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
|
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
|
3
|
Kishimoto TK, Fournier M, Michaud A, Rizzo G, Roy C, Capela T, Nukolova N, Li N, Doyle L, Fu FN, VanDyke D, Traber PG, Spangler JB, Leung SS, Ilyinskii PO. Rapamycin nanoparticles increase the therapeutic window of engineered interleukin-2 and drive expansion of antigen-specific regulatory T cells for protection against autoimmune disease. J Autoimmun 2023; 140:103125. [PMID: 37844543 DOI: 10.1016/j.jaut.2023.103125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/26/2023] [Accepted: 10/04/2023] [Indexed: 10/18/2023]
Abstract
Interleukin-2 (IL-2) therapies targeting the high affinity IL-2 receptor expressed on regulatory T cells (Tregs) have shown promising therapeutic benefit in autoimmune diseases through nonselective expansion of pre-existing Treg populations, but are potentially limited by the inability to induce antigen-specific Tregs, as well as by dose-limiting activation of effector immune cells in settings of inflammation. We recently developed biodegradable nanoparticles encapsulating rapamycin, called ImmTOR, which induce selective immune tolerance to co-administered antigens but do not increase total Treg numbers. Here we demonstrate that the combination of ImmTOR and an engineered Treg-selective IL-2 variant (termed IL-2 mutein) increases the number and durability of total Tregs, as well as inducing a profound synergistic increase in antigen-specific Tregs when combined with a target antigen. We demonstrate that the combination of ImmTOR and an IL-2 mutein leads to durable inhibition of antibody responses to co-administered AAV gene therapy capsid, even at sub-optimal doses of ImmTOR, and provides protection in autoimmune models of type 1 diabetes and primary biliary cholangitis. Importantly, ImmTOR also increases the therapeutic window of engineered IL-2 molecules by mitigating effector immune cell expansion and preventing exacerbation of disease in a model of graft-versus-host-disease. At the same time, IL-2 mutein shows potential for dose-sparing of ImmTOR. Overall, these results establish that the combination of ImmTOR and an IL-2 mutein show synergistic benefit on both safety and efficacy to provide durable antigen-specific immune tolerance to mitigate drug immunogenicity and to treat autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | - Gina Rizzo
- Selecta Biosciences, Watertown, MA, 02472, USA
| | | | | | | | - Ning Li
- Selecta Biosciences, Watertown, MA, 02472, USA
| | - Liam Doyle
- Selecta Biosciences, Watertown, MA, 02472, USA
| | - Fen-Ni Fu
- Selecta Biosciences, Watertown, MA, 02472, USA
| | - Derek VanDyke
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | | | - Jamie B Spangler
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | | | | |
Collapse
|
4
|
Liu W, Zeng X, Liu Y, Liu J, Li C, Chen L, Chen H, Ouyang D. The Immunological Mechanisms and Immune-Based Biomarkers of Drug-Induced Liver Injury. Front Pharmacol 2021; 12:723940. [PMID: 34721020 PMCID: PMC8554067 DOI: 10.3389/fphar.2021.723940] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Drug-induced liver injury (DILI) has become one of the major challenges of drug safety all over the word. So far, about 1,100 commonly used drugs including the medications used regularly, herbal and/or dietary supplements, have been reported to induce liver injury. Moreover, DILI is the main cause of the interruption of new drugs development and drugs withdrawn from the pharmaceutical market. Acute DILI may evolve into chronic DILI or even worse, commonly lead to life-threatening acute liver failure in Western countries. It is generally considered to have a close relationship to genetic factors, environmental risk factors, and host immunity, through the drug itself or its metabolites, leading to a series of cellular events, such as haptenization and immune response activation. Despite many researches on DILI, the specific biomarkers about it are not applicable to clinical diagnosis, which still relies on the exclusion of other causes of liver disease in clinical practice as before. Additionally, circumstantial evidence has suggested that DILI is mediated by the immune system. Here, we review the underlying mechanisms of the immune response to DILI and provide guidance for the future development of biomarkers for the early detection, prediction, and diagnosis of DILI.
Collapse
Affiliation(s)
- Wenhui Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Xiangchang Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Yating Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Jinfeng Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Chaopeng Li
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Lulu Chen
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Hongying Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Dongsheng Ouyang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China.,Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| |
Collapse
|
5
|
Sohrabi M, Gholami A, Amir Kalali B, Khoonsari M, Sahraei R, Nasiri Toosi M, Zamani F, Keyvani H. Are Serum Levels of Nuclear Factor Kappa B and Forkhead Box Protein P3 in Patients with Non-Alcoholic Fatty Liver Disease Related to Severity of Fibrosis? Middle East J Dig Dis 2021; 13:356-362. [PMID: 36606010 PMCID: PMC9489454 DOI: 10.34172/mejdd.2021.247] [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] [Received: 01/29/2021] [Accepted: 06/02/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Inflammation has a significant impact on the development and progression of fatty liver diseases.In this study, we aimed to investigate the relation between serum levels of nuclear factor kappa B (NFkB) and Forkhead box protein P3 (FOXP3)with fibrosis severity among patients with non-alcoholic fatty liver disease(NAFLD). METHODS In a prospective study, the patients suspicios of havingfatty liver were enrolled. The exclusion criteria lack of viral hepatitis, autoimmune hepatitis, Wilson's or other known liver diseases,history of liver or biliary surgery,bariatric surgery, and medications that influence liver metabolism. The participantsunderwent liver fibroscan. According to liver fibrosis, the patients weredivided into two groups; 1)fibrosis less than 7.2 KP,2)advanced NAFLD, fibrosis ≥7.3 KP. A10 cc fasting blood sample was taken from each patient for laboratory assessments.The variables between the two groups were compared using Chi-square or Fisher's exact test.The independence of cytokines was assessed by a logistic regression test. RESULTS Totally 90 patients were enrolled.The mean age was 42.21 ± 11 years. Of them, 50 and 47 participants were allocated to groups 1 and 2, respectively. In the univariate analysis, we revealed asignificant difference between age, body mass index (BMI), fasting blood glucose, liver enzymes, total cholesterol, andtriglyceride levels. Also, there was a significant difference betweenthe levels of NFKB and FOXP3 in group one compared with group two of the participants,as FOXP3(9.17 ± 10.0 vs. 18.63 ± 12.9; p < 0.001) and NFKB (1.70 ± 1.70; p < 0.01). After excluding the confounding factors, we observed a significant association between fibrosis level and cytokine levels in logistic regression. CONCLUSION Serum levels of NFKB and FOXP3 increased by advancing liver fibrosis in patients with NAFLD.This is an independent association. The identification of intermediary regulatory factors would be necessary.
Collapse
Affiliation(s)
- Masoudreza Sohrabi
- Gastrointestinal and Liver Disease Research Center (GILDRC), Iran University of Medical Sciences,Tehran, Iran
| | - Ali Gholami
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
,Department of Epidemiology & Biostatistics, School of Public Health, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Bahareh Amir Kalali
- Gastrointestinal and Liver Disease Research Center (GILDRC), Iran University of Medical Sciences,Tehran, Iran
| | - Mahmoodreza Khoonsari
- Gastrointestinal and Liver Disease Research Center (GILDRC), Iran University of Medical Sciences,Tehran, Iran
| | - Roghieh Sahraei
- Gastrointestinal and Liver Disease Research Center (GILDRC), Iran University of Medical Sciences,Tehran, Iran
| | - Mohsen Nasiri Toosi
- Liver transplantation Research Center. Imam Khomeini Hospital, Tehran University of Medical Sciences.Tehran iran
| | - Farhad Zamani
- Gastrointestinal and Liver Disease Research Center (GILDRC), Iran University of Medical Sciences,Tehran, Iran
| | - Hossein Keyvani
- Gastrointestinal and Liver Disease Research Center (GILDRC), Iran University of Medical Sciences,Tehran, Iran
,Department of Virology, Iran University of Medical Sciences, Tehran, IR Iran
,Corresponding Author: Hossein Keyvani, PhD Department of Virology, Iran University of Medical Sciences, Tehran, IR Iran. Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, IR Iran. Tel : + 98 21 88941831 Fax : + 98 21 88941831
| |
Collapse
|
6
|
Gough NR, Xiang X, Mishra L. TGF-β Signaling in Liver, Pancreas, and Gastrointestinal Diseases and Cancer. Gastroenterology 2021; 161:434-452.e15. [PMID: 33940008 PMCID: PMC8841117 DOI: 10.1053/j.gastro.2021.04.064] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/05/2021] [Accepted: 04/25/2021] [Indexed: 02/06/2023]
Abstract
Genetic alterations affecting transforming growth factor-β (TGF-β) signaling are exceptionally common in diseases and cancers of the gastrointestinal system. As a regulator of tissue renewal, TGF-β signaling and the downstream SMAD-dependent transcriptional events play complex roles in the transition from a noncancerous disease state to cancer in the gastrointestinal tract, liver, and pancreas. Furthermore, this pathway also regulates the stromal cells and the immune system, which may contribute to evasion of the tumors from immune-mediated elimination. Here, we review the involvement of the TGF-β pathway mediated by the transcriptional regulators SMADs in disease progression to cancer in the digestive system. The review integrates human genomic studies with animal models that provide clues toward understanding and managing the complexity of the pathway in disease and cancer.
Collapse
Affiliation(s)
- Nancy R. Gough
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, New York
| | - Xiyan Xiang
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, New York
| | - Lopa Mishra
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, New York; Center for Translational Medicine, Department of Surgery, The George Washington University, Washington, District of Columbia.
| |
Collapse
|
7
|
Silva RCP, da Silva RPC, Souto FO, de Lorena VMB, Aires ADL, Costa VMA, Albuquerque MCPDA, de Souza VMO. Extract from Ascaris suum and N-acetyl-L-cysteine induces an immunosuppressant effect in model of autoimmune hepatitis. Parasite Immunol 2021; 43:e12826. [PMID: 33586210 DOI: 10.1111/pim.12826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/27/2021] [Accepted: 02/10/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Extract of adult Ascaris suum (ASC) worms attenuated the liver damage in experimental autoimmune hepatitis (EAH) with induction of Th2 immune response, but fibrosis occurred. N-acetyl-L-cysteine (NAC) has protective effects against liver fibrosis. OBJECTIVES Evaluate the association ASC + NAC on the T- and B-cell activation, inflammation and fibrogenic markers in the liver in EAH. METHODS Experimental autoimmune hepatitis was induced intravenously with concanavalin A in BALB/c mice. EAH + ASC+NAC group received NAC and ASC; EAH + ASC group received ASC; EAH group received PBS. Doubly labelled CD4+ T (CD28, CTLA-4, CD40L or IL-10) and CD45R+ B lymphocytes (IL-10) and CD4+ CD25+ FoxP3+ cells were evaluated, along with gene expression of Col1a1, α-SMA, Fizz1, Arg1 and PPAR-γ and histomorphometry. RESULTS Experimental autoimmune hepatitis group showed high frequency of CD28+ and CD40L+ T lymphocytes, but not the EAH + ASC group. In relation to EAH group, the Fizz1 expression was lower in both groups treated, but Arg1 expression was lower in only EAH + ASC+NAC group. In the EAH + ASC+NAC group, there were higher frequencies of CD4+ IL-10+ and CD4+ CD25+ FoxP3+ cells, but not CD45R+ IL-10+ , along with mitigated inflammation and collagen production. CONCLUSIONS Ascaris suum favoured immunosuppression in EAH limiting the T cells activation. However, association ASC and NAC was necessary for attenuating the inflammatory process and collagen production.
Collapse
Affiliation(s)
- Roeckson Carlos Peixoto Silva
- Keizo Asami Immunopathology Laboratory (LIKA), Federal University of Pernambuco, Pernambuco, Brazil.,Department of Tropical Medicine, Federal University of Pernambuco, Pernambuco, Brazil
| | - Raul Penaforte Correia da Silva
- Keizo Asami Immunopathology Laboratory (LIKA), Federal University of Pernambuco, Pernambuco, Brazil.,Department of Tropical Medicine, Federal University of Pernambuco, Pernambuco, Brazil
| | - Fabrício Oliveira Souto
- Keizo Asami Immunopathology Laboratory (LIKA), Federal University of Pernambuco, Pernambuco, Brazil.,Agreste Academic Centre, Federal University of Pernambuco, Pernambuco, Brazil
| | | | - André de Lima Aires
- Keizo Asami Immunopathology Laboratory (LIKA), Federal University of Pernambuco, Pernambuco, Brazil.,Department of Tropical Medicine, Federal University of Pernambuco, Pernambuco, Brazil
| | - Vlaudia Maria Assis Costa
- Keizo Asami Immunopathology Laboratory (LIKA), Federal University of Pernambuco, Pernambuco, Brazil.,Department of Tropical Medicine, Federal University of Pernambuco, Pernambuco, Brazil
| | - Mônica Camelo Pessôa de Azevedo Albuquerque
- Keizo Asami Immunopathology Laboratory (LIKA), Federal University of Pernambuco, Pernambuco, Brazil.,Department of Tropical Medicine, Federal University of Pernambuco, Pernambuco, Brazil
| | - Valdênia Maria Oliveira de Souza
- Keizo Asami Immunopathology Laboratory (LIKA), Federal University of Pernambuco, Pernambuco, Brazil.,Department of Pharmaceutical Sciences, Federal University of Pernambuco, Pernambuco, Brazil
| |
Collapse
|
8
|
Ishay Y, Rotnemer-Golinkin D, Ilan Y. The role of the sphingosine axis in immune regulation: A dichotomy in the anti-inflammatory effects between sphingosine kinase 1 and sphingosine kinase 2-dependent pathways. Int J Immunopathol Pharmacol 2021; 35:20587384211053274. [PMID: 34789044 PMCID: PMC8645305 DOI: 10.1177/20587384211053274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/21/2021] [Indexed: 01/05/2023] Open
Abstract
Background: Sphingosine kinase has been identified as playing a central role in the immune cascade, being a common mediator in the cellular response to a variety of signals. The different effects of sphingosine kinase 1 and 2 (SphK1 and SphK2, respectively) activity have not been completely characterized. Aim: To determine the different roles played by SphK1 and SphK2 in the regulation of immune-mediated disorders. Methods: Nine groups of mice were studied. Concanavalin A (ConA) injection was used to induce immune-mediated hepatitis. Mice were treated with SphK1 inhibitor (termed SphK-I) and SphK2 inhibitor (termed ABC294640), prior to ConA injection, and effects of treatment on liver enzymes, subsets of T lymphocytes, and serum levels of cytokines were observed. Results: While liver enzyme elevation was ameliorated by administration of SphK1 inhibitor, SphK2 inhibitor-treated mice did not show this tendency. A marked decrease in expression of CD25+ T-cells and Foxp+ T-cells was observed in mice treated with a high dose of SphK1 inhibitor. Alleviation of liver damage was associated with a statistically significant reduction of serum IFNγ levels in mice treated with SphK1 inhibitor and not in those treated with SphK2 inhibitor. Conclusions: Early administration of SphK1 inhibitor in a murine model of immune-mediated hepatitis alleviated liver damage and inflammation with a statistically significant reduction in IFN-γ levels. The data support a dichotomy in the anti-inflammatory effects of SphK1 and SphK2, and suggests that isoenzyme-directed therapies can improve the effect of targeting these pathways.
Collapse
Affiliation(s)
- Yuval Ishay
- Department of Medicine, Hadassah-Hebrew University Medical
Center, Jerusalem Israel
| | | | - Yaron Ilan
- Department of Medicine, Hadassah-Hebrew University Medical
Center, Jerusalem Israel
| |
Collapse
|
9
|
Hu WC. A Framework of All Discovered Immunological Pathways and Their Roles for Four Specific Types of Pathogens and Hypersensitivities. Front Immunol 2020; 11:1992. [PMID: 32849663 PMCID: PMC7426516 DOI: 10.3389/fimmu.2020.01992] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 07/23/2020] [Indexed: 01/05/2023] Open
Affiliation(s)
- Wan-Chung Hu
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| |
Collapse
|
10
|
Silva RCP, Silva RPCD, Silva MDC, Nascimento WRCD, Costa VMA, Azevedo Albuquerque MCPD, Souza VMOD. Extract of Ascaris suum induces TGF-β and early production of IgG1 in experimental autoimmune hepatitis. REVISTA BRASILEIRA DE PARASITOLOGIA VETERINARIA = BRAZILIAN JOURNAL OF VETERINARY PARASITOLOGY : ORGAO OFICIAL DO COLEGIO BRASILEIRO DE PARASITOLOGIA VETERINARIA 2020; 29:e023419. [PMID: 32520090 DOI: 10.1590/s1984-29612020031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/06/2020] [Indexed: 06/11/2023]
Abstract
In experimental autoimmune hepatitis (EAH) of Th1 profile, an extract of adult Ascaris suum worms (ASC) was previously found to deviate the immune response to a Th2/IL-10 pattern. Here, the effects of treatment with ASC on production of TGF-β and the anti-Ascaris isotypes IgG1 and IgG2a in EAH were evaluated. EAH was induced in BALB/c mice, intravenously with concanavalin A. Two hours later, these animals received ASC (EAH+ASC group) or PBS vehicle (EAH group). IgG1 and IgG2a were evaluated 8 h, 24 h and 7 d after induction. TGF-β was measured in a splenocyte culture at this last time. The isotype levels in the EAH group were low throughout the kinetics. In the EAH+ASC group, there was significant production of IgG1 at 24 h and 7 d, but of IgG2a only at 7 d. There was statistically greater production of TGF-β in the EAH+ASC group. The higher levels of IgG1 and TGF-β in this group suggest that an additional Th1 response control route exists in EAH, which needs to be investigated.
Collapse
Affiliation(s)
- Roeckson Carlos Peixoto Silva
- Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco - UFPE, Recife, PE, Brasil
- Departamento de Medicina Tropical, Universidade Federal de Pernambuco - UFPE, Recife, PE, Brasil
| | - Raul Penaforte Correia da Silva
- Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco - UFPE, Recife, PE, Brasil
- Departamento de Medicina Tropical, Universidade Federal de Pernambuco - UFPE, Recife, PE, Brasil
| | - Maria da Conceição Silva
- Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco - UFPE, Recife, PE, Brasil
- Departamento de Medicina Tropical, Universidade Federal de Pernambuco - UFPE, Recife, PE, Brasil
| | | | - Vlaudia Maria Assis Costa
- Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco - UFPE, Recife, PE, Brasil
- Departamento de Medicina Tropical, Universidade Federal de Pernambuco - UFPE, Recife, PE, Brasil
| | - Mônica Camelo Pessôa de Azevedo Albuquerque
- Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco - UFPE, Recife, PE, Brasil
- Departamento de Medicina Tropical, Universidade Federal de Pernambuco - UFPE, Recife, PE, Brasil
| | - Valdênia Maria Oliveira de Souza
- Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco - UFPE, Recife, PE, Brasil
- Departamento de Ciências Farmacêuticas, Universidade Federal de Pernambuco - UFPE, Recife, PE, Brasil
| |
Collapse
|
11
|
Ge W, Gao Y, Zhao Y, Yang Y, Sun Q, Yang X, Xu X, Zhang J. Decreased T-cell mediated hepatic injury in concanavalin A-treated PLRP2-deficient mice. Int Immunopharmacol 2020; 85:106604. [PMID: 32428799 DOI: 10.1016/j.intimp.2020.106604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022]
Abstract
Concanavalin A (Con A) activates innate immunity and causes liver damage mediated by cytotoxic T lymphocytes (CTL) in mice. The Pancreatic lipase-related protein 2 (PLRP2) is induced by interleukin (IL)-4 in vitro in CTLs and associated with CTL functions. We examined the role of PLRP2 in a mouse model of Con A-induced T cell-mediated hepatitis. PLRP2-knockout and wild-type (WT) mice were inoculated with 20 mg/kg Con A. Mice lacking PLRP2 reduced Con A-induced hepatitis, which was manifested by a decrease in serum aminotransferase and histopathological assessment. The expression and secretion of cytokines including tumor necrosis factor-alpha (TNF-α), interferon (IFN)-γ, IL-6, and IL-1β were suppressed in Con A-treated PLRP2-knockout mice. In PLRP2 knockout mice, Con A-induced liver chemokines and adhesion molecules (such as MIP-1α, MIP-1β, ICAM-1 and MCP-1) were also down regulated. In the WT liver treated with Con A, the number of T cells (CD4+ and CD8+) and macrophages (CD11b+ F4/80+) increased significantly, while the lack of PLRP2 reduced the number of T cells in the liver, but had no effect on macrophages. The shift of the metabolic profiles was impaired in Con A-treated PLRP2-knockout mice compared to WT mice. In conclusion, these results indicate that PLRP2 deficiency reduces T-cell mediated Con A-induced hepatitis, and suggest PLRP2 is a potential target of anti-inflammatory and immunomodulatory drugs to treat immune-mediated hepatitis.
Collapse
Affiliation(s)
- Wenhao Ge
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Yan Gao
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Yang Zhao
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Yunxia Yang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Qi Sun
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Xiao Yang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Xi Xu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Jianfa Zhang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China.
| |
Collapse
|
12
|
Wang H, Lu CH, Ho PC. Metabolic adaptation orchestrates tissue context-dependent behavior in regulatory T cells. Immunol Rev 2020; 295:126-139. [PMID: 32147869 DOI: 10.1111/imr.12844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/19/2020] [Indexed: 02/07/2023]
Abstract
The diverse distribution and functions of regulatory T cells (Tregs) ensure tissue and immune homeostasis; however, it remains unclear which factors can guide distribution, local differentiation, and tissue context-specific behavior in Tregs. Although the emerging concept that Tregs could re-adjust their transcriptome based on their habitations is supported by recent findings, the underlying mechanisms that reprogram transcriptome in Tregs are unknown. In the past decade, metabolic machineries have been revealed as a new regulatory circuit, known as immunometabolic regulation, to orchestrate activation, differentiation, and functions in a variety of immune cells, including Tregs. Given that systemic and local alterations of nutrient availability and metabolite profile associate with perturbation of Treg abundance and functions, it highlights that immunometabolic regulation may be one of the mechanisms that orchestrate tissue context-specific regulation in Tregs. The understanding on how metabolic program instructs Tregs in peripheral tissues not only represents a critical opportunity to delineate a new avenue in Treg biology but also provides a unique window to harness Treg-targeting approaches for treating cancer and autoimmunity with minimizing side effects. This review will highlight the metabolic features on guiding Treg formation and function in a disease-oriented perspective and aim to pave the foundation for future studies.
Collapse
Affiliation(s)
- Haiping Wang
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Chun-Hao Lu
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
13
|
El-Kashef DH, Abdelrahman RS. Montelukast ameliorates Concanavalin A-induced autoimmune hepatitis in mice via inhibiting TNF-α/JNK signaling pathway. Toxicol Appl Pharmacol 2020; 393:114931. [PMID: 32109511 DOI: 10.1016/j.taap.2020.114931] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 02/17/2020] [Accepted: 02/24/2020] [Indexed: 01/19/2023]
Abstract
BACKGROUND Concanavalin A (ConA) is a well-established model to induce autoimmune hepatitis (AIH) in mice which mimics pathological alterations that occur in human. The pathogenesis of ConA-induced AIH involves many signaling pathways. Montelukast is a leukotriene receptor antagonist that is mainly used in the management of asthma. The antioxidant, anti-inflammatory and anti-apoptotic effects of montelukast have been reported in previous studies. Lately, montelukast has been documented to confer protection against various inflammatory diseases. Up to date, no study has explored the effect of montelukast on AIH induced by ConA. AIM AND METHOD This study aims to detect the protective effects of montelukast (10 mg/kg) on ConA (20 mg/kg)- induced AIH in mice and to demonstrate its hepatoprotective mechanisms. Hepatic function, histological changes, oxidative stress, inflammation, autophagy, and apoptotic markers were investigated. RESULTS Hepatic function and histological data revealed that treatment with montelukast significantly attenuated ConA-induced hepatic damage. Montelukast significantly reduced JNK level and NFκB p65 expression, and inhibited proinflammatory cytokines (TNF-α and IL-6) as well as oxidative stress (MDA, NO, and GSH). Moreover, inflammatory cells (CD4+ infiltration and the levels of apoptotic markers (Bax and caspase-3) besides autophagy biomarkers (Beclin1 and LC3) were reduced. CONCLUSION Our results suggest that montelukast could be a potential therapeutic drug against the ConA-induced AIH through its anti-oxidant, anti-inflammatory, anti- autophagy as well as anti-apoptotic properties.
Collapse
Affiliation(s)
- Dalia H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt
| | - Rehab S Abdelrahman
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al-Madina Al-Munawwarah, 30001, Saudi Arabia. Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt.
| |
Collapse
|
14
|
Koda Y, Nakamoto N, Chu PS, Ugamura A, Mikami Y, Teratani T, Tsujikawa H, Shiba S, Taniki N, Sujino T, Miyamoto K, Suzuki T, Yamaguchi A, Morikawa R, Sato K, Sakamoto M, Yoshimoto T, Kanai T. Plasmacytoid dendritic cells protect against immune-mediated acute liver injury via IL-35. J Clin Invest 2019; 129:3201-3213. [PMID: 31264967 DOI: 10.1172/jci125863] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 05/14/2019] [Indexed: 12/12/2022] Open
Abstract
Acute liver failure (ALF) is a life-threatening condition, and liver transplantation is the only therapeutic option. Although immune dysregulation is central to its pathogenesis, the precise mechanism remains unclear. Here, we show that the number of peripheral and hepatic plasmacytoid DCs (pDCs) decrease during acute liver injury in both humans and mice. Selective depletion of pDCs in Siglechdtr/+ mice exacerbated concanavalin A-induced acute liver injury. In contrast, adoptively transferred BM-derived pDCs preferentially accumulated in the inflamed liver and protected against liver injury. This protective effect was independent of TLR7 and TLR9 signaling, since a similar effect occurred following transfer of MyD88-deficient pDCs. Alternatively, we found an unexpected immunosuppressive role of pDCs in an IL-35-dependent manner. Both Il12a and Ebi3, heterodimeric components of IL-35, were highly expressed in transferred pDCs and CD4+CD25+ Tregs. However, the protective effect of pDC transfer was completely lost in mice depleted of Tregs by anti-CD25 antibody. Moreover, pDCs derived from IL-35-deficient mice had less of a protective effect both in vivo and in vitro even in the presence of Tregs. These results highlight a unique aspect of pDCs in association with Tregs, serving as a guide for immunotherapeutic options in ALF.
Collapse
Affiliation(s)
- Yuzo Koda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.,Mitsubishi Tanabe Pharma Corporation, Kanagawa, Japan
| | - Nobuhiro Nakamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Po-Sung Chu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Aya Ugamura
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Toshiaki Teratani
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hanako Tsujikawa
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Shunsuke Shiba
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Nobuhito Taniki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tomohisa Sujino
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kentaro Miyamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takahiro Suzuki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akihiro Yamaguchi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Rei Morikawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Katsuaki Sato
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Michiie Sakamoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Takayuki Yoshimoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.,Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| |
Collapse
|
15
|
Treg/Th17 imbalance is associated with poor autoimmune hepatitis prognosis. Clin Immunol 2019; 198:79-88. [DOI: 10.1016/j.clim.2018.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 10/04/2018] [Accepted: 11/09/2018] [Indexed: 12/27/2022]
|
16
|
Gazdic M, Markovic BS, Arsenijevic A, Jovicic N, Acovic A, Harrell CR, Fellabaum C, Djonov V, Arsenijevic N, Lukic ML, Volarevic V. Crosstalk between mesenchymal stem cells and T regulatory cells is crucially important for the attenuation of acute liver injury. Liver Transpl 2018; 24:687-702. [PMID: 29500914 DOI: 10.1002/lt.25049] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/08/2018] [Accepted: 02/12/2018] [Indexed: 12/15/2022]
Abstract
One of the therapeutic options for the treatment of fulminant hepatitis is repopulation of intrahepatic regulatory cells because their pool is significantly reduced during acute liver failure. Although it is known that mesenchymal stem cells (MSCs), which have beneficent effects in the therapy of fulminant hepatitis, may promote expansion of regulatory T cells (Tregs) and regulatory B cells (Bregs), the role of these regulatory cells in MSC-mediated attenuation of acute liver injury is unknown. Herewith, we described the molecular mechanisms involved in the crosstalk between MSCs and liver regulatory cells and analyzed the potential of MSC-based therapy for the expansion of intrahepatic regulatory cells in mouse model of acute liver failure. MSC-dependent attenuation of α-galactosylceramide (α-GalCer)-induced acute liver injury in mice was accompanied with an increased presence of interleukin (IL) 10-producing CD4+ CD25+ forkhead box P3+ Tregs and IL10- and transforming growth factor β-producing marginal zone-like Bregs in the liver. Depletion of Bregs did not alter MSC-based alleviation of acute liver failure, whereas depletion of Tregs completely abrogated hepatoprotective effects of MSCs and inhibited their capacity to attenuate hepatotoxicity of liver natural killer T cells (NKTs), indicating that Tregs, and not Bregs, were critically involved in MSC-based modulation of acute liver inflammation. MSCs, in a paracrine, indoleamine 2,3-dioxygenase-dependent manner, significantly increased the capacity of Tregs to produce immunosuppressive IL10 and to suppress hepatotoxicity of liver NKTs. Accordingly, adoptive transfer of MSC-primed Tregs resulted in the complete attenuation of α-GalCer-induced acute liver failure. In conclusion, our findings highlighted the crucial importance of Tregs for MSC-based attenuation of acute liver failure and indicated the significance of MSC-mediated priming of Tregs as a new therapeutic approach in Treg-based therapy of acute liver injury. Liver Transplantation 24 687-702 2018 AASLD.
Collapse
Affiliation(s)
- Marina Gazdic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Bojana Simovic Markovic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Aleksandar Arsenijevic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nemanja Jovicic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Aleksandar Acovic
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | | | | | - Nebojsa Arsenijevic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Miodrag L Lukic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Vladislav Volarevic
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
17
|
Marshall GP, Cserny J, Perry DJ, Yeh WI, Seay HR, Elsayed AG, Posgai AL, Brusko TM. Clinical Applications of Regulatory T cells in Adoptive Cell Therapies. CELL & GENE THERAPY INSIGHTS 2018; 4:405-429. [PMID: 34984106 PMCID: PMC8722436 DOI: 10.18609/cgti.2018.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Interest in adoptive T-cell therapies has been ignited by the recent clinical success of genetically-modified T cells in the cancer immunotherapy space. In addition to immune targeting for malignancies, this approach is now being explored for the establishment of immune tolerance with regulatory T cells (Tregs). Herein, we will summarize the basic science and clinical results emanating from trials directed at inducing durable immune regulation through administration of Tregs. We will discuss some of the current challenges facing the field in terms of maximizing cell purity, stability and expansion capacity, while also achieving feasibility and GMP production. Indeed, recent advances in methodologies for Treg isolation, expansion, and optimal source materials represent important strides toward these considerations. Finally, we will review the emerging genetic and biomaterial-based approaches on the horizon for directing Treg specificity to augment tissue-targeting and regenerative medicine.
Collapse
Affiliation(s)
| | - Judit Cserny
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Daniel J Perry
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Wen-I Yeh
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Howard R Seay
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Ahmed G Elsayed
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA.,Department of Microbiology and Immunology, Faculty of Medicine, Mansoura University, Egypt
| | - Amanda L Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Todd M Brusko
- OneVax LLC, Sid Martin Biotechnology Institute, Alachua, Florida, USA.,Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| |
Collapse
|
18
|
Ma WT, Liu QZ, Yang JB, Yang YQ, Zhao ZB, Ma HD, Gershwin ME, Lian ZX. A Mouse Model of Autoimmune Cholangitis via Syngeneic Bile Duct Protein Immunization. Sci Rep 2017; 7:15246. [PMID: 29127360 PMCID: PMC5681628 DOI: 10.1038/s41598-017-15661-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 10/26/2017] [Indexed: 12/22/2022] Open
Abstract
Primary biliary cholangitis (PBC) is an autoimmune liver disease characterized by the destruction of interlobular biliary ductules, which progressively leads to cholestasis, hepatic fibrosis, cirrhosis, and eventually liver failure. Several mouse models have been used to clarify the pathogenesis of PBC and are generally considered reflective of an autoimmune cholangitis. Most models focus on issues of molecular mimicry between the E2 subunit of the pyruvate dehydrogenase complex (PDC-E2), the major mitochondrial autoantigen of PBC and xenobiotic cross reactive chemicals. None have focused on the classic models of breaking tolerance, namely immunization with self-tissue. Here, we report a novel mouse model of autoimmune cholangitis via immunization with syngeneic bile duct protein (BDP). Our results demonstrate that syngeneic bile duct antigens efficiently break immune tolerance of recipient mice, capturing several key features of PBC, including liver-specific inflammation focused on portal tract areas, increased number and activation state of CD4 and CD8 T cells in the liver and spleen. Furthermore, the germinal center (GC) responses in the spleen were more enhanced in our mouse model. Finally, these mice were 100% positive for anti-mitochondrial antibodies (AMAs). In conclusion, we developed a novel mouse model of PBC that may help to elucidate the detailed mechanism of this complex disease.
Collapse
Affiliation(s)
- Wen-Tao Ma
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, 510006, China.,Liver Immunology Laboratory, Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.,College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Qing-Zhi Liu
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, 510006, China.,Liver Immunology Laboratory, Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Jing-Bo Yang
- Liver Immunology Laboratory, Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Yan-Qing Yang
- Liver Immunology Laboratory, Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Zhi-Bin Zhao
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, 510006, China.,Liver Immunology Laboratory, Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Hong-Di Ma
- Liver Immunology Laboratory, Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, Davis, CA, USA
| | - Zhe-Xiong Lian
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, 510006, China. .,Liver Immunology Laboratory, Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China. .,Innovation Center for Cell Signaling Network, Hefei National Laboratory for Physical Sciences at Microscale, Hefei, 230027, China.
| |
Collapse
|
19
|
Wang L, Zhang W, Ge CH, Yin RH, Xiao Y, Zhan YQ, Yu M, Li CY, Ge ZQ, Yang XM. Toll-like receptor 5 signaling restrains T-cell/natural killer T-cell activation and protects against concanavalin A-induced hepatic injury. Hepatology 2017; 65:2059-2073. [PMID: 28273362 DOI: 10.1002/hep.29140] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 02/27/2017] [Indexed: 01/06/2023]
Abstract
UNLABELLED Toll-like receptor-5 (TLR5) signaling regulates the immune privileged status of the liver and is involved in hepatic immune disorders. However, the role of TLR5 has not yet been investigated in experimental models of concanavalin A (Con A)-mediated liver injury. Here, we show that TLR5 is highly up-regulated in the hepatic mononuclear cells of mice during Con A-induced hepatitis. Increased mortality and liver histopathology of TLR5-deficient mice correlated with excessive production of proinflammatory cytokines, suggesting that TLR5 knockout mice were more susceptible to Con A-induced hepatitis. We also report that administration of CBLB502, an exogenous TLR5 agonist, substantially alleviated Con A-mediated hepatitis in wild-type mice as shown by increased survival rates, reduced aminotransferase and proinflammatory cytokine production, impaired lymphocyte infiltration, and ameliorated hepatocyte necrosis and/or apoptosis. Mechanistic studies revealed that CBLB502 acts as a negative regulator in limiting T-cell/natural killer T-cell activity and cytokine production in the Con A-hepatitis model. Bone marrow transplantation experiments showed that TLR5 in bone marrow-derived cells contributed to the hepatoprotective efficacy of CBLB502 against Con A-induced liver injury. Moreover, interleukin-6 elevation induced by CBLB502 is an important protective factor against Con A-induced liver injury. In addition, we demonstrate that CBLB502 suppresses α-galactosylceramide-induced natural killer T cell-dependent inflammatory liver injury. CONCLUSION The TLR5 signaling pathway plays an important role in T cell-mediated hepatic injury and may be exploited for therapeutic treatment of inflammatory liver diseases. (Hepatology 2017;65:2059-2073).
Collapse
Affiliation(s)
- Lei Wang
- Department of Pharmaceutical Engineering, Tianjin University, Tianjin, China.,Beijing Institute of Radiation Medicine, Beijing, China
| | - Wen Zhang
- Department of Pharmaceutical Engineering, Tianjin University, Tianjin, China.,Beijing Institute of Radiation Medicine, Beijing, China
| | - Chang-Hui Ge
- Beijing Institute of Radiation Medicine, Beijing, China.,State Key Laboratory of Proteomics, Beijing, China
| | - Rong-Hua Yin
- Beijing Institute of Radiation Medicine, Beijing, China.,State Key Laboratory of Proteomics, Beijing, China
| | - Yang Xiao
- Beijing Institute of Radiation Medicine, Beijing, China.,State Key Laboratory of Proteomics, Beijing, China
| | - Yi-Qun Zhan
- Beijing Institute of Radiation Medicine, Beijing, China.,State Key Laboratory of Proteomics, Beijing, China
| | - Miao Yu
- Beijing Institute of Radiation Medicine, Beijing, China.,State Key Laboratory of Proteomics, Beijing, China
| | - Chang-Yan Li
- Beijing Institute of Radiation Medicine, Beijing, China.,State Key Laboratory of Proteomics, Beijing, China
| | - Zhi-Qiang Ge
- Department of Pharmaceutical Engineering, Tianjin University, Tianjin, China
| | - Xiao-Ming Yang
- Department of Pharmaceutical Engineering, Tianjin University, Tianjin, China.,Beijing Institute of Radiation Medicine, Beijing, China.,State Key Laboratory of Proteomics, Beijing, China
| |
Collapse
|
20
|
Pereira LMS, Gomes STM, Ishak R, Vallinoto ACR. Regulatory T Cell and Forkhead Box Protein 3 as Modulators of Immune Homeostasis. Front Immunol 2017; 8:605. [PMID: 28603524 PMCID: PMC5445144 DOI: 10.3389/fimmu.2017.00605] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 05/08/2017] [Indexed: 12/15/2022] Open
Abstract
The transcription factor forkhead box protein 3 (FOXP3) is an essential molecular marker of regulatory T cell (Treg) development in different microenvironments. Tregs are cells specialized in the suppression of inadequate immune responses and the maintenance of homeostatic tolerance. Studies have addressed and elucidated the role played by FOXP3 and Treg in countless autoimmune and infectious diseases as well as in more specific cases, such as cancer. Within this context, the present article reviews aspects of the immunoregulatory profile of FOXP3 and Treg in the management of immune homeostasis, including issues relating to pathology as well as immune tolerance.
Collapse
Affiliation(s)
- Leonn Mendes Soares Pereira
- Laboratório de Virologia, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil.,Programa de Pós-Graduação em Biologia de Agentes Infecciosos e Parasitários, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Samara Tatielle Monteiro Gomes
- Laboratório de Virologia, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil.,Programa de Pós-Graduação em Biologia de Agentes Infecciosos e Parasitários, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Ricardo Ishak
- Laboratório de Virologia, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | | |
Collapse
|
21
|
Salecan protected against concanavalin A-induced acute liver injury by modulating T cell immune responses and NMR-based metabolic profiles. Toxicol Appl Pharmacol 2017; 317:63-72. [PMID: 28109817 DOI: 10.1016/j.taap.2017.01.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/07/2017] [Accepted: 01/16/2017] [Indexed: 12/24/2022]
Abstract
Salecan, a water-soluble extracellular β-glucan produced by Agrobacterium sp. ZX09, has been reported to exhibit a wide range of biological effects. The aims of the present study were to investigate the protective effect of salecan against Concanavalin A (ConA)-induced hepatitis, a well-established animal model of immune-mediated liver injury, and to search for possible mechanisms. C57BL/6 mice were pretreated with salecan followed by ConA injection. Salecan treatment significantly reduced ConA-induced acute liver injury, and suppressed the expression and secretion of inflammatory cytokines including interferon (IFN)-γ, interleukin (IL)-6 and IL-1β in ConA-induced liver injury model. The high expression levels of chemokines and adhesion molecules such as MIP-1α, MIP-1β, ICAM-1, MCP-1 and RANTES in the liver induced by ConA were also down-regulated after salecan treatment. Salecan inhibited the infiltration and activation of inflammatory cells, especially T cells, in the liver induced by ConA. Moreover, salecan reversed the metabolic profiles of ConA-treated mice towards the control group by partly recovering the metabolic perturbations induced by ConA. Our results suggest the preventive and therapeutic potential of salecan in immune-mediated hepatitis.
Collapse
|
22
|
Wang L, Wang J, Liu Y, Wang B, Yang S, Yu Q, Roggendorf M, Lu M, Liu J, Yang D. Molecular cloning, characterization and expression analysis of TGF-β and receptor genes in the woodchuck model. Gene 2016; 595:1-8. [PMID: 27637515 DOI: 10.1016/j.gene.2016.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 08/22/2016] [Accepted: 09/12/2016] [Indexed: 12/16/2022]
Abstract
Transforming growth factor beta (TGF-β) is an important cytokine with pleiotropic regulatory functions in the immune system and in the responses against viral infections. TGF-β acts on a variety of immune cells through the cell surface TGF-β receptor (University of Duisburg-EssenTGFBR). The woodchuck has been used as a biomedical model for studies of obesity and energy balance, endocrine and metabolic function, cardiovascular, cerebrovascular and neoplastic disease. Woodchucks infected with woodchuck hepatitis virus (WHV) represent an informative animal model to study hepatitis B virus (HBV) infection. In this study, the cDNA sequences of woodchuck TGF-β1, TGF-β2, TGFBR1 and TGFBR2 were cloned, sequenced and characterized. The full-length TGFBR1 cDNA sequence consisted of 1305bp coding sequence (CDS) that encoded 434 amino acids with a molecular weight of 48.9kDa. The phylogenetic tree analysis revealed that the woodchuck TGF-β family genes had a closer genetic relationship with Ictidomys tridecemlineatus. One antibody with cross-reactivity to woodchuck TGFBR1 was identified by flow cytometry. Moreover, the expression of these genes were analyzed at the transcriptional level. The quantitative PCR analysis showed that the TGF-β family transcripts were constitutively expressed in many tissues tested. Altered expression levels of the TGF-β family transcripts in the liver of WHV infected woodchucks were observed. These results serve as a foundation for further insight into the role of the TGF-β family in viral hepatitis in woodchuck model. Our work also possesses the potential value for characterizing the TGF-β family in other related diseases, such as obesity-related diseases, metabolic disorder, cardiovascular disease and cancer.
Collapse
Affiliation(s)
- Lu Wang
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Junzhong Wang
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Yana Liu
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Baoju Wang
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Shangqing Yang
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Qing Yu
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Michael Roggendorf
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Germany
| | - Mengji Lu
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Germany
| | - Jia Liu
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| | - Dongliang Yang
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
23
|
Schrager LK, Izzo A, Velmurugan K. Immunopathogenesis of tuberculosis and novel mechanisms of vaccine activity. Tuberculosis (Edinb) 2016; 99 Suppl 1:S3-7. [DOI: 10.1016/j.tube.2016.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
24
|
Adar T, Edden Y, Shteingart S, Ben Ya’acov A, Mahamid M, Broide E, Shmorak S, Bar-Gil Shitrit A, Reinus C, Goldin E. Portal hypertension is associated with modulation of regulatory T cells. EUR J INFLAMM 2016; 14:40-47. [DOI: 10.1177/1721727x15622960] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Background: Portal hypertension is a complication of liver cirrhosis. The portal vein drains the spleen and the intestines, which are both rich in inflammatory mediators. Portal hypertension- induced stress within these organs that may result in pro-inflammatory changes. The association of these changes with regulatory T cells was not addressed before. Aim: Our aim is to investigate the involvement of some subsets of regulatory T cells in portal hypertension. Methods: In the current study we used the partial portal vein ligation model to demonstrate differences in the distribution of regulatory T cells within the portal vein and the inferior vena cava associated with portal hypertension. Results: We show that CD4+CD25+FoxP3+ regulatory T cells are significantly ( P <0.05) increased only in the inferior vena cava of partial portal vein ligation-rats. The development of portal hypertension was associated with the reversal of the distribution patterns in the portal vein and inferior vena cava for both CD4+ and CD8+ cells. We further show that in naïve rats CD4+IL17+ cells were significantly ( P <0.05) and specifically enriched in inferior vena cava compared to the portal vein. Conclusions: These novel findings support the involvement of regulatory T cells in the inflammatory signals accompanied with acute portal hypertension.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Constantin Reinus
- Department of Pathology, Shaare Zedek Medical Center, affiliated with the Hebrew University School of Medicine, Jerusalem, Israel
| | | |
Collapse
|
25
|
Zhang X, Feng M, Liu X, Bai L, Kong M, Chen Y, Zheng S, Liu S, Wan YJY, Duan Z, Han YP. Persistence of cirrhosis is maintained by intrahepatic regulatory T cells that inhibit fibrosis resolution by regulating the balance of tissue inhibitors of metalloproteinases and matrix metalloproteinases. Transl Res 2016; 169:67-79.e1-2. [PMID: 26613891 DOI: 10.1016/j.trsl.2015.10.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 10/07/2015] [Accepted: 10/29/2015] [Indexed: 12/26/2022]
Abstract
Fibrosis is the result of the abnormal accumulation of the extracellular matrix and ineffective clearance of fibroplasia. CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) are immunosuppressive lymphocytes that are highly expressed in the fibrotic tissues and peripheral blood of patients with cirrhosis or hepatocellular carcinoma. The role of Tregs in the progression of liver fibrosis is not well understood. Our experiments reveal that abundant of Tregs was scattered around sites of fibroplasia. Conversely, the depletion of Tregs promoted the resolution of liver fibrosis. As a consequence of Tregs depletion, the expression of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) was altered; mmp9 and timp1 were reduced, whereas mmp2 and mmp14 were enhanced. The mmp9/timp1, mmp13/timp1, and mmp14/timp2 ratios were significantly increased in association with fibrosis resolution. Kupffer cells (KCs) are the main source of MMP. We observed that when KCs were cocultured with Tregs, the Tregs were able to inhibit MMP expression of KCs even at a low ratio; and anti-transforming growth factor-β (TGF-β) significantly reversed the inhibition of Tregs on MMP. Meanwhile, we also found that after Tregs depletion, TGF-β levels decreased in the mice liver, unlike in fibrosis. Furthermore, double depletion of both KCs and Tregs did not cause fiber resolution in mice. Thus, our results demonstrate that the persistence of liver cirrhosis is maintained by increased Tregs in the sites of fibroplasia and the subsequent regulation of the MMP/TIMP balance and that the suppression of KC-mediated MMP expression contributed to the regulatory process.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Artificial Liver Treatment Center, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Min Feng
- Department of Liver Transplantation, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Xin Liu
- Artificial Liver Treatment Center, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Li Bai
- Artificial Liver Treatment Center, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Ming Kong
- Artificial Liver Treatment Center, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Yu Chen
- Artificial Liver Treatment Center, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Sujun Zheng
- Artificial Liver Treatment Center, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Shuang Liu
- Artificial Liver Treatment Center, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Yu-Jui Yvonne Wan
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis Health Systems, Sacramento, Calif
| | - Zhongping Duan
- Artificial Liver Treatment Center, Beijing YouAn Hospital, Capital Medical University, Beijing, China.
| | - Yuan-Ping Han
- The Center for Growth, Metabolism and Aging, The Key Laboratory for Bio-Resource and Eco-Environment, College of Life Science, and The National Key Laboratory of Biotherapy, Sichuan University, Chengdu, China.
| |
Collapse
|
26
|
an Haack I, Derkow K, Riehn M, Rentinck MN, Kühl AA, Lehnardt S, Schott E. The Role of Regulatory CD4 T Cells in Maintaining Tolerance in a Mouse Model of Autoimmune Hepatitis. PLoS One 2015; 10:e0143715. [PMID: 26599014 PMCID: PMC4658037 DOI: 10.1371/journal.pone.0143715] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 11/09/2015] [Indexed: 11/19/2022] Open
Abstract
Background The role of regulatory CD4 T cells (Treg) in immune-mediated liver disease is still under debate. It remains disputed whether Treg suppress T cell-mediated hepatitis in vivo and whether hepatic regulatory T cells are functional in patients with autoimmune hepatitis. Methods We used TF-OVA mice, which express ovalbumin in hepatocytes, to investigate the impact of Treg in a model of autoimmune hepatitis. Treg isolated from inflamed livers of TF-OVA mice were tested for their functionality in vitro. By employing double transgenic TF-OVAxDEREG (DEpletion of REGulatory T cells) mice we analyzed whether Treg-depletion aggravates autoimmune inflammation in the liver in vivo. Results CD25+Foxp3+ CD4 T cells accumulated in the liver in the course of CD8 T cell-mediated hepatitis. Treg isolated from inflamed livers were functional to suppress CD8 T-cell proliferation in vitro. Depletion of Treg in TF-OVAxDEREG mice dramatically amplified T cell-mediated hepatitis. Repeated administration of antigen-specific CD8 T cells led to a second wave of inflammation only after depletion of Treg. Conclusion Our data add to the evidence for an important role of Treg in autoimmune hepatitis and show that Treg reduce the severity of T-cell mediated hepatitis in vivo. They constitute a key immune cell population that actively maintains a tolerogenic milieu in the liver and protects the liver against repeated inflammatory challenges.
Collapse
Affiliation(s)
- Ira an Haack
- Dept. of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Katja Derkow
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Mathias Riehn
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research Braunschweig, Braunschweig, Germany
| | - Marc-Nicolas Rentinck
- Dept. of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Anja A. Kühl
- Dept. of Medicine I for Gastroenterology, Infectious Diseases and Rheumatology/Research Center ImmunoSciences, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Seija Lehnardt
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité Universitätsmedizin Berlin, Berlin, Germany
- Cluster of Excellence NeuroCure, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Eckart Schott
- Dept. of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
27
|
Lee YS, Yi HS, Suh YG, Byun JS, Eun HS, Kim SY, Seo W, Jeong JM, Choi WM, Kim MH, Kim JH, Park KG, Jeong WI. Blockade of Retinol Metabolism Protects T Cell-Induced Hepatitis by Increasing Migration of Regulatory T Cells. Mol Cells 2015; 38:998-1006. [PMID: 26537191 PMCID: PMC4673415 DOI: 10.14348/molcells.2015.0218] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 08/30/2015] [Accepted: 08/31/2015] [Indexed: 12/12/2022] Open
Abstract
Retinols are metabolized into retinoic acids by alcohol dehydrogenase (ADH) and retinaldehyde dehydrogenase (Raldh). However, their roles have yet to be clarified in hepatitis despite enriched retinols in hepatic stellate cells (HSCs). Therefore, we investigated the effects of retinols on Concanavalin A (Con A)-mediated hepatitis. Con A was injected into wild type (WT), Raldh1 knock-out (Raldh1(-/-)), CCL2(-/-) and CCR2(-/-) mice. For migration study of regulatory T cells (Tregs), we used in vivo and ex vivo adoptive transfer systems. Blockade of retinol metabolism in mice given 4-methylpyrazole, an inhibitor of ADH, and ablated Raldh1 gene manifested increased migration of Tregs, eventually protected against Con A-mediated hepatitis by decreasing interferon-γ in T cells. Moreover, interferon-γ treatment increased the expression of ADH3 and Raldh1, but it suppressed that of CCL2 and IL-6 in HSCs. However, the expression of CCL2 and IL-6 was inversely increased upon the pharmacologic or genetic ablation of ADH3 and Raldh1 in HSCs. Indeed, IL-6 treatment increased CCR2 expression of Tregs. In migration assay, ablated CCR2 in Tregs showed reduced migration to HSCs. In adoptive transfer of Tregs in vivo and ex vivo, Raldh1-deficient mice showed more increased migration of Tregs than WT mice. Furthermore, inhibited retinol metabolism increased survival rate (75%) compared with that of the controls (25%) in Con A-induced hepatitis. These results suggest that blockade of retinol metabolism protects against acute liver injury by increased Treg migration, and it may represent a novel therapeutic strategy to control T cell-mediated acute hepatitis.
Collapse
Affiliation(s)
- Young-Sun Lee
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
- Department of Internal Medicine, Korea University College of Medicine, Seoul 136-705,
Korea
| | - Hyon-Seung Yi
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 305-764,
Korea
| | - Yang-Gun Suh
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408,
Korea
| | - Jin-Seok Byun
- Department of Oral Medicine, School of Dentistry, Kyungpook National University, Daegu 41566,
Korea
| | - Hyuk Soo Eun
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - So Yeon Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Wonhyo Seo
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Jong-Min Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Won-Mook Choi
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Myung-Ho Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Ji Hoon Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul 136-705,
Korea
| | - Keun-Gyu Park
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu 41566,
Korea
| | - Won-Il Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| |
Collapse
|
28
|
Treg Cell Differentiation: From Thymus to Peripheral Tissue. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 136:175-205. [PMID: 26615097 DOI: 10.1016/bs.pmbts.2015.07.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Regulatory T cells (Tregs) are crucial mediators of self-tolerance in the periphery. They differentiate in the thymus, where interactions with thymus-resident antigen-presenting cells, an instructive cytokine milieu, and stimulation of the T cell receptor lead to the selection into the Treg lineage and the induction of Foxp3 gene expression. Once mature, Treg cells leave the thymus and migrate into either the secondary lymphoid tissues, e.g., lymph nodes and spleen, or peripheral nonlymphoid tissues. There is growing evidence that Treg cells go beyond the classical modulation of immune responses and also play important functional roles in nonlymphoid peripheral tissues. In this review, we summarize recent findings about the thymic Treg lineage differentiation as well as the further specialization of Treg cells in the secondary lymphoid and in the peripheral nonlymphoid organs.
Collapse
|
29
|
Dietary n-3 PUFA Protects Mice from Con A Induced Liver Injury by Modulating Regulatory T Cells and PPAR-γ Expression. PLoS One 2015; 10:e0132741. [PMID: 26177196 PMCID: PMC4503783 DOI: 10.1371/journal.pone.0132741] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 06/17/2015] [Indexed: 01/01/2023] Open
Abstract
Background Dietary n-3 polyunsaturated fatty acids (PUFA) exert anti-inflammatory and immunoregulatory effects through down-regulating the innate and adoptive immune response. However, the effect of dietary n-3 PUFA on CD4+CD25+ regulatory T cells (Tregs) is unclear. Aims The current study was to examine the relationship between n-3 PUFA and Tregs as well as their immunoregulatory effect in immune-mediated liver injury. Methods The mice model feeding with n-3 PUFA-enriched diet was established and Tregs were analyzed. Effect of docosahexaenoic acid (DHA) on Tregs proliferation and induction was determined in vitro. The potential immunotherapeutic effect of dietary n-3 PUFA was investigated through Con A-induced hepatitis model. Results Long-term administration of dietary n-3 PUFA significantly increased hepatic Tregs and modulated their phenotype. n-3 PUFA or DHA directly increased natural Tregs (nTreg) proliferation but didn’t increase inducible Tregs (iTreg). In addition, the expression of peroxisome proliferator activated receptor gamma (PPAR-γ), transforming growth factor β (TGF-β) and interleukin (IL)-10 were significantly up-regulated in n-3 PUFA-enriched diet-fed mice. Finally, n-3 PUFA-enriched diet alleviated liver injury induced by Con A and down-regulated pro-inflammatory cytokines expression, accompanied by increased PPAR-γ expression. Conclusion Dietary n-3 PUFA enhanced Tregs generation through up-regulating PPAR-γ and TGF-β expression, and protected mice from Con A-induced liver injury. This finding provides a promising potential therapeutic method in treating inflammatory and autoimmune disease.
Collapse
|
30
|
Chen Y, Sun R, Wu X, Cheng M, Wei H, Tian Z. CD4+CD25+ Regulatory T Cells Inhibit Natural Killer Cell Hepatocytotoxicity of Hepatitis B Virus Transgenic Mice via Membrane-Bound TGF-β and OX40. J Innate Immun 2015; 8:30-42. [PMID: 26067079 DOI: 10.1159/000431150] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 05/05/2015] [Indexed: 12/13/2022] Open
Abstract
CD4+CD25+ regulatory T cells (Tregs) are involved in the regulation of physiological and pathological hepatic immune responses, but the roles are not well explored in natural killer (NK) cell-mediated liver diseases. In this study, using the NK cell-mediated oversensitive liver injury model of hepatitis B virus transgenic (HBs-Tg) mice triggered by a low dose of concanavalin A, it was observed that an increased number of CD4+CD25+Foxp3+ Tregs were accumulated in the liver, along with the recovery of liver injury. Adoptive transfer of hepatic Tregs from HBs-Tg mice but not wild B6 mice could significantly attenuate the oversensitive liver injury via inhibiting liver accumulation and decreasing NK cell group 2D-mediated activation of NK cells in the recipient HBs-Tg mice. Furthermore, upregulated expression of membrane-bound TGF-β (mTGF-β) and OX40 on hepatic Tregs were demonstrated to account for inhibiting the NK cell-mediated hepatic injury in HBs-Tg mice through cell-cell contact, confirmed by antibody blockade and cell Transwell experiments in vivo and in vitro. Our findings for the first time indicated that CD4+CD25+ Tregs directly suppressed NK cell-mediated hepatocytotoxicity through mTGF-β and OX40/OX40L interaction in a cell-cell contact manner in HBV-associated liver disease.
Collapse
Affiliation(s)
- Yongyan Chen
- Department of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, PR China
| | | | | | | | | | | |
Collapse
|
31
|
Volarevic V, Markovic BS, Bojic S, Stojanovic M, Nilsson U, Leffler H, Besra GS, Arsenijevic N, Paunovic V, Trajkovic V, Lukic ML. Gal-3 regulates the capacity of dendritic cells to promote NKT-cell-induced liver injury. Eur J Immunol 2014; 45:531-43. [DOI: 10.1002/eji.201444849] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 09/09/2014] [Accepted: 10/28/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Vladislav Volarevic
- Centre for Molecular Medicine and Stem Cell Research; Faculty of Medical Sciences; University of Kragujevac; Kragujevac Serbia
| | - Bojana Simovic Markovic
- Centre for Molecular Medicine and Stem Cell Research; Faculty of Medical Sciences; University of Kragujevac; Kragujevac Serbia
| | - Sanja Bojic
- Centre for Molecular Medicine and Stem Cell Research; Faculty of Medical Sciences; University of Kragujevac; Kragujevac Serbia
| | - Maja Stojanovic
- Centre for Molecular Medicine and Stem Cell Research; Faculty of Medical Sciences; University of Kragujevac; Kragujevac Serbia
| | - Ulf Nilsson
- Centre for Analysis and Synthesis; Department of Chemistry; Lund University; Lund Sweden
| | - Hakon Leffler
- Section MIG; Department of Laboratory Medicine; Lund University; Lund Sweden
| | | | - Nebojsa Arsenijevic
- Centre for Molecular Medicine and Stem Cell Research; Faculty of Medical Sciences; University of Kragujevac; Kragujevac Serbia
| | - Verica Paunovic
- Institute for Microbiology and Immunology; School of Medicine; University of Belgrade; Belgrade Serbia
| | - Vladimir Trajkovic
- Institute for Microbiology and Immunology; School of Medicine; University of Belgrade; Belgrade Serbia
| | - Miodrag L. Lukic
- Centre for Molecular Medicine and Stem Cell Research; Faculty of Medical Sciences; University of Kragujevac; Kragujevac Serbia
| |
Collapse
|
32
|
CD4(+)Foxp3(+) Tregs protect against innate immune cell-mediated fulminant hepatitis in mice. Mol Immunol 2014; 63:420-7. [PMID: 25315497 DOI: 10.1016/j.molimm.2014.09.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 09/23/2014] [Accepted: 09/23/2014] [Indexed: 01/03/2023]
Abstract
UNLABELLED Foxp3(+) Tregs play important roles in maintaining homeostasis by suppressing excessive immune responses that result in serious tissue damage; yet, it is largely unknown about the impact of Tregs on innate immune cells in hepatitis models in vivo. In this study, we examined the effect of hepatic Tregs on innate immune-mediated liver injury by using the murine model of polyI:C and d-galactosamine (d-GalN)-induced hepatitis. Administration of polyI:C/d-GalN increased the number of CD4(+)Foxp3(+) Tregs in the liver. Depletion of Tregs leaded to higher levels of proinflammatory cytokine expression and severer liver injury, whereas adoptive transfer of Foxp3(+) Tregs attenuated liver injury in polyI:C/d-GalN-treated mice. In addition, depletion of Tregs leaded to a reduction in TGF-β and IL-10 expression in polyI:C/d-GalN-treated mice. Both of these cytokines were important for suppression of polyI:C/d-GalN-induced liver injury. TGF-β was derived from Tregs. IL-10 was derived from active Kupffer cells, and coincubation of Kupffer cells with Tregs increased IL-10 secretion. Furthermore, TGF-β blockade abrogated Treg-mediated suppression of proinflammatory cytokine production by innate immune cell in vitro. CONCLUSION CD4(+)Foxp3(+) Tregs modify innate immune responses in polyI:C/d-GalN-induced fulminant hepatitis via producing TGF-β and enhancing IL-10 secretion by Kupffer cells.
Collapse
|
33
|
Mahalingam J, Lin CY, Chiang JM, Su PJ, Chu YY, Lai HY, Fang JH, Huang CT, Lin YC. CD4⁺ T cells expressing latency-associated peptide and Foxp3 are an activated subgroup of regulatory T cells enriched in patients with colorectal cancer. PLoS One 2014; 9:e108554. [PMID: 25268580 PMCID: PMC4182495 DOI: 10.1371/journal.pone.0108554] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 08/24/2014] [Indexed: 01/29/2023] Open
Abstract
Latency-associated peptide (LAP) - expressing regulatory T cells (Tregs) are important for immunological self-tolerance and immune homeostasis. In order to investigate the role of LAP in human CD4+Foxp3+ Tregs, we designed a cross-sectional study that involved 42 colorectal cancer (CRC) patients. The phenotypes, cytokine-release patterns, and suppressive ability of Tregs isolated from peripheral blood and tumor tissues were analyzed. We found that the population of LAP-positive CD4+Foxp3+ Tregs significantly increased in peripheral blood and cancer tissues of CRC patients as compared to that in the peripheral blood and tissues of healthy subjects. Both LAP+ and LAP− Tregs had a similar effector/memory phenotype. However, LAP+ Tregs expressed more effector molecules, including tumor necrosis factor receptor II, granzyme B, perforin, Ki67, and CCR5, than their LAP− negative counterparts. The in vitro immunosuppressive activity of LAP+ Tregs, exerted via a transforming growth factor-β–mediated mechanism, was more potent than that of LAP− Tregs. Furthermore, the enrichment of LAP+ Treg population in peripheral blood mononuclear cells (PBMCs) of CRC patients correlated with cancer metastases. In conclusion, we found that LAP+ Foxp3+ CD4+ Treg cells represented an activated subgroup of Tregs having more potent regulatory activity in CRC patients. The increased frequency of LAP+ Tregs in PBMCs of CRC patients suggests their potential role in controlling immune response to cancer and presents LAP as a marker of tumor-specific Tregs in CRC patients.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/immunology
- Case-Control Studies
- Colorectal Neoplasms/diagnosis
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/immunology
- Colorectal Neoplasms/pathology
- Female
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Gene Expression Regulation, Neoplastic
- Granzymes/genetics
- Granzymes/immunology
- Humans
- Immune Tolerance
- Immunologic Memory
- Ki-67 Antigen/genetics
- Ki-67 Antigen/immunology
- Lymphatic Metastasis
- Male
- Middle Aged
- Peptides/genetics
- Peptides/immunology
- Perforin/genetics
- Perforin/immunology
- Protein Precursors/genetics
- Protein Precursors/immunology
- Receptors, CCR5/genetics
- Receptors, CCR5/immunology
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Receptors, Tumor Necrosis Factor, Type II/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/immunology
Collapse
Affiliation(s)
- Jayashri Mahalingam
- College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
- Department of Gastroenterology-Hepatology, Linkou Medical Centre, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
| | - Chun-Yen Lin
- College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
- Department of Gastroenterology-Hepatology, Linkou Medical Centre, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
| | - Jy-Ming Chiang
- College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
- Colorectal Surgery Section, Department of Surgery, Linkou Medical Centre, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
| | - Po-Jung Su
- Department of Hematology-Oncology, Linkou Medical Centre, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
| | - Yu-Yi Chu
- Department of Hematology-Oncology, Linkou Medical Centre, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
| | - Hsin-Yi Lai
- College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Jian-He Fang
- Department of Gastroenterology-Hepatology, Linkou Medical Centre, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
| | - Ching-Tai Huang
- College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
- Department of Infectious Diseases, Linkou Medical Centre, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
| | - Yung-Chang Lin
- College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
- Department of Hematology-Oncology, Linkou Medical Centre, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
- * E-mail:
| |
Collapse
|
34
|
Czaja AJ. Review article: chemokines as orchestrators of autoimmune hepatitis and potential therapeutic targets. Aliment Pharmacol Ther 2014; 40:261-79. [PMID: 24890045 DOI: 10.1111/apt.12825] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 04/10/2014] [Accepted: 05/14/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND Chemokines contribute to the pathogenesis of autoimmune hepatitis by directing the migration and positioning of inflammatory and immune cells within the liver. AIM Describe the liver-infiltrating effector cell populations in autoimmune hepatitis, indicate the chemokines that influence their migration, describe the role of chemokines in hepatic fibrosis and identify chemokine-directed treatment opportunities. METHODS Studies cited in Pub Med from 1972 to 2014 for autoimmune hepatitis, chemokines in liver disease, pathogenesis of autoimmune hepatitis and chemokine therapy were selected. RESULTS T helper type 17 lymphocytes expressing CXCR3 and CCR6 are attracted to the liver by the secretion of CXCL9, CXCL10 and CXCL11. These cells recruit pro-inflammatory T helper type 1 lymphocytes expressing CXCR3 and CCR5 by secreting CXCL10. Resident natural killer T cells expressing CXCR6 migrate in response to the local secretion of CXCL16, and they modulate the inflammatory response. T helper type 2 lymphocytes expressing CCR4 are attracted by CCL17 and CCL22, and they dampen the expansion of pro-inflammatory cells. Regulatory T cells expressing CXCR3 are attracted by the secretion of CXCL9, and they help dampen the pro-inflammatory responses. CCL2, CCL3, CCL5, CXCL4, CXCL10 and CXCL16 promote fibrosis by activating or attracting hepatic stellate cells, and CX3CL1 may prevent fibrosis by affecting the apoptosis of monocytes. CONCLUSIONS Chemokines are requisites for mobilising, directing and positioning the effector cells in immune-mediated liver disease. They are feasible therapeutic targets in autoimmune hepatitis, and the evaluation of monoclonal antibodies that neutralise the pro-inflammatory ligands or designer peptides that block receptor activity are investigational opportunities.
Collapse
Affiliation(s)
- A J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
35
|
The effect of citrus peel extracts on cytokines levels and T regulatory cells in acute liver injury. BIOMED RESEARCH INTERNATIONAL 2014; 2014:127879. [PMID: 25126542 PMCID: PMC4121996 DOI: 10.1155/2014/127879] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 06/13/2014] [Accepted: 06/24/2014] [Indexed: 01/26/2023]
Abstract
Background. T cell-mediated immune responses contribute to the hepatocellular injury during autoimmune hepatitis, viral infection, and hepatotoxins. Pharmacological compounds regulating immune responses are suitable candidates for prevention/treatment of this pathology. Therefore, the main aim of this study was to define the effects of antioxidant, anti-inflammatory mixture of citrus peel extract (CPE) on the immune-mediated liver injury. Methods. The influence of CPE on liver injury was determined by the activity of transaminases in plasma and the histological changes. Anti-inflammatory and antioxidant effects were studied by measuring frequency of T regulatory cells (Tregs), cytokines (TNF-α, IL-10, and IFN-γ), and nitric oxide levels. Results. The CPE application notably prevents development of liver injury through decreasing levels of both cytokines (TNF-alpha, INF) and regulatory T cells and increasing levels of IL-10. CPE injection also diminished the serum NO, which in turn resulted in evident reduction of the liver damage. Conclusion. Our findings represent the primary preclinical data indicating that the CPE in vivo could ameliorate Con A induced hepatitis. The low dose of CPE most likely can be used for the treatment of the T cell-mediated liver injury as in autoimmune hepatitis, alcoholic hepatitis, and chronic viral hepatitis.
Collapse
|
36
|
Burghardt S, Claass B, Erhardt A, Karimi K, Tiegs G. Hepatocytes induce Foxp3⁺ regulatory T cells by Notch signaling. J Leukoc Biol 2014; 96:571-7. [PMID: 24970859 DOI: 10.1189/jlb.2ab0613-342rr] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The liver plays a pivotal role in maintaining immunological tolerance, although the exact molecular mechanism is still largely unknown. The induction of systemic tolerance by liver resident APCs has been attributed to peripheral deletion and to the induction of Tregs. HCs, the parenchymal cells in the liver, could function as nonprofessional APCs and interact and establish cell-cell contact with T lymphocytes. We hypothesized that HCs from healthy or regenerated livers may contribute to induction of functional Tregs. Here, we show that murine HCs induced Foxp3(+) Tregs within CD4(+) T cells in vitro, which increased in the presence of TGF-β. Interestingly, a further Foxp3(+) Treg expansion was observed if HCs were isolated from regenerated livers. Additionally, the induction of Foxp3(+) Tregs was associated with the Notch signaling pathway, as the ability of HCs to enhance Foxp3 was abolished by γ-secretase inhibition. Furthermore, HC-iTregs showed ability to suppress the proliferative response of CD4(+) T cells to anti-CD3 stimulation in vitro. Thus, HCs may play a pivotal role in the induction of tolerance via Notch-mediated conversion of CD4(+) T cells into Foxp3(+) Tregs upon TCR stimulation.
Collapse
Affiliation(s)
- Sven Burghardt
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Benjamin Claass
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Annette Erhardt
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Khalil Karimi
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| |
Collapse
|
37
|
Lu L, Feng M, Gu J, Xia Z, Zhang H, Zheng S, Duan Z, Hu R, Wang J, Shi W, Ji C, Shen Y, Chen G, Zheng SG, Han YP. Restoration of intrahepatic regulatory T cells through MMP-9/13-dependent activation of TGF-β is critical for immune homeostasis following acute liver injury. J Mol Cell Biol 2013; 5:369-79. [PMID: 24280647 PMCID: PMC3841112 DOI: 10.1093/jmcb/mjt042] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 07/24/2013] [Accepted: 08/08/2013] [Indexed: 12/24/2022] Open
Abstract
During the acute liver injury, immune responses are provoked into eliciting inflammation in the acute phase. In the healing phase, the inflammation is terminated for wound healing and restoration of immune homeostasis. In this study, we sought to address how regulatory T cells (Tregs) are involved in the progression of liver injury and repair. In the acute phase, intrahepatic Tregs (CD4(+)FoxP3(+)Helios(+)) diminished promptly through apoptosis, which was followed by inflammation and tissue injury. In the healing phase, a new subset of Tregs (CD4(+)Foxp3(+)Helios(-)) was generated in correlation with the matrix metalloproteinase (MMP) cascade and transforming growth factor-beta (TGF-β) activation that were manifested mainly by hepatic stellate cells. Moreover, the induction of induced Tregs and wound healing were both impaired in mice lacking TGF-β signaling or MMPs. The depletion of induced Tregs also impeded wound healing for tissue repair. Together, this study demonstrates the mechanism that the loss of nTregs through apoptosis in the acute phase may facilitate inflammation, while regenerated Tregs through MMP9/13-dependent activation of TGF-β in the healing phase are critical to terminate inflammation and allow for wound healing.
Collapse
Affiliation(s)
- Ling Lu
- Department of Liver Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Min Feng
- Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jia Gu
- Department of Liver Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zanxian Xia
- School of Biological Sciences and Technology, Central South University, Changsha 410013, China
| | - Hongjun Zhang
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Sujun Zheng
- Beijing YouAn Hospital, Capital Medical University, Beijing 100069, China
| | - Zhongping Duan
- Beijing YouAn Hospital, Capital Medical University, Beijing 100069, China
| | - Richard Hu
- Olive View-UCLA Medical Center, Los Angeles, CA 91342, USA
| | - Julie Wang
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Wei Shi
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cheng Ji
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yi Shen
- Penn State University Hershey College of Medicine, Hershey, PA 17033, USA
| | - Guihua Chen
- Department of Liver Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Song Guo Zheng
- Penn State University Hershey College of Medicine, Hershey, PA 17033, USA
- Tongji University Shanghai East Hospital, Institute of Immunology, Shanghai 200120, China
| | - Yuan-Ping Han
- The Center for Growth, Metabolism and Aging, the Key Laboratory for Bio-Resource and Eco-Environment, and the National Key Laboratory of Biotherapy, Sichuan University, Chengdu 610064, China
| |
Collapse
|
38
|
Abstract
The liver is the largest organ in the body and is generally regarded by nonimmunologists as having little or no lymphoid function. However, such is far from accurate. This review highlights the importance of the liver as a lymphoid organ. Firstly, we discuss experimental data surrounding the role of liver as a lymphoid organ. The liver facilitates tolerance rather than immunoreactivity, which protects the host from antigenic overload of dietary components and drugs derived from the gut and it is instrumental to fetal immune tolerance. Loss of liver tolerance leads to autoaggressive phenomena, which if not controlled by regulatory lymphoid populations, may lead to the induction of autoimmune liver diseases. Liver-related lymphoid subpopulations also act as critical antigen-presenting cells. The study of the immunological properties of liver and delineation of the microenvironment of the intrahepatic milieu in normal and diseased livers provides a platform to understand the hierarchy of a series of detrimental events that lead to immune-mediated destruction of the liver and the rejection of liver allografts. The majority of emphasis within this review will be on the normal mononuclear cell composition of the liver. However, within this context, we will discuss selected, but not all, immune-mediated liver disease and attempt to place these data in the context of human autoimmunity.
Collapse
Affiliation(s)
- Dimitrios P Bogdanos
- Institute of Liver Studies, Transplantation Immunology and Mucosal Biology, King's College London School of Medicine at King's College Hospital, London, UK
| | | | | |
Collapse
|
39
|
Regulatory T-cell directed therapies in liver diseases. J Hepatol 2013; 59:1127-34. [PMID: 23727305 DOI: 10.1016/j.jhep.2013.05.034] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 05/17/2013] [Accepted: 05/22/2013] [Indexed: 01/07/2023]
|
40
|
Lehtimäki S, Lahesmaa R. Regulatory T Cells Control Immune Responses through Their Non-Redundant Tissue Specific Features. Front Immunol 2013; 4:294. [PMID: 24069022 PMCID: PMC3780303 DOI: 10.3389/fimmu.2013.00294] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 09/07/2013] [Indexed: 01/11/2023] Open
Abstract
Regulatory T cells (Treg) are needed in the control of immune responses and to maintain immune homeostasis. Of this subtype of regulatory lymphocytes, the most potent are Foxp3 expressing CD4+ T cells, which can be roughly divided into two main groups; natural Treg cells (nTreg), developing in the thymus, and induced or adaptive Treg cells (iTreg), developing in the periphery from naïve, conventional T cells. Both nTreg cells and iTreg cells have their own, non-redundant roles in the immune system, with nTreg cells mainly maintaining tolerance toward self-structures, and iTreg developing in response to externally delivered antigens or commensal microbes. In addition, Treg cells acquire tissue specific features and are adapted to function in the tissue they reside. This review will focus on some specific features of Treg cells in different compartments of the body.
Collapse
Affiliation(s)
- Sari Lehtimäki
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University , Turku , Finland
| | | |
Collapse
|
41
|
Zhang B, Hu M, Zhang P, Cao H, Wang Y, Wang Z, Su T. BAFF promotes regulatory T-cell apoptosis and blocks cytokine production by activating B cells in primary biliary cirrhosis. Braz J Med Biol Res 2013; 46:433-9. [PMID: 23681290 PMCID: PMC3854395 DOI: 10.1590/1414-431x20132665] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Accepted: 03/11/2013] [Indexed: 11/22/2022] Open
Abstract
Primary biliary cirrhosis (PBC) is a chronic and slowly progressive cholestatic
liver disease of autoimmune etiology. A number of questions regarding its
etiology are unclear. CD4+CD25+ regulatory T cells (Tregs) play a
critical role in self-tolerance and, for unknown reasons, their relative number
is reduced in PBC patients. B-cell-activating factor (BAFF) is a key survival
factor during B-cell maturation and its concentration is increased in peripheral
blood of PBC patients. It has been reported that activated B cells inhibit Treg
cell proliferation and there are no BAFF receptors on Tregs. Therefore, we
speculated that excessive BAFF may result in Treg reduction via B cells. To
prove our hypothesis, we isolated Tregs and B cells from PBC and healthy donors.
BAFF and IgM concentrations were then analyzed by ELISA and CD40, CD80, CD86,
IL-10, and TGF-β expression in B cells and Tregs were measured by flow
cytometry. BAFF up-regulated CD40, CD80, CD86, and IgM expression in B cells.
However, BAFF had no direct effect on Treg cell apoptosis and cytokine
secretion. Nonetheless, we observed that BAFF-activated B cells could induce
Treg cell apoptosis and reduce IL-10 and TGF-β expression. We also showed
that BAFF-activated CD4+ T cells had no effect on Treg apoptosis.
Furthermore, we verified that bezafibrate, a hypolipidemic drug, can inhibit
BAFF-induced Treg cell apoptosis. In conclusion, BAFF promotes Treg cell
apoptosis and inhibits cytokine production by activating B cells in PBC
patients. The results of this study suggest that inhibition of BAFF activation
is a strategy for PBC treatment.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Hepatology, Wuxi Infectious Diseases Hospital, Wuxi, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|
42
|
Li F, Tian Z. The liver works as a school to educate regulatory immune cells. Cell Mol Immunol 2013; 10:292-302. [PMID: 23604044 DOI: 10.1038/cmi.2013.7] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 02/21/2013] [Indexed: 12/24/2022] Open
Abstract
Because of its unique blood supply, the liver maintains a special local immune tolerogenic microenvironment. Moreover, the liver can impart this immune tolerogenic effect on other organs, thus inducing systemic immune tolerance. The network of hepatic regulatory cells is an important mechanism underlying liver tolerance. Many types of liver-resident antigen-presenting cells (APCs) have immune regulatory function, and more importantly, they can also induce the differentiation of circulating immune cells into regulatory cells to further extend systemic tolerance. Thus, the liver can be seen as a type of 'school', where liver APCs function as 'teachers' and circulating immune cells function as 'students.'
Collapse
Affiliation(s)
- Fenglei Li
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | | |
Collapse
|
43
|
Milovanovic M, Volarevic V, Radosavljevic G, Jovanovic I, Pejnovic N, Arsenijevic N, Lukic ML. IL-33/ST2 axis in inflammation and immunopathology. Immunol Res 2012; 52:89-99. [PMID: 22392053 DOI: 10.1007/s12026-012-8283-9] [Citation(s) in RCA: 193] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Interleukin-33 (IL-33), a member of the IL-1 family of cytokines, binds to its plasma membrane receptor, heterodimeric complex consisted of membrane-bound ST2L and IL-1R accessory protein, inducing NFkB and MAPK activation. IL-33 exists as a nuclear precursor and may act as an alarmin, when it is released after cell damage or as negative regulator of NFκB gene transcription, when acts in an intracrine manner. ST2L is expressed on several immune cells: Th2 lymphocytes, NK, NKT and mast cells and on cells of myeloid lineage: monocytes, dendritic cells and granulocytes. IL-33/ST2 axis can promote both Th1 and Th2 immune responses depending on the type of activated cell and microenvironment and cytokine network in damaged tissue. We previously described and discuss here the important role of IL-33/ST2 axis in experimental models of type 1 diabetes, experimental autoimmune encephalomyelitis, fulminant hepatitis and breast cancer. We found that ST2 deletion enhance the development of T cell-mediated autoimmune disorders, EAE and diabetes mellitus type I. Disease development was accompanied by dominantly Th1/Th17 immune response but also higher IL-33 production, which suggest that IL-33 in receptor independent manner could promote the development of inflammatory autoreactive T cells. IL-33/ST2 axis has protective role in Con A hepatitis. ST2-deficient mice had more severe hepatitis with higher influx of inflammatory cells in liver and dominant Th1/Th17 systemic response. Pretreatment of mice with IL-33 prevented Con A-induced liver damage through prevention of apoptosis of hepatocytes and Th2 amplification. Deletion of IL-33/ST2 axis enhances cytotoxicity of NK cells, production of IFN-γ in these cells and systemic production of IFN-γ, IL-17 and TNF-α, which leads to attenuated tumor growth. IL-33 treatment of tumor-bearing mice suppresses activity of NK cells, dendritic cell maturation and enhances alternative activation of macrophages. In conclusion, we observed that IL-33 has attenuated anti-inflammatory effects in T cell-mediated responses and that both IL-33 and ST2 could be further explored as potential therapeutic targets in treatment of immune-mediated diseases.
Collapse
Affiliation(s)
- Marija Milovanovic
- Faculty of Medicine, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | | | | | | | | | | | | |
Collapse
|
44
|
Li J, Qiu SJ, She WM, Wang FP, Gao H, Li L, Tu CT, Wang JY, Shen XZ, Jiang W. Significance of the balance between regulatory T (Treg) and T helper 17 (Th17) cells during hepatitis B virus related liver fibrosis. PLoS One 2012; 7:e39307. [PMID: 22745730 PMCID: PMC3380028 DOI: 10.1371/journal.pone.0039307] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 05/18/2012] [Indexed: 12/19/2022] Open
Abstract
Background Hepatitis B virus-related liver fibrosis (HBV-LF) always progresses from inflammation to fibrosis. However, the relationship between these two pathological conditions is not fully understood. Here, it is postulated that the balance between regulatory T (Treg) cells and T helper 17 (Th17) cells as an indicator of inflammation may predict fibrosis progression of HBV-LF. Methodology/Principal Findings The frequencies and phenotypes of peripheral Treg and Th17 cells of seventy-seven HBeAg-positive chronic hepatitis B (CHB) patients who underwent liver biopsies and thirty healthy controls were determined by flow cytometry. In the periphery of CHB patients, both Treg and Th17 frequencies were significantly increased and correlated, and a lower Treg/Th17 ratio always indicated more liver injury and fibrosis progression. To investigate exact effects of Treg and Th17 cells during HBV-LF, a series of in vitro experiments were performed using purified CD4+, CD4+CD25+, or CD4+CD25− cells from the periphery, primary human hepatic stellate cells (HSCs) isolated from healthy liver specimens, human recombinant interleukin (IL)-17 cytokine, anti-IL-17 antibody and HBcAg. In response to HBcAg, CD4+CD25+ cells significantly inhibited cell proliferation and cytokine production (especially IL-17 and IL-22) by CD4+CD25− cells in cell-contact and dose-dependent manners. In addition, CD4+ cells from CHB patients, compared to those from HC subjects, dramatically promoted proliferation and activation of human HSCs. Moreover, in a dramatically dose-dependent manner, CD4+CD25+ cells from CHB patients inhibited, whereas recombinant IL-17 response promoted the proliferation and activation of HSCs. Finally, in vivo evidence about effects of Treg/Th17 balance during liver fibrosis was obtained in concanavalin A-induced mouse fibrosis models via depletion of CD25+ or IL-17+ cells, and it’s observed that CD25 depletion promoted, whereas IL-17 depletion, alleviated liver injury and fibrosis progression. Conclusions/Significance The Treg/Th17 balance might influence fibrosis progression in HBV-LF via increase of liver injury and promotion of HSCs activation.
Collapse
Affiliation(s)
- Jing Li
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuang-Jian Qiu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei-Min She
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fu-Ping Wang
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hong Gao
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Li
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuan-Tao Tu
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ji-Yao Wang
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xi-Zhong Shen
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Jiang
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
- * E-mail:
| |
Collapse
|
45
|
Uibo R, Kisand K, Yang CY, Gershwin ME. Primary biliary cirrhosis: a multi-faced interactive disease involving genetics, environment and the immune response. APMIS 2012; 120:857-71. [PMID: 23009110 DOI: 10.1111/j.1600-0463.2012.02914.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 04/10/2012] [Indexed: 12/12/2022]
Abstract
Primary biliary cirrhosis (PBC) is considered a model autoimmune disease based on several features, including the presence of a highly directed and very specific immune response to mitochondrial autoantigens, a female predominance, a targeted destruction of the biliary epithelium, and homogeneity between patients. It is essentially a chronic progressive cholestatic liver disease characterized by immune-mediated destruction of small- and medium-sized intrahepatic bile ducts. There is considerable variation in the incidence and prevalence of the disease between regions of the world, although such differences likely reflect not only a true disparity in disease but also differences in awareness; for example, in the United States, PBC is often detected in an asymptomatic stage based on multi-phasic clinical testing. There has been considerable progress at defining the immune response in this disease, including quantitation of autoreactive T cells against PDC-E2, the major mitochondrial autoantigen. The overwhelming data suggests that patients develop PBC based on a genetic predisposition and loss of tolerance to one or more environmental agents. In this review, we will present an updated overview of PBC and place it in the context of autoimmunity.
Collapse
Affiliation(s)
- Raivo Uibo
- Institute of General and Molecular Pathology, Centre of Excellence for Translational Medicine, University of Tartu, Tartu, Estonia.
| | | | | | | |
Collapse
|
46
|
Erhardt A, Tiegs G. IL-33--a cytokine which balances on a knife's edge? J Hepatol 2012; 56:7-10. [PMID: 21703171 DOI: 10.1016/j.jhep.2011.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 05/18/2011] [Accepted: 05/20/2011] [Indexed: 01/01/2023]
|
47
|
Tian Z, Chen Y. Immunology of Liver. PRIMARY LIVER CANCER 2012:233-275. [DOI: 10.1007/978-3-642-28702-2_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
48
|
Feng M, Wang Q, Zhang F, Lu L. Ex vivo induced regulatory T cells regulate inflammatory response of Kupffer cells by TGF-beta and attenuate liver ischemia reperfusion injury. Int Immunopharmacol 2011; 12:189-96. [PMID: 22155100 DOI: 10.1016/j.intimp.2011.11.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Revised: 10/29/2011] [Accepted: 11/23/2011] [Indexed: 02/06/2023]
Abstract
In the presence of TGF-β, CD4+CD62L+T cells can be induced to CD4+CD25+FoxP3+ regulatory T cells (iTregs). In our previous work, we have shown that adoptive transfer of iTregs promoted liver recovery from ischemia reperfusion injury (IRI). In this study, we examined the molecular mechanism underlying the liver IRI attenuation by iTregs in a mouse partial hepatic IRI model. We found that the population of hepatic Tregs decreased significantly at 24 h after reperfusion. Adoptive transfer of iTregs before IRI markedly increased the numbers of hepatic Tregs and attenuated liver IRI as indicated by reduced serum aminotransferases and proinflammatory cytokines, such as interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α). Ex vivo study indicated that iTregs suppressed IL-1β and TNF-α expression, promoted transcription of interleukin-10 (IL-10), and elevated phosphorylation of SMAD3 in Kupffer cells (KCs). Furthermore, inhibition of TGF-β signaling by anti-TGF-β abolished the effects on KCs. Treatment with TGF-β suppressed matrix metalloprotease (MMP9) production in KCs and protected liver from IRI. In conclusion, our results suggest that iTregs play a critical role in hepatic IRI by regulating pro-inflammatory and anti-inflammatory function of KCs through TGF-β.
Collapse
Affiliation(s)
- Min Feng
- Department of Liver Transplantation, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | | | | | | |
Collapse
|
49
|
Relatively increased number of liver Foxp3+ regulatory T cells against hepatic lesions in murine lupus. ACTA ACUST UNITED AC 2011; 31:476. [PMID: 21823008 DOI: 10.1007/s11596-011-0476-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Indexed: 02/05/2023]
|
50
|
Miao J, Zhang J, Zheng L, Yu X, Zhu W, Zou S. Taurine attenuates Streptococcus uberis-induced mastitis in rats by increasing T regulatory cells. Amino Acids 2011; 42:2417-28. [PMID: 21809074 DOI: 10.1007/s00726-011-1047-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 07/18/2011] [Indexed: 01/25/2023]
Abstract
Taurine (Tau) is reported to have a key role in the regulation of the innate immune response and thus reduce tissue damage induced by bacterial infection. In this study, the effects of Tau on a rat model of mastitis induced by Streptococcus uberis (S. uberis) and the changes of T regulatory cells (Tregs) were assessed. Starting on gestation day 14 and continuing until parturition, 100 mg/kg of taurine (group TS) or an equal volume of physiological saline (group CS) was administered daily, per os. Seventy-two hours after parturition, rats were infused with approximately 100 cfu of S. uberis into each of two mammary glands. The results showed that the resultant inflammation, evidenced by swelling, secretory epithelial cell degeneration, increased adipose tissue and neutrophil (PMN) infiltration were evident in mammary tissue following injection with S. uberis. Pre-treatment with Tau attenuated these morphologic changes, the expression of interleukin (IL)-2, interferon (INF)-γ mRNA, myeloperoxidase (MPO) activity and N-acetyl-β-D-glucosaminidase (NAGase) in mammary tissue. The percentages of Foxp3+CD25+CD4+/lymphocytes (Tregs) were dramatically increased after the S. uberis challenge. Significant differences (P<0.05) were observed at 24, and 72 h post S. uberis-injection (PI) in CS. Pre-treatment further increased the percentage of Tregs and a significant difference between CS and TS (P<0.05) was apparent at 24 h PI. Our data indicate that in rats, Tau can be used to regulate the immune response following infection by S. uberis and consequently prevent mammary tissue damage by increasing Tregs.
Collapse
Affiliation(s)
- Jinfeng Miao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | | | | | | | | | | |
Collapse
|