumin-specific OT-II T cells. The tolerogenic environment led to a multi-pronged modulation of hepatic T cell populations, resulting in an increase in T cells with a regulatory phenotype, upregulation of PD-1 on CD4+ and CD8+ T cells, and the emergence of a large population of CD4–CD8– (double negative) T cells. ImmTOR treatment protected mice in a concanavalin A-induced model of acute hepatitis, as evidenced by reduced production of inflammatory cytokines, infiltrate of activated leukocytes, and tissue necrosis. Modulation of T cell phenotype was seen to a lesser extent after administration by empty nanoparticles, but not free rapamycin. The upregulation of PD-1, but not the appearance of double negative T cells, was inhibited by antibodies against PD-L1 or CTLA-4. These results suggest that the liver may contribute to the tolerogenic properties of ImmTOR treatment.
Collapse
Affiliation(s)
| | | | | | - Gina L Rizzo
- Selecta Biosciences, Watertown, MA, United States
| | | |
Collapse
|
8
|
Almishri W, Shaheen AA, Sharkey KA, Swain MG. The Antidepressant Mirtazapine Inhibits Hepatic Innate Immune Networks to Attenuate Immune-Mediated Liver Injury in Mice. Front Immunol 2019; 10:803. [PMID: 31031775 PMCID: PMC6474187 DOI: 10.3389/fimmu.2019.00803] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 03/26/2019] [Indexed: 12/11/2022] Open
Abstract
Activation of the innate immune system, including tissue macrophages and associated neutrophil infiltration, is an important driver of subsequent adaptive immune responses in many autoimmune diseases, including autoimmune hepatitis (AIH). The antidepressant mirtazapine has a unique complex pharmacology, altering signaling through a number of serotonin and histamine receptors that can impact macrophage function; an effect potentially influencing AIH outcome. In the mouse model of concanavalin A (Con A) induced liver injury (mimics many aspects of human AIH), in which early innate immune activation (i.e., stimulated hepatic macrophages/monocytes recruit neutrophils and additional monocytes to the liver) critically drives immune-mediated hepatitis induction, mirtazapine strikingly and dose-dependently inhibited Con A-induced liver injury. This inflammation-suppressing effect of mirtazapine was linked to an attenuation of Con A-stimulated early innate immune responses within the liver, including inhibition of hepatic macrophage/monocyte activation, decreased hepatic macrophage/monocyte-derived pro-inflammatory cytokine (e.g., TNFα) and chemokine (e.g., CXCL1 and CXCL2) production, suppression of Con A-induced increases in the hepatic expression of the neutrophil relevant endothelial cell adhesion molecule ICAM-1, with the resultant significant reduction in neutrophil recruitment into the liver. Consistent with our findings in the Con A model, mirtazapine also significantly reduced activation-induced release of cytokine/chemokine mediators from human CD14+ monocytes in vitro. Conclusion: Our data suggest that mirtazapine can attenuate hepatic innate immune responses that critically regulate the subsequent development of autoimmune liver injury. Therefore, given that it is a safe and widely used medication, mirtazapine may represent a novel therapeutic approach to autoimmune liver disease.
Collapse
Affiliation(s)
- Wagdi Almishri
- Liver Unit, Snyder Institute for Chronic Disease, University of Calgary, Calgary, AB, Canada
| | - Abdel Aziz Shaheen
- Liver Unit, Snyder Institute for Chronic Disease, University of Calgary, Calgary, AB, Canada
| | - Keith A. Sharkey
- Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Mark G. Swain
- Liver Unit, Snyder Institute for Chronic Disease, University of Calgary, Calgary, AB, Canada
- *Correspondence: Mark G. Swain
| |
Collapse
|
9
|
Hazem SH, Hamed MF, Saad MAA, Gameil NM. Comparison of lactate and β-hydroxybutyrate in the treatment of concanavalin-A induced hepatitis. Int Immunopharmacol 2018; 61:376-384. [DOI: 10.1016/j.intimp.2018.06.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/01/2018] [Accepted: 06/15/2018] [Indexed: 01/21/2023]
|
10
|
Ko YE, Yoon SY, Ly SY, Kim JH, Sohn KY, Kim JW. 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) reduces hepatic injury in concanavalin A-treated mice. J Cell Biochem 2017; 119:1392-1405. [PMID: 28749086 DOI: 10.1002/jcb.26299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 07/18/2017] [Indexed: 11/10/2022]
Abstract
1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG), a chemically synthesized monoacetyldiaglyceride, is one of the constituents in Sika deer antlers and has been known traditionally as having immunomodulatory effects. However, the mechanism by which PLAG controls neutrophil migration, which evokes liver injury in the hepatitis animal model, remains largely unknown. This study was designed to evaluate the immunomodulatory effects of PLAG on cytokine secretion and neutrophil migration in vivo and in vitro. Concanavalin A (Con A) induced leukocyte infiltration in the liver and increased plasma cytokine levels. Pretreatment with PLAG reduced the levels of interleukin (IL)-4, IL-6, IL-10, and CXCL2, but maintained interferon (IFN)-γ levels and modulated neutrophil recruitment toward the liver. Furthermore, the mRNA and protein levels of IL-4 and CXCL2 in liver tissue were also decreased in the Con A-treated mice. Liver histology analyses showed that PLAG reduced Con A-induced hepatic necrosis, which was accompanied by leukocyte infiltration. The in vitro studies revealed that PLAG reduced IL-4 secretion in Con A stimulated T cell and blocked signal transducer and activator of transcription 6 (STAT6) Con A induced hepatocyte. PLAG attenuated IL-4 induced activation of atypical protein kinase C (PKC)/STAT6 in hepatocytes and inhibited neutrophil migration toward the liver tissue through suppression of IL-8/vascular cell adhesion molecule (VCAM) expression. These results suggest that PLAG could mitigate excess neutrophil migration into liver tissue and potentially have a therapeutic effect on immune-mediated liver injury.
Collapse
Affiliation(s)
- Young E Ko
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,Department of Food and Nutrition, Chungnam National University, Daejeon, Republic of Korea
| | - Sun Y Yoon
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Daejeon, Korea
| | - Sun Y Ly
- Department of Food and Nutrition, Chungnam National University, Daejeon, Republic of Korea
| | - Joo H Kim
- Department of Pathology, EulJi University School of Medicine, Daejeon, Republic of Korea
| | - Ki Y Sohn
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Daejeon, Korea
| | - Jae W Kim
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| |
Collapse
|
11
|
Hsu MC, Liu SH, Wang CW, Hu NY, Wu ES, Shih YC, Chiu PJ. JKB-122 is effective, alone or in combination with prednisolone in Con A-induced hepatitis. Eur J Pharmacol 2017; 812:113-120. [DOI: 10.1016/j.ejphar.2017.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 12/28/2022]
|
12
|
Rani R, Tandon A, Wang J, Kumar S, Gandhi CR. Stellate Cells Orchestrate Concanavalin A-Induced Acute Liver Damage. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2008-2019. [PMID: 28710903 DOI: 10.1016/j.ajpath.2017.05.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/13/2017] [Accepted: 05/22/2017] [Indexed: 12/12/2022]
Abstract
Concanavalin A (ConA) causes immune cell-mediated liver damage, but the contribution of resident nonparenchymal cells (NPCs) is also evident. Hepatic stellate cells (HSCs) induce hepatic inflammation and immunological reactions; we therefore investigated their role in ConA-induced liver injury. ConA was administered i.v. to control or HSC-depleted mice; hepatic histopathology and cytokines/chemokines were determined after 6 hours. In vitro, effects of ConA-conditioned HSC medium on hepatocytes were determined. ConA induced inflammation, sinusoidal congestion, and extensive midzonal hepatocyte death in control mice, which were strongly minimized in HSC-depleted mice. CD4 and natural killer T cells and neutrophils were markedly reduced in ConA-treated HSC-depleted mice compared with control mice. The increase in cytokines/chemokines of hepatic injury was much higher in ConA-treated control mice than in HSC-depleted mice. ConA-treated HSCs showed increased expression of interferon-β, tumor necrosis factor-α, and CXCL1, induced oxidative stress in hepatocytes, and caused hepatocyte apoptosis. ConA induced nuclear translocation of interferon-regulatory factor-1 (IRF1) in hepatocytes in vivo, and ConA/HSC induced a similar effect in cultured hepatocytes. IRF1-knockout mice were resistant to ConA-induced liver damage, and anti-interferon β antibody mitigated ConA/HSC-induced injury. In HSC-NPC co-culture, ConA-induced expression of inflammatory cytokines/chemokines was significantly augmented compared with NPCs alone. HSCs play an essential role in ConA-induced liver injury directly via the interferon-β/IRF1 axis, and by modulating properties of NPCs.
Collapse
Affiliation(s)
- Richa Rani
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio; Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ashish Tandon
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Jiang Wang
- Department of Pathology, University of Cincinnati, Cincinnati, Ohio
| | - Sudhir Kumar
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio; Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Chandrashekhar R Gandhi
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio; Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| |
Collapse
|
13
|
Kodama S, Shimura T, Kuribayashi H, Abe T, Yoshinari K. Pregnenolone 16α-carbonitrile ameliorates concanavalin A-induced liver injury in mice independent of the nuclear receptor PXR activation. Toxicol Lett 2017; 271:58-65. [PMID: 28237809 DOI: 10.1016/j.toxlet.2017.02.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/02/2017] [Accepted: 02/20/2017] [Indexed: 12/19/2022]
Abstract
The pregnane X receptor (PXR) is well-known as a key regulator of drug/xenobiotic clearance. Upon activation by ligand, PXR transcriptionally upregulates the expression of drug-metabolizing enzymes and drug transporters. Recent studies have revealed that PXR also plays a role in regulating immune/inflammatory responses. Specific PXR activators, including synthetic ligands and phytochemicals, have been shown to ameliorate chemically induced colitis in mice. In this study, we investigated an anti-inflammatory effect of pregnenolone 16α-carbonitrile (PCN), a prototypical activator for rodent PXR, in concanavalin A (Con A)-induced liver injury, a model of immune-mediated liver injury, using wild-type and Pxr-/- mice. Unexpectedly, pretreatment with PCN significantly ameliorated Con A-induced liver injury in not only wild-type but Pxr-/- mice as well, accompanied with lowered plasma ALT levels and histological improvements. Pretreatment with PCN was found to significantly repress the induction of Cxcl2 and Ccl2 mRNA expression and neutrophil infiltration into the liver of both wild-type and Pxr-/- mice at the early time point of Con A-induced liver injury. Our results indicate that PCN has unexpected immunosuppressive activity independent of PXR activation to protect mice from immune-mediated liver injury induced by Con A.
Collapse
Affiliation(s)
- Susumu Kodama
- Division of Drug Metabolism and Molecular Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan; Laboratory of Pharmacotherapy of Life-Style Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan.
| | - Takuto Shimura
- Division of Drug Metabolism and Molecular Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Hideaki Kuribayashi
- Division of Drug Metabolism and Molecular Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Taiki Abe
- Division of Drug Metabolism and Molecular Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Kouichi Yoshinari
- Division of Drug Metabolism and Molecular Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan; Department of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| |
Collapse
|
14
|
Ishiguro T, Fukawa T, Akaki K, Nagaoka K, Takeda T, Iwakura Y, Inaba K, Takahara K. Absence of DCIR1 reduces the mortality rate of endotoxemic hepatitis in mice. Eur J Immunol 2017; 47:704-712. [PMID: 28127756 DOI: 10.1002/eji.201646814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/06/2017] [Accepted: 01/23/2017] [Indexed: 01/23/2023]
Abstract
Dendritic cell immunoreceptor (DCIR) is a C-type lectin with an immunoreceptor tyrosine-based inhibitory motif (ITIM). Mice lacking DCIR1 (Dcir1-/- mice) show higher susceptibility to chronic arthritis with increasing age, suggesting that DCIR1 is involved in immune modulation via its ITIM. However, the role of DCIR1 in acute immune responses is not clear. In this study, we explored its role in acute experimental hepatitis. Upon injection of d-galactosamine and lipopolysaccharide, Dcir1-/- mice showed decreased mortality rates and serum levels of alanine aminotransferase. In early onset hepatitis, serum levels of TNF-α, which primarily cause inflammation and hepatocyte apoptosis, were significantly lower in Dcir1-/- mice than in WT mice. In the liver of Dcir1-/- mice, influx of neutrophils and other leukocytes decreased. Consistently, the levels of neutrophil-chemoattractant chemokine CXCL1/KC, but not CXCL2/MIP-2, were lower in Dcir1-/- mice than in WT mice. However, chemotaxis of Dcir1-/- neutrophils to CXCL1/KC appeared normal. Pervanadate treatment induced binding of DCIR1 and Src homology region 2 domain-containing phosphatase (SHP)-2, possibly leading to CXCL1/KC expression. These results suggest that DCIR1 is involved in exacerbation of endotoxemic hepatitis, providing a new therapeutic target for lethal hepatitis.
Collapse
Affiliation(s)
- Toshifumi Ishiguro
- Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe, Sakyo, Kyoto, Japan
| | - Tetsuya Fukawa
- Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe, Sakyo, Kyoto, Japan
| | - Kotaro Akaki
- Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe, Sakyo, Kyoto, Japan
| | - Koji Nagaoka
- Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe, Sakyo, Kyoto, Japan
| | - Tatsuki Takeda
- Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe, Sakyo, Kyoto, Japan
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Kayo Inaba
- Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe, Sakyo, Kyoto, Japan
| | - Kazuhiko Takahara
- Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe, Sakyo, Kyoto, Japan
| |
Collapse
|
15
|
Fei M, Xie Q, Zou Y, He R, Zhang Y, Wang J, Bo L, Li J, Deng X. Alpha-lipoic acid protects mice against concanavalin A-induced hepatitis by modulating cytokine secretion and reducing reactive oxygen species generation. Int Immunopharmacol 2016; 35:53-60. [PMID: 27018751 DOI: 10.1016/j.intimp.2016.03.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/01/2016] [Accepted: 03/18/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Alpha-lipoic acid (α-LA), which exits in almost all types of prokaryotic and eukaryotic cells, is a key regulator of energy metabolism in mitochondria. This study was designed to explore the protective effect of α-LA against concanavalin A (Con A)-induced hepatitis in mice and explore the potential mechanism. METHODS Acute autoimmune hepatitis was induced by intravenous (IV) injection of Con A (15mg/kg) in C57BL/6 mice. α-LA (100mg/kg) was administered four days before Con A injection. Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and histopathological change of the liver tissue were measured. Serum cytokine TNF-α, IL-6, IFN-γ and IL-10 were detected by ELISA. The mRNA levels of these inflammatory cytokines in the liver were detected by RT-PCR. Malondialdehyde (MDA), myeloperoxidase (MPO), superoxide dismutase (SOD) and reduced/oxidized glutathione (GSH/GSSG) in liver were determined using commercial kits. Phosphorylated NF-κB p65, IκBα and phosphorylated MAPK were measured by Western blot. RESULTS Con A injection induced severe immune responses and extensive hepatocellular apoptosis within 12h. Pretreatment of α-LA markedly reduced the serum ALT and AST activity and the increase of plasma TNF-α, IL-6, IFN-γ and IL-10. In addition, α-LA pretreatment decreased the tissue MPO activity and lipid peroxidation, but increased SOD and GSH levels. α-LA inhibited the phosphorylation of NF-κB p65, IκBα and JNK. CONCLUSION Pretreatment of α-LA markedly attenuated Con A-induced hepatitis by modulating cytokine secretion and reducing reactive oxygen species generation.
Collapse
Affiliation(s)
- Miaomiao Fei
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Qun Xie
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yun Zou
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Rong He
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou 221004, Jiangsu, China
| | - Yan Zhang
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Jun Wang
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Lulong Bo
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Jinbao Li
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, China.
| | - Xiaoming Deng
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, China.
| |
Collapse
|
16
|
Ji YR, Kim HJ, Yu DH, Bae KB, Park SJ, Park SJ, Jang WY, Kang MC, Jeong J, Sung YH, Choi M, Park T, Park T, Yun JW, Lee HS, Lee S, Kim MO, Ryoo ZY. Over-expression of Roquin aggravates T cell mediated hepatitis in transgenic mice using T cell specific promoter. Biochem Biophys Res Commun 2014; 452:822-7. [PMID: 25201726 DOI: 10.1016/j.bbrc.2014.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 09/01/2014] [Indexed: 12/13/2022]
Abstract
Chronic hepatitis is a major cause of liver cancer, so earlier treatment of hepatitis might be reducing liver cancer incidence. Hepatitis can be induced in mice by treatment with Concanavalin A (Con A); the resulting liver injury causes significant CD4(+) T cell activation and infiltration. In these T cells, Roquin, a ring-type E3 ubiquitin ligase, is activated. To investigate the role of Roquin, we examined Con A-induced liver injury and T cell infiltration in transgenic (Tg) mice overexpressing Roquin specifically in T cells. In Roquin Tg mice, Con A treatment caused greater increases in both the levels of liver injury enzymes and liver tissue apoptosis, as revealed by TUNEL and H&E staining, than wild type (WT) mice. Further, Roquin Tg mice respond to Con A treatment with greater increases in the T cell population, particularly Th17 cells, though Treg cell counts are lower. Roquin overexpression also enhances increases in pro-inflammatory cytokines, including IFN-γ, TNF-α and IL-6, upon liver injury. Furthermore, Roquin regulates the immune response and apoptosis in Con A induced hepatitis via STATs, Bax and Bcl2. These findings suggest that over-expression of Roquin exacerbates T-cell mediated hepatitis.
Collapse
Affiliation(s)
- Young Rae Ji
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea
| | - Hei Jung Kim
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea
| | - Dong Hun Yu
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea
| | - Ki Beom Bae
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea
| | - Seo Jin Park
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea
| | - Si Jun Park
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea
| | - Woo Young Jang
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea
| | - Min-Cheol Kang
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea
| | - Jain Jeong
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea
| | - Yong Hun Sung
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea
| | - Minjee Choi
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea
| | - Taejun Park
- Department of Biotechnology, Daegu University, Kyungsan, Kyungbuk, Republic of Korea
| | - Taesun Park
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - Jong Won Yun
- Department of Biotechnology, Daegu University, Kyungsan, Kyungbuk, Republic of Korea
| | - Hyun-Shik Lee
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea
| | - Sanggyu Lee
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea
| | - Myoung Ok Kim
- Departments of Animal Science, Kyungpook National University, Sangju, Republic of Korea
| | - Zae Young Ryoo
- School of Life Science, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Buk-ku, Daegu, Republic of Korea.
| |
Collapse
|
17
|
Celaj S, Gleeson MW, Deng J, O'Toole GA, Hampton TH, Toft MF, Morrison HG, Sogin ML, Putra J, Suriawinata AA, Gorham JD. The microbiota regulates susceptibility to Fas-mediated acute hepatic injury. J Transl Med 2014; 94:938-49. [PMID: 25068658 PMCID: PMC4152405 DOI: 10.1038/labinvest.2014.93] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/11/2014] [Accepted: 06/17/2014] [Indexed: 12/28/2022] Open
Abstract
Whereas a significant role for intestinal microbiota in affecting the pathogenesis and progression of chronic hepatic diseases is well documented, the contribution of the intestinal flora to acute liver injury has not been extensively addressed. Elucidating the influence of the intestinal microbiota on acute liver inflammation would be important for better understanding the transition from acute injury to chronic liver disease. Using the Concanavalin A (ConA)-induced liver injury model in laboratory mice, we show that the severity of acute hepatic damage varies greatly among genetically identical mice raised in different environments and harboring distinct microbiota. Through reconstitution of germ-free (GF) mice, and the co-housing of conventional mice, we provide direct evidence that manipulation of the intestinal flora alters susceptibility to ConA-induced liver injury. Through deep sequencing of the fecal microbiome, we observe that the relative abundance of Ruminococcaceae, a Gram(+) family within the class Clostridia, but distinct from segmented filamentous bacteria, is positively associated with the degree of liver damage. Searching for the underlying mechanism(s) that regulate susceptibility to ConA, we provide evidence that the extent of liver injury following triggering of the death receptor Fas varies greatly as a function of the microbiota. We demonstrate that the extent of Fas-induced liver injury increases in GF mice after microbiota reconstitution, and decreases in conventionally raised mice following reduction in intestinal bacterial load, by antibiotic treatment. We also show that the regulation of sensitivity to Fas-induced liver injury is dependent upon the toll-like receptor signaling molecule MyD88. In conclusion, the status and composition of the intestinal microbiota determine the susceptibility to ConA-induced acute liver injury. The microbiota acts as a rheostat, actively modulating the extent of liver damage in response to Fas triggering.
Collapse
Affiliation(s)
- Stela Celaj
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Michael W Gleeson
- Department of Medicine, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Jie Deng
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - George A O'Toole
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Thomas H Hampton
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Martin F Toft
- Taconic Farms Inc., One Hudson City Center, Hudson, NY, USA
| | - Hilary G Morrison
- Josephine Bay Paul Center for Comparative Molecular Biology and Evolution, Marine Biological Laboratory, Woods Hole, MA, USA
| | - Mitchell L Sogin
- Josephine Bay Paul Center for Comparative Molecular Biology and Evolution, Marine Biological Laboratory, Woods Hole, MA, USA
| | - Juan Putra
- Department of Pathology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Arief A Suriawinata
- Department of Pathology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - James D Gorham
- 1] Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA [2] Department of Pathology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
18
|
Xia S, Han M, Li X, Cheng L, Qiang Y, Wu S, Zhang M, Xu H, Liu X, Shao Q. Dietary fish oil exacerbates concanavalin A induced hepatitis through promoting hepatocyte apoptosis and altering immune cell populations. J Toxicol Sci 2014; 39:179-90. [DOI: 10.2131/jts.39.179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Sheng Xia
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, China
- Institute of Clinic Laboratory Diagnosis, School of Medical Science and Laboratory Medicine, Jiangsu University, China
| | - Mutian Han
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, China
| | - Xiaoping Li
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, China
| | - Lu Cheng
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, China
| | - Yetao Qiang
- Institute of Clinic Laboratory Diagnosis, School of Medical Science and Laboratory Medicine, Jiangsu University, China
| | - Shuiyun Wu
- Institute of Clinic Laboratory Diagnosis, School of Medical Science and Laboratory Medicine, Jiangsu University, China
| | - Miaomiao Zhang
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, China
| | - Huaxi Xu
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, China
- Institute of Clinic Laboratory Diagnosis, School of Medical Science and Laboratory Medicine, Jiangsu University, China
| | - Xia Liu
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, China
| | - Qixiang Shao
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, China
- Institute of Clinic Laboratory Diagnosis, School of Medical Science and Laboratory Medicine, Jiangsu University, China
| |
Collapse
|
19
|
Fullerton AM, Roth RA, Ganey PE. Pretreatment with TCDD exacerbates liver injury from Concanavalin A: critical role for NK cells. Toxicol Sci 2013; 136:72-85. [PMID: 23970800 PMCID: PMC3829569 DOI: 10.1093/toxsci/kft174] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 08/09/2013] [Indexed: 12/13/2022] Open
Abstract
For many liver diseases, including viral and autoimmune hepatitis, immune cells play an important role in the development and progression of liver injury. Concanavalin A (Con A) administration to rodents has been used as a model of immune-mediated liver injury resembling human autoimmune hepatitis. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has been demonstrated to alter the development of immune-mediated diseases. Mice pretreated with TCDD developed exacerbated liver injury in response to administration of a mild dose (6 mg/kg) of Con A. In the present study, we tested the hypothesis that TCDD pretreatment exacerbates Con A-induced liver injury by enhancing the activation and recruitment of accessory cell types including neutrophils, macrophages, and natural killer (NK) cells. Mice were treated with 0, 0.3, 3, or 30 μg/kg TCDD and 4 days later with Con A or saline. TCDD pretreatment with doses of 3 and 30 μg/kg significantly increased liver injury from Con A administration. The plasma concentrations of neutrophil chemokines were significantly increased in TCDD-pretreated mice after Con A administration. NKT cell-deficient (CD1d KO) mice were used to examine whether NKT cells were required for TCDD/Con A-induced liver injury. CD1d KO mice were completely protected from liver injury induced by treatment with Con A alone, whereas the injury from TCDD/Con A treatment was reduced but not eliminated. However, T-cell deficient (RAG1 KO) mice were protected from liver injury induced by Con A irrespective of pretreatment with TCDD. TCDD/Con A treatment increased the percentage of NK cells expressing the activation marker CD69. Depletion of NK cells prior to treatment resulted in significant reductions in plasma interferon-γ and liver injury from TCDD/Con A treatment. In summary, exposure to TCDD exacerbated the immune-mediated liver injury induced by Con A, and our findings suggest that NK cells play a critical role in this response.
Collapse
Affiliation(s)
- Aaron M. Fullerton
- Department of Pharmacology & Toxicology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - Robert A. Roth
- Department of Pharmacology & Toxicology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - Patricia E. Ganey
- Department of Pharmacology & Toxicology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| |
Collapse
|
20
|
YANG QIAO, SHI YU, YANG YING, CHEN ZHI. Deactivation and apoptosis of hepatic macrophages are involved in the development of concanavalin A-induced acute liver failure. Mol Med Rep 2013; 8:757-62. [DOI: 10.3892/mmr.2013.1575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 06/26/2013] [Indexed: 11/05/2022] Open
|
21
|
Siegmund K, Lee WY, Tchang VS, Stiess M, Terracciano L, Kubes P, Pieters J. Coronin 1 is dispensable for leukocyte recruitment and liver injury in concanavalin A-induced hepatitis. Immunol Lett 2013; 153:62-70. [DOI: 10.1016/j.imlet.2013.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 06/25/2013] [Accepted: 06/30/2013] [Indexed: 01/13/2023]
|
22
|
Ma C, Kapanadze T, Gamrekelashvili J, Manns MP, Korangy F, Greten TF. Anti-Gr-1 antibody depletion fails to eliminate hepatic myeloid-derived suppressor cells in tumor-bearing mice. J Leukoc Biol 2012; 92:1199-206. [PMID: 23077247 PMCID: PMC3501895 DOI: 10.1189/jlb.0212059] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 08/31/2012] [Accepted: 09/18/2012] [Indexed: 12/29/2022] Open
Abstract
Recent studies show that the liver is a preferred organ for the accumulation of MDSC. In this study, we examined the effect of systemic RB6-8C5 treatment on hepatic MDSC in tumor-bearing mice. EL4 tumor-bearing mice were injected i.p. with RB6-8C5, and hepatic, splenic, and blood MDSCs were analyzed by flow cytometry. Unexpectedly, hepatic MDSC remained in the liver, although RB6-8C5 completely eliminated them from the spleen and peripheral blood 24 h after treatment. Secondary antibody staining confirmed the presence of RB6-8C5-bound MDSC in the liver of mice with s.c. tumors. Similar observations were made in two other (colon and melanoma) tumor models. Whereas RB6-8C5 injection induced cell death of hepatic MDSC, as shown by Annexin V/7-AAD staining, these cells were replaced immediately, leading to a constant, increased frequency of hepatic MDSC. Adoptively transferred MDSC migrated preferentially to the liver after RB6-8C5 treatment, suggesting that hepatic MDSCs are reconstituted rapidly after depletion. Finally, hepatic MDSC remained immunosuppressive despite RB6-8C5 injection. Our study demonstrates that RB6-8C5 is not suitable for depletion of hepatic MDSCs and analysis of their function.
Collapse
Affiliation(s)
- Chi Ma
- Gastrointestinal Malignancy Section, Medical Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and
| | - Tamar Kapanadze
- Gastrointestinal Malignancy Section, Medical Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Jaba Gamrekelashvili
- Gastrointestinal Malignancy Section, Medical Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Michael P. Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Firouzeh Korangy
- Gastrointestinal Malignancy Section, Medical Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and
| | - Tim F. Greten
- Gastrointestinal Malignancy Section, Medical Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; and
| |
Collapse
|
23
|
Malchow S, Thaiss W, Jänner N, Waetzig GH, Gewiese-Rabsch J, Garbers C, Yamamoto K, Rose-John S, Scheller J. Essential role of neutrophil mobilization in concanavalin A-induced hepatitis is based on classic IL-6 signaling but not on IL-6 trans-signaling. Biochim Biophys Acta Mol Basis Dis 2011; 1812:290-301. [DOI: 10.1016/j.bbadis.2010.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 11/18/2010] [Accepted: 11/23/2010] [Indexed: 10/18/2022]
|
24
|
Yin H, Cheng L, Agarwal C, Agarwal R, Ju C. Lactoferrin protects against concanavalin A-induced liver injury in mice. Liver Int 2010; 30:623-32. [PMID: 20136718 DOI: 10.1111/j.1478-3231.2009.02199.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Liver diseases, caused by viral infection, autoimmune conditions, alcohol ingestion or the use of certain drugs, are a significant health issue, as many can develop into liver failure. Lactoferrin (Lac) is an iron-binding glycoprotein that belongs to the transferrin family. Owing to its multiple biological functions, Lac has been evaluated in a number of clinical trials to treat infections, inflammation and cancer. AIM The present study aims to reveal a profound hepatoprotective effect of Lac, using a mouse model of Concanavalin A (Con A)-induced hepatitis, which mimics the pathophysiology of human viral and autoimmune hepatitis. METHOD C57Bl/6J mice were injected with bovine Lac following Con A challenge. The effects of Lac on interferon (IFN)-gamma and interleukin (IL)-4 expression were determined. The roles of Lac on T-cell apoptosis and activation, and leukocytes infiltration were examined. RESULT The data demonstrated that the protective effect of Lac was attributed to its ability to inhibit T-cell activation and production of IFN-gamma, as well as to suppress IL-4 production by hepatic natural killer T cells. CONCLUSION These findings indicate a great therapeutic potential of Lac in treating in treating inflammatory hepatitis and possibly other inflammatory diseases.
Collapse
Affiliation(s)
- Hao Yin
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO, USA
| | | | | | | | | |
Collapse
|
25
|
Corazza N, Badmann A, Lauer C. Immune cell-mediated liver injury. Semin Immunopathol 2009; 31:267-77. [DOI: 10.1007/s00281-009-0168-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 05/27/2009] [Indexed: 02/07/2023]
|
26
|
Induction of hepatitis by JNK-mediated expression of TNF-alpha. Cell 2009; 136:249-60. [PMID: 19167327 DOI: 10.1016/j.cell.2008.11.017] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Revised: 08/21/2008] [Accepted: 11/04/2008] [Indexed: 11/23/2022]
Abstract
The c-Jun NH(2)-terminal kinase (JNK) signaling pathway has been implicated in the development of tumor necrosis factor (TNF)-dependent hepatitis. JNK may play a critical role in hepatocytes during TNF-stimulated cell death in vivo. To test this hypothesis, we examined the phenotype of mice with compound disruption of the Jnk1 and Jnk2 genes. Mice with loss of JNK1/2 expression in hepatocytes exhibited no defects in the development of hepatitis compared with control mice, whereas mice with loss of JNK1/2 in the hematopoietic compartment exhibited a profound defect in hepatitis that was associated with markedly reduced expression of TNF-alpha. These data indicate that JNK is required for TNF-alpha expression but not for TNF-alpha-stimulated death of hepatocytes. Indeed, TNF-alpha induced similar hepatic damage in both mice with hepatocyte-specific JNK1/2 deficiency and control mice. These observations confirm a role for JNK in the development of hepatitis but identify hematopoietic cells as the site of the essential function of JNK.
Collapse
|
27
|
Njoku DB, Li Z, Mellerson JL, Sharma R, Talor MV, Barat N, Rose NR. IP-10 protects while MIP-2 promotes experimental anesthetic hapten - induced hepatitis. J Autoimmun 2009; 32:52-9. [PMID: 19131211 DOI: 10.1016/j.jaut.2008.11.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Revised: 11/06/2008] [Accepted: 11/07/2008] [Indexed: 01/01/2023]
Abstract
MIP-2 and IFN-gamma inducible protein-10 (IP-10) and their respective receptors, CXCR2 and CXCR3, modulate tissue inflammation by recruiting neutrophils or T cells from the spleen or bone marrow. Yet, how these chemokines modulate diseases such as immune-mediated drug-induced liver injury (DILI) is essentially unknown. To investigate how chemokines modulate experimental DILI in our model we used susceptible BALB/c (WT) and IL-4-/- (KO) mice that develop significantly reduced hepatitis and splenic T cell priming to anesthetic haptens and self proteins following TFA-S100 immunizations. We detected CXCR2+ splenic granulocytes in all mice two weeks following immunizations; by three weeks, MIP-2 levels (p<0.001) and GR1+ cells were elevated in WT livers, suggesting MIP-2-recruited granulocytes. Elevated splenic CXCR3+CD4+T cells were identified after two weeks in KO mice indicating elevated IP-10 levels which were confirmed during T cell priming. This result suggested that IP-10 reduced T cell priming to critical DILI antigens. Increased T cell proliferation following co-culture of TFA-S100-primed WT splenocytes with anti-IP-10 (p<0.05) confirmed that IP-10 reduced T cell priming to CYP2E1 and TFA. We propose that MIP-2 promotes and IP-10 protects against the development of hepatitis and T cell priming in this murine model.
Collapse
Affiliation(s)
- Dolores B Njoku
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21287, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Moiseeva EG, Pasechnik AV, Drozdova GA, Frolov VA. Possible mechanisms for the regulation of neutrophil apoptosis during allergic inflammation. Bull Exp Biol Med 2008; 143:305-7. [PMID: 18225748 DOI: 10.1007/s10517-007-0096-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The neutrophil-mediated inflammatory response is regulated via activation of the apoptosis program, which decreases the degree of tissue alteration. In rabbits with allergic inflammation a significant negative correlation was revealed between the intensity of neutrophil apoptosis and blood interferon-gamma concentration.
Collapse
Affiliation(s)
- E G Moiseeva
- Department of General Pathology and Pathophysiology, Russian Peoples' Friendship University of Russia, Moscow
| | | | | | | |
Collapse
|
29
|
Hatano M, Sasaki S, Ohata S, Shiratsuchi Y, Yamazaki T, Nagata K, Kobayashi Y. Effects of Kupffer cell-depletion on Concanavalin A-induced hepatitis. Cell Immunol 2008; 251:25-30. [DOI: 10.1016/j.cellimm.2008.02.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2007] [Revised: 01/17/2008] [Accepted: 02/07/2008] [Indexed: 01/22/2023]
|
30
|
Liu LL, Gong LK, Wang H, Xiao Y, Wu XF, Zhang YH, Xue X, Qi XM, Ren J. Baicalin protects mouse from Concanavalin A-induced liver injury through inhibition of cytokine production and hepatocyte apoptosis. Liver Int 2007; 27:582-91. [PMID: 17403198 DOI: 10.1111/j.1478-3231.2007.01450.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
BACKGROUND Baicalin (BA) exhibits an anti-inflammatory effect in vivo and in vitro and is used to treat chronic hepatitis. However, the mechanism by which BA exerts the liver-protective effect remains largely unknown. AIMS The present study reports that BA inhibits cytokine production and hepatocyte apoptosis to protect mice from liver injury induced by concanavalin A (Con A), a T-cell-dependent liver injury model. RESULTS Con A injection of mice induced severe immune responses and extensive hepatocellular apoptosis within 24 h. Pretreatment of 200 or 100 mg/kg BA markedly reduced serum aminotransferase activities, protected hepatoycte apoptosis and reduced the increase of plasma cytokine levels, including tumor necrosis factor-alpha (TNF-alpha), interferon-gamma (IFN-gamma) and interleukin-6 (IL-6). Furthermore, BA pretreatment decreased tissue myeloperoxidase activity and lipid peroxidation, but increased the superoxide dismutase level. In vitro studies indicated that the beneficial effect of BA was associated with reduced cytokine production from lymphocytes and reduced TNF-alpha-induced hepatocyte apoptosis. CONCLUSION These results suggest that BA has therapeutic potential for T-cell-mediated liver injury.
Collapse
Affiliation(s)
- Lin-Lin Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Mei Y, Wang Y, Xu L. Suppression of immune-mediated liver injury after vaccination with attenuated pathogenic cells. Immunol Lett 2007; 110:29-35. [PMID: 17395274 DOI: 10.1016/j.imlet.2007.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2006] [Revised: 02/16/2007] [Accepted: 02/19/2007] [Indexed: 11/24/2022]
Abstract
Cell vaccination via immunization with attenuated pathogenic cells is an effective preventive method that has been successfully applied in several animal models of inflammatory or autoimmune diseases. Concanavalin A (Con A)-induced hepatitis (CIH) is a commonly used experimental model to study immune-mediated liver injury. Multiple cell types including T lymphocytes, macrophages and neutrophils have been found to be involved in the pathogenesis of CIH. In this study, we used attenuated spleen lymphocytes or peripheral blood lymphocytes as vaccines to investigate whether they could induce protective immune responses to prevent mice from developing CIH. We found that mice receiving such vaccination before CIH induction developed much milder diseases, exhibited a lower level of alanine aminotransferase (ALT) released into their plasma and had less inflammatory lesions in their livers. Such CIH-suppression is dose- and frequency-dependent. The suppressive effect was associated with inhibition of several major inflammatory mediators, pro-inflammatory cytokines and chemokines.
Collapse
Affiliation(s)
- Yunhua Mei
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | | | | |
Collapse
|