1
|
Alonso-Guallart P, Harle D. Role of chemokine receptors in transplant rejection and graft-versus-host disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 388:95-123. [PMID: 39260939 DOI: 10.1016/bs.ircmb.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Organ transplantation increases life expectancy and improves the quality of life of patients experiencing specific conditions such as terminal organ failure. Despite matching efforts between donor and recipient, immune activation can interfere with allograft survival after transplantation if immunosuppression is not used. With both innate and adaptive responses, this is a complicated immunological process. This can lead to organ rejection, or graft-versus-host disease (GVHD), depending on the origin of the immune response. Inflammatory factors, such as chemokine receptors and their ligands, are involved in a wide variety of immunological processes, including modulating transplant rejection or GVHD, therefore, chemokine biology has been a major focus of transplantation studies. These molecules attract circulating peripheral leukocytes to infiltrate into the allograft and facilitate dendritic and T cell trafficking between lymph nodes and the graft during the allogeneic response. In this chapter, we will review the most relevant chemokine receptors such as CXCR3 and CCR5, among others, and their ligands involved in the process of allograft rejection for solid organ transplantation and graft-versus-host disease in the context of hematopoietic cell transplantation.
Collapse
Affiliation(s)
| | - David Harle
- Columbia Center for Translational Immunology
| |
Collapse
|
2
|
Döring Y, van der Vorst EP, Yan Y, Neideck C, Blanchet X, Jansen Y, Kemmerich M, Bayasgalan S, Peters LJ, Hristov M, Bidzhekov K, Yin C, Zhang X, Leberzammer J, Li Y, Park I, Kral M, Nitz K, Parma L, Gencer S, Habenicht A, Faussner A, Teupser D, Monaco C, Holdt L, Megens RT, Atzler D, Santovito D, von Hundelshausen P, Weber C. Identification of a non-canonical chemokine-receptor pathway suppressing regulatory T cells to drive atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:221-242. [PMID: 39044999 PMCID: PMC7616283 DOI: 10.1038/s44161-023-00413-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 12/14/2023] [Indexed: 07/25/2024]
Abstract
CCL17 is produced by conventional dendritic cells (cDCs), signals through CCR4 on regulatory T cells (Tregs), and drives atherosclerosis by suppressing Treg functions through yet undefined mechanisms. Here we show that cDCs from CCL17-deficient mice display a pro-tolerogenic phenotype and transcriptome that is not phenocopied in mice lacking its cognate receptor CCR4. In the plasma of CCL17-deficient mice, CCL3 was the only decreased cytokine/chemokine. We found that CCL17 signaled through CCR8 as an alternate high-affinity receptor, which induced CCL3 expression and suppressed Treg functions in the absence of CCR4. Genetic ablation of CCL3 and CCR8 in CD4+ T cells reduced CCL3 secretion, boosted FoxP3+ Treg numbers, and limited atherosclerosis. Conversely, CCL3 administration exacerbated atherosclerosis and restrained Treg differentiation. In symptomatic versus asymptomatic human carotid atheroma, CCL3 expression was increased, while FoxP3 expression was reduced. Together, we identified a non-canonical chemokine pathway whereby CCL17 interacts with CCR8 to yield a CCL3-dependent suppression of atheroprotective Tregs.
Collapse
Affiliation(s)
- Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Emiel P.C. van der Vorst
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Yi Yan
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Pediatric Translational Medicine Institute and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Carlos Neideck
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | - Xavier Blanchet
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | - Yvonne Jansen
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | - Manuela Kemmerich
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | | | - Linsey J.F. Peters
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Michael Hristov
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | - Kiril Bidzhekov
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | - Changjun Yin
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Xi Zhang
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | - Julian Leberzammer
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Ya Li
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | - Inhye Park
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom
| | - Maria Kral
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Katrin Nitz
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | - Laura Parma
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Selin Gencer
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | - Andreas Habenicht
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Alexander Faussner
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | - Daniel Teupser
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Germany
| | - Claudia Monaco
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Lesca Holdt
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Germany
| | - Remco T.A. Megens
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich
| | - Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, Milan, Italy
| | | | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands
| |
Collapse
|
3
|
Yero A, Shi T, Routy JP, Tremblay C, Durand M, Costiniuk CT, Jenabian MA. FoxP3+ CD8 T-cells in acute HIV infection and following early antiretroviral therapy initiation. Front Immunol 2022; 13:962912. [PMID: 35967314 PMCID: PMC9372390 DOI: 10.3389/fimmu.2022.962912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 06/29/2022] [Indexed: 12/21/2022] Open
Abstract
ObjectivesBesides CD4 regulatory T-cells (Tregs), immunosuppressor FoxP3+ CD8 T-cells are emerging as an important subset of Tregs, which contribute to immune dysfunction and disease progression in HIV infection. However, FoxP3+ CD8 T-cell dynamics in acute HIV infection and following early antiretroviral therapy (ART) initiation remain understudied.MethodsSubsets of FoxP3+ CD8 T-cells were characterized both prospectively and cross-sectionally in PBMCs from untreated acute (n=26) and chronic (n=10) HIV-infected individuals, early ART-treated in acute infection (n=10, median of ART initiation: 5.5 months post-infection), ART-treated in chronic infection (n=10), elite controllers (n=18), and HIV-uninfected controls (n=21).ResultsAcute and chronic infection were associated with increased total, effector memory, and terminally differentiated FoxP3+ CD8 T-cells, while early ART normalized only the frequencies of total FoxP3+ CD8 T-cells. We observed an increase in FoxP3+ CD8 T-cell immune activation (HLADR+/CD38+), senescence (CD57+/CD28-), and PD-1 expression during acute and chronic infection, which were not normalized by early ART. FoxP3+ CD8 T-cells in untreated participants expressed higher levels of immunosuppressive LAP(TGF-β1) and CD39 than uninfected controls, whereas early ART did not affect their expression. The expression of gut-homing markers CCR9 and Integrin-β7 by total FoxP3+ CD8 T-cells and CD39+ and LAP(TGF-β1)+ FoxP3+ CD8 T-cells increased in untreated individuals and remained higher than in uninfected controls despite early ART. Elite controllers share most of the FoxP3+ CD8 T-cell characteristics in uninfected individuals.ConclusionsAlthough early ART normalized total FoxP3+ CD8 T-cells frequencies, it did not affect the persistent elevation of the gut-homing potential of CD39+ and LAP(TGF-β1)+ FoxP3+ CD8 T-cell, which may contribute to immune dysfunction.
Collapse
Affiliation(s)
- Alexis Yero
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC, Canada
| | - Tao Shi
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC, Canada
| | - Jean-Pierre Routy
- Research Institute of McGill University Health Centre, Montreal, QC, Canada
- Chronic Viral Illness Service, Department of Medicine, Glen Site, McGill University Health Centre, Montreal, QC, Canada
| | - Cécile Tremblay
- Centre hospitalier de l'Université de Montréal (CHUM) Research Centre, Montreal, QC, Canada
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Madeleine Durand
- Centre hospitalier de l'Université de Montréal (CHUM) Research Centre, Montreal, QC, Canada
| | - Cecilia T. Costiniuk
- Research Institute of McGill University Health Centre, Montreal, QC, Canada
- Chronic Viral Illness Service, Department of Medicine, Glen Site, McGill University Health Centre, Montreal, QC, Canada
| | - Mohammad-Ali Jenabian
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC, Canada
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- *Correspondence: Mohammad-Ali Jenabian,
| |
Collapse
|
4
|
Neves EGA, Koh CC, Souza-Silva TG, Passos LSA, Silva ACC, Velikkakam T, Villani F, Coelho JS, Brodskyn CI, Teixeira A, Gollob KJ, Nunes MDCP, Dutra WO. T-Cell Subpopulations Exhibit Distinct Recruitment Potential, Immunoregulatory Profile and Functional Characteristics in Chagas versus Idiopathic Dilated Cardiomyopathies. Front Cardiovasc Med 2022; 9:787423. [PMID: 35187122 PMCID: PMC8847602 DOI: 10.3389/fcvm.2022.787423] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic Chagas cardiomyopathy (CCC) is one of the deadliest cardiomyopathies known and the most severe manifestation of Chagas disease, which is caused by infection with the parasite Trypanosoma cruzi. Idiopathic dilated cardiomyopathies (IDC) are a diverse group of inflammatory heart diseases that affect the myocardium and are clinically similar to CCC, often causing heart failure and death. While T-cells are critical for mediating cardiac pathology in CCC and IDC, the mechanisms underlying T-cell function in these cardiomyopathies are not well-defined. In this study, we sought to investigate the phenotypic and functional characteristics of T-cell subpopulations in CCC and IDC, aiming to clarify whether the inflammatory response is similar or distinct in these cardiomyopathies. We evaluated the expression of systemic cytokines, determined the sources of the different cytokines, the expression of their receptors, of cytotoxic molecules, and of molecules associated with recruitment to the heart by circulating CD4+, CD8+, and CD4-CD8- T-cells from CCC and IDC patients, using multiparameter flow cytometry combined with conventional and unsupervised machine-learning strategies. We also used an in silico approach to identify the expression of genes that code for key molecules related to T-cell function in hearts of patient with CCC and IDC. Our data demonstrated that CCC patients displayed a more robust systemic inflammatory cytokine production as compared to IDC. While CD8+ T-cells were highly activated in CCC as compared to IDC, CD4+ T-cells were more activated in IDC. In addition to differential expression of functional molecules, these cells also displayed distinct expression of molecules associated with recruitment to the heart. In silico analysis of gene transcripts in the cardiac tissue demonstrated a significant correlation between CD8 and inflammatory, cytotoxic and cardiotropic molecules in CCC transcripts, while no correlation with CD4 was observed. A positive correlation was observed between CD4 and perforin transcripts in hearts from IDC but not CCC, as compared to normal tissue. These data show a clearly distinct systemic and local cellular response in CCC and IDC, despite their similar cardiac impairment, which may contribute to identifying specific immunotherapeutic targets in these diseases.
Collapse
Affiliation(s)
- Eula G. A. Neves
- Department of Morphology, Cell-Cell Interactions Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Carolina C. Koh
- Department of Morphology, Cell-Cell Interactions Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Thaiany G. Souza-Silva
- Department of Morphology, Cell-Cell Interactions Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Lívia Silva Araújo Passos
- Department of Morphology, Cell-Cell Interactions Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Brigham and Womens Hospital, Harvard University, Boston, MA, United States
| | - Ana Carolina C. Silva
- Department of Morphology, Cell-Cell Interactions Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Teresiama Velikkakam
- Department of Morphology, Cell-Cell Interactions Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Villani
- Department of Morphology, Cell-Cell Interactions Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Minas Gerais State University, Divinópolis, Brazil
| | - Janete Soares Coelho
- Department of Morphology, Cell-Cell Interactions Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Ezequiel Dias Foundation, Belo Horizonte, Brazil
| | - Claudia Ida Brodskyn
- Gonçalo Moniz Research Center, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | - Andrea Teixeira
- Rene Rachou Institute, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
| | - Kenneth J. Gollob
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, INCT-DT, Salvador, Brazil
| | - Maria do Carmo P. Nunes
- Graduate Program in Infectology and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Walderez O. Dutra
- Department of Morphology, Cell-Cell Interactions Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, INCT-DT, Salvador, Brazil
- Graduate Program in Infectology and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
5
|
Antiretroviral therapy-treated HIV-infected adults with coronary artery disease are characterized by a distinctive regulatory T-cell signature. AIDS 2021; 35:1003-1014. [PMID: 33587446 DOI: 10.1097/qad.0000000000002842] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Despite the success of antiretroviral therapy (ART) to control viral replication, people living with HIV (PWH) have high levels of chronic systemic inflammation and immune dysregulation which drives accelerated co-morbidities including coronary artery disease (CAD). Regulatory T cells (Tregs) and ectonucleotidases CD39/CD73 are known to be athero-protective via their immunosuppressive and anti-inflammatory functions. DESIGN We assessed the dynamics of Treg subsets in ART-treated PWH with or without CAD vs. HIV-uninfected individuals. METHODS Blood specimens were obtained from 142 participants including ART-treated HIV-infected adults with (n = 43) or without CAD (n = 41), as well as HIV-uninfected controls with (n = 31) or without CAD (n = 27). CAD was determined by the presence of atherosclerotic features on computed tomography angiography of the coronary arteries performed on all study participants. Treg subsets frequencies were assessed by flow cytometry. RESULTS Regardless of statin treatment or ART regimen, HIV+CAD+ individuals had the highest total Treg frequencies and increased thymic generation and output of Tregs (Helios/CD31 expression), while athero-protective CD39+/CD73+ Tregs were significantly depleted in this group. Tregs from PWH had higher expression of CCR6/CXCR3 than uninfected individuals regardless of CAD, while in HIV+CAD+ individuals Tregs expressed the highest levels of CCR4, which limits their maintenance. The lowest levels of CD4+ and CD8+ T-cell immune activation has been observed in HIV+CAD+ within study groups. CONCLUSION ART-treated PWH with diagnosed CAD are characterized by profound alterations in populations of anti-inflammatory and athero-protective Treg subsets. These changes may contribute to atherosclerotic plaque formation and progression during chronic HIV infection in the ART era.
Collapse
|
6
|
Palchevskiy V, Xue YY, Kern R, Weigt SS, Gregson AL, Song SX, Fishbein MC, Hogaboam CM, Sayah DM, Lynch JP, Keane MP, Brooks DG, Belperio JA. CCR4 expression on host T cells is a driver for alloreactive responses and lung rejection. JCI Insight 2019; 5:121782. [PMID: 31085832 PMCID: PMC6629140 DOI: 10.1172/jci.insight.121782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 05/08/2019] [Indexed: 12/16/2022] Open
Abstract
Despite current immunosuppressive strategies, long-term lung transplant outcomes remain poor due to rapid allogenic responses. Using a stringent mouse model of allo-airway transplantation, we identify the CCR4-ligand axis as a central node driving secondary lymphoid tissue homing and activation of the allogeneic T cells that prevent long-term allograft survival. CCR4 deficiency on transplant recipient T cells diminishes allograft injury and when combined with CTLA4-Ig leads to an unprecedented long-term lung allograft accommodation. Thus, we identify CCR4-ligand interactions as a central mechanism driving allogeneic transplant rejection and suggest it as a potential target to enhance long-term lung transplant survival.
Collapse
Affiliation(s)
- Vyacheslav Palchevskiy
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Ying Ying Xue
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Rita Kern
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Stephen S. Weigt
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Aric L. Gregson
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Sophie X. Song
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Michael C. Fishbein
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Cory M. Hogaboam
- Pulmonary & Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - David M. Sayah
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Joseph P. Lynch
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Michael P. Keane
- University College Dublin School of Medicine, Respiratory Medicine, St Vincent’s University Hospital, Dublin, Ireland
| | - David G. Brooks
- Princess Margaret Cancer Center, University Health Network and Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - John A. Belperio
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| |
Collapse
|
7
|
Baur J, Otto C, Steger U, Klein-Hessling S, Muhammad K, Pusch T, Murti K, Wismer R, Germer CT, Klein I, Müller N, Serfling E, Avots A. The Transcription Factor NFATc1 Supports the Rejection of Heterotopic Heart Allografts. Front Immunol 2018; 9:1338. [PMID: 29946322 PMCID: PMC6005848 DOI: 10.3389/fimmu.2018.01338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/29/2018] [Indexed: 12/17/2022] Open
Abstract
The immune suppressants cyclosporin A (CsA) and tacrolimus (FK506) are used worldwide in transplantation medicine to suppress graft rejection. Both CsA and FK506 inhibit the phosphatase calcineurin (CN) whose activity controls the immune receptor-mediated activation of lymphocytes. Downstream targets of CN in lymphocytes are the nuclear factors of activated T cells (NFATs). We show here that the activity of NFATc1, the most prominent NFAT factor in activated lymphocytes supports the acute rejection of heterotopic heart allografts. While ablation of NFATc1 in T cells prevented graft rejection, ectopic expression of inducible NFATc1/αA isoform led to rejection of heart allografts in recipient mice. Acceptance of transplanted hearts in mice bearing NFATc1-deficient T cells was accompanied by a reduction in number and cytotoxicity of graft infiltrating cells. In CD8+ T cells, NFATc1 controls numerous intracellular signaling pathways that lead to the metabolic switch to aerobic glycolysis and the expression of numerous lymphokines, chemokines, and their receptors, including Cxcr3 that supports the rejection of allogeneic heart transplants. These findings favors NFATc1 as a molecular target for the development of new strategies to control the cytotoxicity of T cells upon organ transplantation.
Collapse
Affiliation(s)
- Johannes Baur
- Department of General, Visceral, Vascular, and Pediatric Surgery, University Hospital of Würzburg, Würzburg, Germany
| | - Christoph Otto
- Experimental Surgery, Department of General, Visceral, Vascular, and Pediatric Surgery, University Hospital of Würzburg, Würzburg, Germany
| | - Ulrich Steger
- Department of General, Visceral, Vascular, and Pediatric Surgery, University Hospital of Würzburg, Würzburg, Germany
| | - Stefan Klein-Hessling
- Department of Molecular Pathology, Institute of Pathology, Comprehensive Cancer Center Mainfranken, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Khalid Muhammad
- Department of Molecular Pathology, Institute of Pathology, Comprehensive Cancer Center Mainfranken, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Tobias Pusch
- Department of Molecular Pathology, Institute of Pathology, Comprehensive Cancer Center Mainfranken, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Krisna Murti
- Department of Molecular Pathology, Institute of Pathology, Comprehensive Cancer Center Mainfranken, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Rhoda Wismer
- Department of Molecular Pathology, Institute of Pathology, Comprehensive Cancer Center Mainfranken, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Christoph-Thomas Germer
- Department of General, Visceral, Vascular, and Pediatric Surgery, University Hospital of Würzburg, Würzburg, Germany
| | - Ingo Klein
- Transplant and Hepatobiliary Surgery, Department of General, Visceral, Vascular, and Pediatric Surgery, University Hospital of Würzburg, Würzburg, Germany
| | - Nora Müller
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Edgar Serfling
- Department of Molecular Pathology, Institute of Pathology, Comprehensive Cancer Center Mainfranken, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Andris Avots
- Department of Molecular Pathology, Institute of Pathology, Comprehensive Cancer Center Mainfranken, Julius-Maximilians University of Würzburg, Würzburg, Germany
| |
Collapse
|
8
|
Meng X, Gao W, Tang Y, Shen Z, Wang Z. Alterations of Serum IP-10 and TARC in Patients with Early Acute Rejection after Liver Transplantation. Cell Physiol Biochem 2017; 41:1063-1071. [DOI: 10.1159/000464114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 12/27/2016] [Indexed: 12/26/2022] Open
Abstract
Background/Aims: To analyze alterations of interferon-γ-induced protein 10 (IP-10) and thymus and activation-regulated chemokine (TARC) levels in early acute liver transplantation rejection. Methods: Thirty-six patients with early acute liver transplantation rejection were classified as non-, mild, moderate, and severe rejection groups. The levels of serum IP-10 and TARC were determined on days 3, 2, 1, and 0 before biopsy. Results: The IP-10 activities in all rejection groups were significantly higher (p < 0.05) than those in the non-rejection group at all time points and correlated with the extent of rejection (p < 0.05). The differences in TARC among the three rejection groups were significant (p < 0.05), and its highest level was found in the mild rejection group at all observed time points, whereas its lowest level was detected in the severe rejection group. The analysis of the TARC/IP-10 ratio revealed that the volume was correlated with the rejection degree. This ratio in the moderate and severe rejection groups on days 2, 1, and 0 before biopsy were 20% lower than that before transplantation. Conclusion: Serum IP-10 showed an increasing trend during early acute liver transplantation rejection. IP-10 increase or TARC/IP-10 ratio decrease combining with abnormal hepatic enzymatic alteration could be a valuable and specific sign for early rejection of the transplanted liver.
Collapse
|
9
|
Piñeros AR, Campos LW, Fonseca DM, Bertolini TB, Gembre AF, Prado RQ, Alves-Filho JC, Ramos SG, Russo M, Bonato VLD. M2 macrophages or IL-33 treatment attenuate ongoing Mycobacterium tuberculosis infection. Sci Rep 2017; 7:41240. [PMID: 28128217 PMCID: PMC5269597 DOI: 10.1038/srep41240] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 12/19/2016] [Indexed: 11/09/2022] Open
Abstract
The protective effects of mycobacterial infections on lung allergy are well documented. However, the inverse relationship between tuberculosis and type 2 immunity is still elusive. Although type 1 immunity is essential to protection against Mycobacterium tuberculosis it might be also detrimental to the host due to the induction of extensive tissue damage. Here, we determined whether lung type 2 immunity induced by allergen sensitization and challenge could affect the outcome of M. tuberculosis infection. We used two different protocols in which sensitization and allergen challenge were performed before or after M. tuberculosis infection. We found an increased resistance to M. tuberculosis only when allergen exposure was given after, but not before infection. Infected mice exposed to allergen exhibited lower bacterial load and cellular infiltrates in the lungs. Enhanced resistance to infection after allergen challenge was associated with increased gene expression of alternatively activated macrophages (M2 macrophages) and IL-33 levels. Accordingly, either adoptive transfer of M2 macrophages or systemic IL-33 treatment was effective in attenuating M. tuberculosis infection. Notably, the enhanced resistance induced by allergen exposure was dependent on IL-33 receptor ST2. Our work indicates that IL-33 might be an alternative therapeutic treatment for severe tuberculosis.
Collapse
Affiliation(s)
- A R Piñeros
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - L W Campos
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - D M Fonseca
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - T B Bertolini
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - A F Gembre
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - R Q Prado
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - J C Alves-Filho
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - S G Ramos
- Department of Pathology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - M Russo
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - V L D Bonato
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
10
|
Stephenson E, Savvatis K, Mohiddin SA, Marelli-Berg FM. T-cell immunity in myocardial inflammation: pathogenic role and therapeutic manipulation. Br J Pharmacol 2016; 174:3914-3925. [PMID: 27590129 DOI: 10.1111/bph.13613] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/11/2016] [Accepted: 08/16/2016] [Indexed: 12/13/2022] Open
Abstract
T-cell-mediated immunity has been linked not only to a variety of heart diseases, including classic inflammatory diseases such as myocarditis and post-myocardial infarction (Dressler's) syndrome, but also to conditions without an obvious inflammatory component such as idiopathic dilated cardiomyopathy and hypertensive cardiomyopathy. It has been recently proposed that in all these conditions, the heart becomes the focus of T-cell-mediated autoimmune inflammation following ischaemic or infectious injury. For example, in acute myocarditis, an inflammatory disease of heart muscle, T-cell responses are thought to arise as a consequence of a viral infection. In a number of patients, persistent T-cell-mediated responses in acute viral myocarditis can lead to autoimmunity and chronic cardiac inflammation resulting in dilated cardiomyopathy. In spite of the major progress made in understanding the mechanisms of pathogenic T-cell responses, effective and safe therapeutic targeting of the immune system in chronic inflammatory diseases of the heart has not yet been developed due to the lack of specific diagnostic and prognostic biomarkers at an early stage. This has also prevented the identification of targets for patient-tailored immunomodulatory therapies that are both disease- and organ-selective. In this review, we discuss current knowledge of the development and functional characteristics of pathogenic T-cell-mediated immune responses in the heart, and, in particular, in myocarditis, as well as recent advances in experimental models which have the potential to translate into heart-selective immunomodulation. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- E Stephenson
- William Harvey Research Institute, London, UK.,Barts and The London School of Medicine, London, UK
| | - K Savvatis
- William Harvey Research Institute, London, UK.,Barts and The London School of Medicine, London, UK.,Department of Cardiology, Barts Heart Centre, St. Bartholomew NHS Trust, London, UK
| | - S A Mohiddin
- William Harvey Research Institute, London, UK.,Barts and The London School of Medicine, London, UK.,Department of Cardiology, Barts Heart Centre, St. Bartholomew NHS Trust, London, UK
| | - F M Marelli-Berg
- William Harvey Research Institute, London, UK.,Barts and The London School of Medicine, London, UK
| |
Collapse
|
11
|
Fu H, Ward EJ, Marelli-Berg FM. Mechanisms of T cell organotropism. Cell Mol Life Sci 2016; 73:3009-33. [PMID: 27038487 PMCID: PMC4951510 DOI: 10.1007/s00018-016-2211-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 02/06/2023]
Abstract
Protective immunity relies upon T cell differentiation and subsequent migration to target tissues. Similarly, immune homeostasis requires the localization of regulatory T cells (Tregs) to the sites where immunity takes place. While naïve T lymphocytes recirculate predominantly in secondary lymphoid tissue, primed T cells and activated Tregs must traffic to the antigen rich non-lymphoid tissue to exert effector and regulatory responses, respectively. Following priming in draining lymph nodes, T cells acquire the 'homing receptors' to facilitate their access to specific tissues and organs. An additional level of topographic specificity is provided by T cells receptor recognition of antigen displayed by the endothelium. Furthermore, co-stimulatory signals (such as those induced by CD28) have been shown not only to regulate T cell activation and differentiation, but also to orchestrate the anatomy of the ensuing T cell response. We here review the molecular mechanisms supporting trafficking of both effector and regulatory T cells to specific antigen-rich tissues.
Collapse
Affiliation(s)
- Hongmei Fu
- William Harvey Research Institute, Heart Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Eleanor Jayne Ward
- William Harvey Research Institute, Heart Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Federica M Marelli-Berg
- William Harvey Research Institute, Heart Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
12
|
Abstract
Expression of distinct homing receptors guides adaptive immune cells to antigen-rich tissue. In this issue of Immunity, Komarowska et al. (2015) describe an autocrine loop that is initiated by cardiac-expressed hepatocyte growth factor to direct T cells into the heart during inflammation and cardiac transplant rejection.
Collapse
Affiliation(s)
- Dennis Wolf
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Jie Li
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| |
Collapse
|
13
|
Hepatocyte Growth Factor Receptor c-Met Instructs T Cell Cardiotropism and Promotes T Cell Migration to the Heart via Autocrine Chemokine Release. Immunity 2015; 42:1087-99. [PMID: 26070483 PMCID: PMC4510150 DOI: 10.1016/j.immuni.2015.05.014] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 10/18/2014] [Accepted: 04/02/2015] [Indexed: 12/20/2022]
Abstract
Effector-T-cell-mediated immunity depends on the efficient localization of antigen-primed lymphocytes to antigen-rich non-lymphoid tissue, which is facilitated by the expression of a unique set of “homing” receptors acquired by memory T cells. We report that engagement of the hepatocyte growth factor (HGF) receptor c-Met by heart-produced HGF during priming in the lymph nodes instructs T cell cardiotropism, which was associated with a specialized homing “signature” (c-Met+CCR4+CXCR3+). c-Met signals facilitated T cell recruitment to the heart via the chemokine receptor CCR5 by inducing autocrine CCR5 ligand release. c-Met triggering was sufficient to support cardiotropic T cell recirculation, while CCR4 and CXCR3 sustained recruitment during heart inflammation. Transient pharmacological blockade of c-Met during T cell priming led to enhanced survival of heart, but not skin, allografts associated with impaired localization of alloreactive T cells to heart grafts. These findings suggest c-Met as a target for development of organ-selective immunosuppressive therapies. HGF-induced c-Met signals during activation induce cardiotropic memory T cells Cardiotropic T cells express a specific molecular signature (c-Met+CCR4+CXCR3+) By inducing an autocrine chemokine loop, c-Met also promotes T cell recruitment Blockade of the HGF-c-Met axis prevents heart, but not skin, allograft rejection
Collapse
|
14
|
Abstract
Organ transplantation appears today to be the best alternative to replace the loss of vital organs induced by various diseases. Transplants can, however, also be rejected by the recipient. In this review, we provide an overview of the mechanisms and the cells/molecules involved in acute and chronic rejections. T cells and B cells mainly control the antigen-specific rejection and act either as effector, regulatory, or memory cells. On the other hand, nonspecific cells such as endothelial cells, NK cells, macrophages, or polymorphonuclear cells are also crucial actors of transplant rejection. Last, beyond cells, the high contribution of antibodies, chemokines, and complement molecules in graft rejection is discussed in this article. The understanding of the different components involved in graft rejection is essential as some of them are used in the clinic as biomarkers to detect and quantify the level of rejection.
Collapse
Affiliation(s)
- Aurélie Moreau
- INSERM UMR 1064, Center for Research in Transplantation and Immunology-ITUN, CHU de Nantes 44093, France
| | | | | | | |
Collapse
|
15
|
Abstract
Renal allograft rejection or dysfunction often results in graft failure, and remains the major obstacle in the success of renal transplantation. Various immunological and nonimmunological factors are involved in allograft rejection. In addition to human leukocyte antigen loci, several genetically controlled molecules have been identified in recent years as playing important roles in the process of rejection. Genetic variants in genes encoding different T-helper (Th) type 1 and Th2 cytokines, chemokines and their receptors, growth factors, molecules of the renin-angiotensin system, enzymes of the homocysteine pathway, and proteins acting as substrates of immunosuppressive drugs impact on the success of engraftment and highlight the concept of genetic predisposition to allograft rejection. This review evaluates specific genetic variants and their functional roles in graft failure, with an emphasis on the latest methodologies available for genotyping, and appropriate strategies to enable them to become a tool of predictive and individualized medicine to ensure better transplant outcome.
Collapse
Affiliation(s)
- Faisal Khan
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, (UP), India
| | | | | |
Collapse
|
16
|
Weber C, Meiler S, Döring Y, Koch M, Drechsler M, Megens RTA, Rowinska Z, Bidzhekov K, Fecher C, Ribechini E, van Zandvoort MAMJ, Binder CJ, Jelinek I, Hristov M, Boon L, Jung S, Korn T, Lutz MB, Förster I, Zenke M, Hieronymus T, Junt T, Zernecke A. CCL17-expressing dendritic cells drive atherosclerosis by restraining regulatory T cell homeostasis in mice. J Clin Invest 2011; 121:2898-910. [PMID: 21633167 DOI: 10.1172/jci44925] [Citation(s) in RCA: 207] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 04/13/2011] [Indexed: 01/13/2023] Open
Abstract
Immune mechanisms are known to control the pathogenesis of atherosclerosis. However, the exact role of DCs, which are essential for priming of immune responses, remains elusive. We have shown here that the DC-derived chemokine CCL17 is present in advanced human and mouse atherosclerosis and that CCL17+ DCs accumulate in atherosclerotic lesions. In atherosclerosis-prone mice, Ccl17 deficiency entailed a reduction of atherosclerosis, which was dependent on Tregs. Expression of CCL17 by DCs limited the expansion of Tregs by restricting their maintenance and precipitated atherosclerosis in a mechanism conferred by T cells. Conversely, a blocking antibody specific for CCL17 expanded Tregs and reduced atheroprogression. Our data identify DC-derived CCL17 as a central regulator of Treg homeostasis, implicate DCs and their effector functions in atherogenesis, and suggest that CCL17 might be a target for vascular therapy.
Collapse
Affiliation(s)
- Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Transplant vasculopathy (TV) remains the leading cause of late death among heart transplant recipients. Transplant vasculopathy is characterized by progressive neointimal proliferation, leading to ischemic failure of the allograft. Multiple experimental and clinical studies have shown that injury to the graft at various stages of transplantation can be a risk factor for development of transplant vasculopathy. The hallmark of cardiac allograft injury is the infiltration of leukocytes. Recruitment of leukocytes requires intercellular communication between infiltrating cells, endothelium, parenchymal cells, and components of extracellular matrix. These events are mediated via the generation of adhesion molecules, cytokines, and chemokines. The chemokines, by virtue of their specific cell receptor expression, can selectively mediate the local recruitment/activation of distinct leukocytes/cells, allowing for migration across the endothelium and beyond the vascular compartment. This report provides a comprehensive review of the chemokines that participate in the development of transplant vasculopathy.
Collapse
Affiliation(s)
- John A Belperio
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angele, CA 90095, USA
| | | |
Collapse
|
18
|
Abstract
CC chemokine receptor 4 (CCR4) and its two ligands, CCL17 and CCL22, are critically involved in different immune processes. In models of lipopolysaccharide-induced shock, CCR4-deficient (CCR4(-/-)) mice showed improved survival rates associated with attenuated proinflammatory cytokine release. Using CCR4(-/-) mice with a C57BL/6 background, this study describes for the first time the role of CCR4 in a murine model of polymicrobial abdominal sepsis, the colon ascendens stent peritonitis (CASP). CASP-induced sepsis led to a massive downregulation of CCR4 in lymphoid and nonlymphoid tissues, whereas the expression of CCL17 and CCL22 was independent of the presence of CCR4. After CASP, CCR4(-/-) animals showed a strongly enhanced bacterial clearance in several organs but not in the peritoneal lavage fluid and the blood. In addition, significantly reduced levels of proinflammatory cytokines/chemokines were measured in organ supernatants as well as in the sera of CCR4(-/-) mice. CCR4 deficiency consequently resulted in an attenuated severity of systemic sepsis and a strongly improved survival rate after CASP or CASP with intervention. Thus, our data provide clear evidence that CCR4 plays a strictly detrimental role in the course of polymicrobial sepsis.
Collapse
|
19
|
Gelman AE, Okazaki M, Lai J, Kornfeld CG, Kreisel FH, Richardson SB, Sugimoto S, Tietjens JR, Patterson GA, Krupnick AS, Kreisel D. CD4+ T lymphocytes are not necessary for the acute rejection of vascularized mouse lung transplants. THE JOURNAL OF IMMUNOLOGY 2008; 180:4754-62. [PMID: 18354199 DOI: 10.4049/jimmunol.180.7.4754] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Acute rejection continues to present a major obstacle to successful lung transplantation. Although CD4(+) T lymphocytes are critical for the rejection of some solid organ grafts, the role of CD4(+) T cells in the rejection of lung allografts is largely unknown. In this study, we demonstrate in a novel model of orthotopic vascularized mouse lung transplantation that acute rejection of lung allografts is independent of CD4(+) T cell-mediated allorecognition pathways. CD4(+) T cell-independent rejection occurs in the absence of donor-derived graft-resident hematopoietic APCs. Furthermore, blockade of the CD28/B7 costimulatory pathways attenuates acute lung allograft rejection in the absence of CD4(+) T cells, but does not delay acute rejection when CD4(+) T cells are present. Our results provide new mechanistic insight into the acute rejection of lung allografts and highlight the importance of identifying differences in pathways that regulate the rejection of various organs.
Collapse
Affiliation(s)
- Andrew E Gelman
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University, St Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Cornell LD, Smith RN, Colvin RB. Kidney transplantation: mechanisms of rejection and acceptance. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2008; 3:189-220. [PMID: 18039144 DOI: 10.1146/annurev.pathmechdis.3.121806.151508] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We describe the molecular and cellular mechanisms believed to be responsible for the rejection of renal allografts, including acute T cell-mediated rejection, acute antibody-mediated (humoral) rejection, rejection mediated by the innate immune system, and chronic rejection. We present mechanisms of graft acceptance, including accommodation, regulation, and tolerance. Studies in animals have replicated many pathologic features of acute and chronic rejection. We illuminate the pathogenesis of human pathology by reflection from experimental models.
Collapse
Affiliation(s)
- Lynn D Cornell
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | | | | |
Collapse
|
21
|
Ness TL, Ewing JL, Hogaboam CM, Kunkel SL. CCR4 is a key modulator of innate immune responses. THE JOURNAL OF IMMUNOLOGY 2007; 177:7531-9. [PMID: 17114422 DOI: 10.4049/jimmunol.177.11.7531] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CCR4 is recognized as a key receptor in Th2-associated immune processes, although very little is known about its role in innate immunity. Previous studies reported increased resistance to LPS-induced lethality in CCR4(-/-) mice compared with wild-type mice. This study demonstrates that CCR4(-/-) mice are similarly resistant to challenge with other TLR agonists, as well as bacterial peritonitis. Resistance was associated with enhanced early leukocyte recruitment, increased TLR expression, a skewed type 2 cytokine/chemokine profile, and improved bacterial clearance. Macrophages from CCR4(-/-) mice exhibited many features consistent with alternative activation, including elevated secretion of type 2 cytokines/chemokines and the found in inflammatory zone 1 (FIZZ1) protein. MyD88-dependent NF-kappaB signaling was significantly down-regulated in CCR4(-/-) macrophages, whereas p38 MAPK and JNK activation were conversely increased. These data stress the importance of CCR4 in macrophage differentiation and innate immune responses to pathogens, as well as the involvement of chemokine receptor expression in TLR signaling regulation.
Collapse
Affiliation(s)
- Traci L Ness
- Department of Pathology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
22
|
Wang X, Xu F, Xu Q, Mahmud H, Houze J, Zhu L, Akerman M, Tonn G, Tang L, McMaster BE, Dairaghi DJ, Schall TJ, Collins TL, Medina JC. Optimization of 2-aminothiazole derivatives as CCR4 antagonists. Bioorg Med Chem Lett 2006; 16:2800-3. [PMID: 16497499 DOI: 10.1016/j.bmcl.2006.01.126] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2005] [Revised: 01/28/2006] [Accepted: 01/31/2006] [Indexed: 11/29/2022]
Abstract
A series of 2-aminothiazole-derived antagonists of the CCR4 receptor has been synthesized and their affinity for the receptor evaluated using a [(125)I]TARC (CCL17) displacement assay. Optimization of these compounds for potency and pharmacokinetic properties led to the discovery of potent, orally bioavailable antagonists.
Collapse
Affiliation(s)
- Xuemei Wang
- Amgen Inc., 1120 Veterans Boulevard, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Carpenter KJ, Hogaboam CM. Immunosuppressive effects of CCL17 on pulmonary antifungal responses during pulmonary invasive aspergillosis. Infect Immun 2005; 73:7198-207. [PMID: 16239514 PMCID: PMC1273903 DOI: 10.1128/iai.73.11.7198-7207.2005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Aspergillus fumigatus-sensitized CCR4-deficient (CCR4-/-) mice exhibit an accelerated clearance of conidia during fungal asthma. In the present study, we examined the roles of CCL17 and CCL22, two CCR4 ligands, during pulmonary invasive aspergillosis in neutropenic mice. Kaplan-Meier survival curve analysis revealed that wild-type C57BL/6 (CCR4+/+) mice were significantly protected from the lethal effects of Aspergillus compared with their wild-type controls following systemic neutralization with anti-CCL17 but not anti-CCL22 antibodies. Systemic neutralization of CCL17 significantly increased whole-lung CCL2 levels. Mouse survival and histological analysis revealed that the receptor mediating the deleterious effects of CCL17 was CCR4 since mice genetically deficit in CCR4 (CCR4-/-) did not develop invasive aspergillosis. Enzyme-linked immunosorbent assay analysis of whole-lung samples at day 2 after conidial challenge in neutrophil-depleted CCR4-/- and CCR4+/+ mice revealed that whole-lung IL-12 levels were significantly increased in the CCR4-/- group compared with the wild-type group. Also at day 2 after conidial challenge, significantly greater numbers of CD11c+ F4/80+ and CD11c+/CD86+ but fewer CD3/NK1.1+ cells were present in the lungs of CCR4-/- mice compared with their wild-type counterparts. Thus, CCL17-CCR4 interactions dramatically impair the pulmonary antifungal response against A. fumigatus in neutropenic mice.
Collapse
MESH Headings
- Animals
- Aspergillosis/immunology
- Bronchoalveolar Lavage
- Chemokine CCL17
- Chemokine CCL2/metabolism
- Chemokine CCL22
- Chemokine CCL3
- Chemokine CCL4
- Chemokines, CC/immunology
- Chemokines, CC/metabolism
- Cytokines/genetics
- Cytokines/metabolism
- Female
- Immune Tolerance/immunology
- Interleukin-12/metabolism
- Lung/immunology
- Lung/metabolism
- Lung/pathology
- Lung Diseases, Fungal/immunology
- Lung Diseases, Fungal/microbiology
- Lymphocytes/cytology
- Lymphocytes/immunology
- Macrophage Inflammatory Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neutropenia/immunology
- Receptors, CCR4
- Receptors, CCR8
- Receptors, Chemokine/deficiency
- Receptors, Chemokine/genetics
- Receptors, Chemokine/immunology
- Specific Pathogen-Free Organisms
- Survival Rate
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Kristin J Carpenter
- Department of Pathology, University of Michigan Medical School, Room 5216B, Med Sci I, 1301 Catherine Road, Ann Arbor, MI 48109-0602, USA
| | | |
Collapse
|