101
|
Variegated RHOA mutations in adult T-cell leukemia/lymphoma. Blood 2015; 127:596-604. [PMID: 26574607 DOI: 10.1182/blood-2015-06-644948] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 11/02/2015] [Indexed: 12/24/2022] Open
Abstract
Adult T-cell leukemia/lymphoma (ATLL) is a distinct form of peripheral T-cell lymphoma with poor prognosis, which is caused by the human T-lymphotropic virus type 1 (HTLV-1). In contrast to the unequivocal importance of HTLV-1 infection in the pathogenesis of ATLL, the role of acquired mutations in HTLV-1 infected T cells has not been fully elucidated, with a handful of genes known to be recurrently mutated. In this study, we identified unique RHOA mutations in ATLL through whole genome sequencing of an index case, followed by deep sequencing of 203 ATLL samples. RHOA mutations showed distinct distribution and function from those found in other cancers. Involving 15% (30/203) of ATLL cases, RHOA mutations were widely distributed across the entire coding sequence but almost invariably located at the guanosine triphosphate (GTP)-binding pocket, with Cys16Arg being most frequently observed. Unexpectedly, depending on mutation types and positions, these RHOA mutants showed different or even opposite functional consequences in terms of GTP/guanosine diphosphate (GDP)-binding kinetics, regulation of actin fibers, and transcriptional activation. The Gly17Val mutant did not bind GTP/GDP and act as a dominant negative molecule, whereas other mutants (Cys16Arg and Ala161Pro) showed fast GTP/GDP cycling with enhanced transcriptional activation. These findings suggest that both loss- and gain-of-RHOA functions could be involved in ATLL leukemogenesis. In summary, our study not only provides a novel insight into the molecular pathogenesis of ATLL but also highlights a unique role of variegation of heterologous RHOA mutations in human cancers.
Collapse
|
102
|
Ahmed R, Westera L, Drylewicz J, Elemans M, Zhang Y, Kelly E, Reljic R, Tesselaar K, de Boer RJ, Macallan DC, Borghans JAM, Asquith B. Reconciling Estimates of Cell Proliferation from Stable Isotope Labeling Experiments. PLoS Comput Biol 2015; 11:e1004355. [PMID: 26437372 PMCID: PMC4593553 DOI: 10.1371/journal.pcbi.1004355] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 05/26/2015] [Indexed: 01/01/2023] Open
Abstract
Stable isotope labeling is the state of the art technique for in vivo quantification of lymphocyte kinetics in humans. It has been central to a number of seminal studies, particularly in the context of HIV-1 and leukemia. However, there is a significant discrepancy between lymphocyte proliferation rates estimated in different studies. Notably, deuterated 2H2-glucose (D2-glucose) labeling studies consistently yield higher estimates of proliferation than deuterated water (D2O) labeling studies. This hampers our understanding of immune function and undermines our confidence in this important technique. Whether these differences are caused by fundamental biochemical differences between the two compounds and/or by methodological differences in the studies is unknown. D2-glucose and D2O labeling experiments have never been performed by the same group under the same experimental conditions; consequently a direct comparison of these two techniques has not been possible. We sought to address this problem. We performed both in vitro and murine in vivo labeling experiments using identical protocols with both D2-glucose and D2O. This showed that intrinsic differences between the two compounds do not cause differences in the proliferation rate estimates, but that estimates made using D2-glucose in vivo were susceptible to difficulties in normalization due to highly variable blood glucose enrichment. Analysis of three published human studies made using D2-glucose and D2O confirmed this problem, particularly in the case of short term D2-glucose labeling. Correcting for these inaccuracies in normalization decreased proliferation rate estimates made using D2-glucose and slightly increased estimates made using D2O; thus bringing the estimates from the two methods significantly closer and highlighting the importance of reliable normalization when using this technique. Stable isotope labeling is used to quantify the rate at which living cells proliferate and die in humans. It has been central to a number of seminal studies, particularly in viral infections such as HIV-1, and leukemia. However, different labels (deuterated water or deuterated glucose) yield different estimates for the rate of cell proliferation and loss; this hampers our understanding and weakens our confidence in this important technique. We performed in vitro and in vivo experiments as well as a new analysis of existing data to directly compare the two labels. This reveals that a major source of the discrepancy lies in the difficulty of assessing deuterated glucose availability. We reconcile published studies and provide recommendations to avoid this problem in the future.
Collapse
Affiliation(s)
- Raya Ahmed
- Institute for Infection and Immunity, St. George’s, University of London, London, United Kingdom
| | - Liset Westera
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Julia Drylewicz
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Theoretical Biology & Bioinformatics, Utrecht University, The Netherlands
| | - Marjet Elemans
- Department of Medicine, Imperial College, London, United Kingdom
| | - Yan Zhang
- Institute for Infection and Immunity, St. George’s, University of London, London, United Kingdom
| | - Elizabeth Kelly
- Institute for Infection and Immunity, St. George’s, University of London, London, United Kingdom
| | - Rajko Reljic
- Institute for Infection and Immunity, St. George’s, University of London, London, United Kingdom
| | - Kiki Tesselaar
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rob J. de Boer
- Theoretical Biology & Bioinformatics, Utrecht University, The Netherlands
| | - Derek C. Macallan
- Institute for Infection and Immunity, St. George’s, University of London, London, United Kingdom
| | - José A. M. Borghans
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Becca Asquith
- Department of Medicine, Imperial College, London, United Kingdom
- * E-mail:
| |
Collapse
|
103
|
Stervbo U, Bozzetti C, Baron U, Jürchott K, Meier S, Mälzer JN, Nienen M, Olek S, Rachwalik D, Schulz AR, Neumann A, Babel N, Grützkau A, Thiel A. Effects of aging on human leukocytes (part II): immunophenotyping of adaptive immune B and T cell subsets. AGE (DORDRECHT, NETHERLANDS) 2015; 37:93. [PMID: 26324156 PMCID: PMC5005833 DOI: 10.1007/s11357-015-9829-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 08/07/2015] [Indexed: 06/04/2023]
Abstract
Immunosenescence results from a continuous deterioration of immune responses resulting in a decreased response to vaccines. A well-described age-related alteration of the immune system is the decrease of de novo generation of T and B cells. In addition, the accumulation of memory cells and loss of diversity in antigen specificities resulting from a lifetime of exposure to pathogens has also been described. However, the effect of aging on subsets of γδTCR(+) T cells and Tregs has been poorly described, and the efficacy of the recall response to common persistent infections in the elderly remains obscure. Here, we investigated alterations in the subpopulations of the B and T cells among 24 healthy young (aged 19-30) and 26 healthy elderly (aged 53-67) individuals. The analysis was performed by flow cytometry using freshly collected peripheral blood. γδTCR(+) T cells were overall decreased, while CD4(+)CD8(-) cells among γδTCR(+) T cells were increased in the elderly. Helios(+)Foxp3(+) and Helios(-)Foxp3(+) Treg cells were unaffected with age. Recent thymic emigrants, based on CD31 expression, were decreased among the Helios(+)Foxp3(+), but not the Helios(-)Foxp3(+) cell populations. We observed a decrease in Adenovirus-specific CD4(+) and CD8(+) T cells and an increase in CMV-specific CD4(+) T cells in the elderly. Similarly, INFγ(+)TNFα(+) double-positive cells were decreased among activated T cells after Adenovirus stimulation but increased after CMV stimulation. The data presented here indicate that γδTCR(+) T cells might stabilize B cells, and functional senescence might dominate at higher ages than those studied here.
Collapse
Affiliation(s)
- Ulrik Stervbo
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
- Deutsches Rheuma-Forschungszentrum – a Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany
- Marienhospital Herne, University Hospital of the Ruhr-University Bochum, Hölkeskampring 40, 44625 Herne, Germany
| | - Cecilia Bozzetti
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Udo Baron
- Epiontis GmbH, Rudower Chaussee 29, 12489 Berlin, Germany
| | - Karsten Jürchott
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Sarah Meier
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Julia Nora Mälzer
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Mikalai Nienen
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
- Marienhospital Herne, University Hospital of the Ruhr-University Bochum, Hölkeskampring 40, 44625 Herne, Germany
| | - Sven Olek
- Epiontis GmbH, Rudower Chaussee 29, 12489 Berlin, Germany
| | - Dominika Rachwalik
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Axel Ronald Schulz
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Avidan Neumann
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Nina Babel
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
- Marienhospital Herne, University Hospital of the Ruhr-University Bochum, Hölkeskampring 40, 44625 Herne, Germany
| | - Andreas Grützkau
- Deutsches Rheuma-Forschungszentrum – a Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany
| | - Andreas Thiel
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – University Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
104
|
Nacka-Aleksić M, Pilipović I, Stojić-Vukanić Z, Kosec D, Bufan B, Vujnović I, Arsenović-Ranin N, Dimitrijević M, Leposavić G. Sexual dimorphism in the aged rat CD4+ T lymphocyte-mediated immune response elicited by inoculation with spinal cord homogenate. Mech Ageing Dev 2015; 152:15-31. [PMID: 26408399 DOI: 10.1016/j.mad.2015.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 09/20/2015] [Indexed: 01/13/2023]
Abstract
Considering the crucial pathogenic role of CD4+ T cells in experimental autoimmune encephalomyelitis (EAE) and the opposite direction of the sexual dimorphism in the severity of the disease in 22-24-and 3-month-old dark agouti rats, sex differences in CD4+ T-cell-mediated immune response in aged rats immunized for EAE were examined and compared with those in young animals. In the inductive phase of EAE, fewer activated CD4+ lymphocytes were retrieved from draining lymph nodes of male (developing less severe disease) compared with female rats, due, at least partly, to their lesser expansion. The former reflected a greater suppressive capacity of CD4+CD25+Foxp3+ cells. Consequently, CD4+ lymphocyte infiltration into the spinal cord of aged male rats was diminished. At the peak of EAE, the frequency of reactivated cells was lower, whereas that of the regulatory CD4+ cells was higher in male rat spinal cord. Consistently, microglial activation and the expression of proinflammatory/damaging cytokines in male rat spinal cord mononuclear cells were diminished. Additionally, the frequency of the highly pathogenic IL-17+IFN-γ+ T lymphocytes infiltrating their spinal cord was lower. Together, these results point to (i) an age-specificity in CD4+ cell-mediated immune response and (ii) mechanisms underlying the sex differences in this response in aged rats.
Collapse
Affiliation(s)
- Mirjana Nacka-Aleksić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Duško Kosec
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Biljana Bufan
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivana Vujnović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Mirjana Dimitrijević
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Gordana Leposavić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia.
| |
Collapse
|
105
|
Sionov RV, Vlahopoulos SA, Granot Z. Regulation of Bim in Health and Disease. Oncotarget 2015; 6:23058-134. [PMID: 26405162 PMCID: PMC4695108 DOI: 10.18632/oncotarget.5492] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 08/08/2015] [Indexed: 11/25/2022] Open
Abstract
The BH3-only Bim protein is a major determinant for initiating the intrinsic apoptotic pathway under both physiological and pathophysiological conditions. Tight regulation of its expression and activity at the transcriptional, translational and post-translational levels together with the induction of alternatively spliced isoforms with different pro-apoptotic potential, ensure timely activation of Bim. Under physiological conditions, Bim is essential for shaping immune responses where its absence promotes autoimmunity, while too early Bim induction eliminates cytotoxic T cells prematurely, resulting in chronic inflammation and tumor progression. Enhanced Bim induction in neurons causes neurodegenerative disorders including Alzheimer's, Parkinson's and Huntington's diseases. Moreover, type I diabetes is promoted by genetically predisposed elevation of Bim in β-cells. On the contrary, cancer cells have developed mechanisms that suppress Bim expression necessary for tumor progression and metastasis. This review focuses on the intricate network regulating Bim activity and its involvement in physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University, Hadassah Medical School, Jerusalem, Israel
| | - Spiros A. Vlahopoulos
- First Department of Pediatrics, University of Athens, Horemeio Research Laboratory, Thivon and Levadias, Goudi, Athens, Greece
| | - Zvi Granot
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University, Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
106
|
Trzonkowski P, Bacchetta R, Battaglia M, Berglund D, Bohnenkamp HR, ten Brinke A, Bushell A, Cools N, Geissler EK, Gregori S, Marieke van Ham S, Hilkens C, Hutchinson JA, Lombardi G, Madrigal JA, Marek-Trzonkowska N, Martinez-Caceres EM, Roncarolo MG, Sanchez-Ramon S, Saudemont A, Sawitzki B. Hurdles in therapy with regulatory T cells. Sci Transl Med 2015; 7:304ps18. [PMID: 26355029 DOI: 10.1126/scitranslmed.aaa7721] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Improper activation of the immune system contributes to a variety of clinical conditions, including autoimmune and allergic diseases as well as solid organ and bone marrow transplantation. One approach to counteract this activation is through adoptive therapy with regulatory T cells (Tregs). Efforts to manufacture these cells have led to good maunfacturing practice-compliant protocols, and Treg products are entering early clinical trials. Here, we report the stance of the European Union Cooperation in Science and Technology Action BM1305, "Action to Focus and Accelerate Cell-based Tolerance-inducing Therapies-A FACTT," which identifies hurdles hindering Treg clinical applications in Europe and provides possible solutions.
Collapse
Affiliation(s)
- Piotr Trzonkowski
- Medical University of Gdansk, Department of Clinical Immunology and Transplantology, Debinki 7, 80-952 Gdansk, Poland. All authors equally contributed to this work.
| | - Rosa Bacchetta
- Department of Pediatric Stem Cell Transplantation and Regenerative Medicine, Stanford School of Medicine, Standford, California, USA
| | - Manuela Battaglia
- Diabetes Research Institute (DRI), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - David Berglund
- Uppsala University, Department of Immunology, Genetics and Pathology; Section of Clinical Immunology, Rudbeck Laboratory, 751 85 Uppsala, Sweden
| | | | - Anja ten Brinke
- Department of Immunopathology, Sanquin Blood Supply, Division Research, Plesmanlaan 125, 1066 CX Amsterdam, Netherland and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Andrew Bushell
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp University Hospital (UZA), Wilrijkstraat 10, B-2650 Edegem, Belgium
| | - Edward K Geissler
- Division of Experimental Surgery, Department of Surgery, University Hospital Regensburg, Regensburg, Bavaria, 93053, Germany
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Blood Supply, Division Research, Plesmanlaan 125, 1066 CX Amsterdam, Netherland and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | | | - James A Hutchinson
- Division of Experimental Surgery, Department of Surgery, University Hospital Regensburg, Regensburg, 93053, Bavaria, Germany
| | - Giovanna Lombardi
- Medical Research Council (MRC) Centre in Transplantation, Kings College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - J Alejandro Madrigal
- Anthony Nolan Research Institute, University College London (UCL) Royal Free Hospital Campus, 77c Fleet Road, London NW3 2QG, UK
| | | | - Eva M Martinez-Caceres
- Immunology Division, Germans Trias i Pujol University Hospital. Campus Can Ruti. Department of Cellular Biology, Physiology, and Immunology, Universitat Autònoma Barcelona 08916, Badalona, Barcelona, Spain
| | - Maria Grazia Roncarolo
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy. Department of Pediatric Stem Cell Transplantation and Regenerative Medicine, Stanford School of Medicine, Stanford, California, USA
| | - Silvia Sanchez-Ramon
- Departamento de Inmunología Clínica, Hospital Clínico San Carlos, Calle Profesor Martín Lagos S/N, E- 28040 Madrid, Spain
| | - Aurore Saudemont
- Anthony Nolan Research Institute, University College London (UCL) Royal Free Hospital Campus, 77c Fleet Road, London NW3 2QG, UK
| | - Birgit Sawitzki
- AG Transplantationstoleranz, Charite Universitätsmedizin, Institut für Med. Imunologie, Augustenburgerplatz 1, 13353 Berlin, Germany
| |
Collapse
|
107
|
Raynor J, Karns R, Almanan M, Li KP, Divanovic S, Chougnet CA, Hildeman DA. IL-6 and ICOS Antagonize Bim and Promote Regulatory T Cell Accrual with Age. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:944-52. [PMID: 26109645 PMCID: PMC4506860 DOI: 10.4049/jimmunol.1500443] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/24/2015] [Indexed: 12/19/2022]
Abstract
Regulatory T cells (Tregs), a subset of CD4(+) T cells, dramatically accumulate with age in humans and mice and contribute to age-related immune suppression. Recently, we showed that a majority of accumulating Tregs in aged mice expressed low levels of CD25, and their accrual is associated with declining levels of IL-2 in aged mice. In this study, we further investigated the origin of CD25(lo) Tregs in aged mice. First, aged Tregs had high expression of neuropilin-1 and Helios, and had a broad Vβ repertoire. Next, we analyzed the gene expression profile of Tregs, naive T cells, and memory T cells in aged mice. We found that the gene expression profile of aged CD25(lo) Tregs were more related to young CD25(lo) Tregs than to either naive or memory T cells. Further, the gene expression profile of aged Tregs was consistent with recently described "effector" Tregs (eTregs). Additional analysis revealed that nearly all Tregs in aged mice were of an effector phenotype (CD44(hi)CD62L(lo)) and could be further characterized by high levels of ICOS and CD69. ICOS contributed to Treg maintenance in aged mice, because in vivo Ab blockade of ICOSL led to a loss of eTregs, and this loss was rescued in Bim-deficient mice. Further, serum levels of IL-6 increased with age and contributed to elevated expression of ICOS on aged Tregs. Finally, Treg accrual was significantly blunted in aged IL-6-deficient mice. Together, our data show a role for IL-6 in promoting eTreg accrual with age likely through maintenance of ICOS expression.
Collapse
Affiliation(s)
- Jana Raynor
- Division of Immunobiology, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Rebekah Karns
- Division of Biomedical Informatics, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH 45229; and
| | - Maha Almanan
- Division of Immunobiology, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Kun-Po Li
- Division of Immunobiology, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Senad Divanovic
- Division of Immunobiology, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH 45229; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Claire A Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH 45229; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - David A Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH 45229; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| |
Collapse
|
108
|
Vukmanovic-Stejic M, Sandhu D, Seidel JA, Patel N, Sobande TO, Agius E, Jackson SE, Fuentes-Duculan J, Suárez-Fariñas M, Mabbott NA, Lacy KE, Ogg G, Nestle FO, Krueger JG, Rustin MHA, Akbar AN. The Characterization of Varicella Zoster Virus-Specific T Cells in Skin and Blood during Aging. J Invest Dermatol 2015; 135:1752-1762. [PMID: 25734814 PMCID: PMC4471118 DOI: 10.1038/jid.2015.63] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 01/14/2015] [Accepted: 02/03/2015] [Indexed: 12/12/2022]
Abstract
Reactivation of the varicella zoster virus (VZV) increases during aging. Although the effects of VZV reactivation are observed in the skin (shingles), the number and functional capacity of cutaneous VZV-specific T cells have not been investigated. The numbers of circulating IFN-γ-secreting VZV-specific CD4(+) T cells are significantly decreased in old subjects. However, other measures of VZV-specific CD4(+) T cells, including proliferative capacity to VZV antigen stimulation and identification of VZV-specific CD4(+) T cells with an major histocompatibility complex class II tetramer (epitope of IE-63 protein), were similar in both age groups. The majority of T cells in the skin of both age groups expressed CD69, a characteristic of skin-resident T cells. VZV-specific CD4(+) T cells were significantly increased in the skin compared with the blood in young and old subjects, and their function was similar in both age groups. In contrast, the number of Foxp3(+) regulatory T cells and expression of the inhibitory receptor programmed cell death -1 PD-1 on CD4(+) T cells were significantly increased in the skin of older humans. Therefore, VZV-specific CD4(+) T cells in the skin of older individuals are functionally competent. However, their activity may be restricted by multiple inhibitory influences in situ.
Collapse
Affiliation(s)
| | - Daisy Sandhu
- Division of Infection and Immunity, University College London, London, UK; Department of Dermatology, Royal Free Hospital, London, UK
| | - Judith A Seidel
- Division of Infection and Immunity, University College London, London, UK
| | - Neil Patel
- Division of Infection and Immunity, University College London, London, UK; Department of Dermatology, Royal Free Hospital, London, UK
| | - Toni O Sobande
- Division of Infection and Immunity, University College London, London, UK
| | - Elaine Agius
- Division of Infection and Immunity, University College London, London, UK; Department of Dermatology, Royal Free Hospital, London, UK
| | - Sarah E Jackson
- Division of Infection and Immunity, University College London, London, UK
| | | | | | - Neil A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, UK
| | - Katie E Lacy
- St Johns Institute of Dermatology, Guys and St Thomas' Hospital, London, UK
| | - Graham Ogg
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Frank O Nestle
- St Johns Institute of Dermatology, Guys and St Thomas' Hospital, London, UK
| | - James G Krueger
- Laboratory for Investigative Dermatology, Rockefeller University, New York, USA
| | | | - Arne N Akbar
- Division of Infection and Immunity, University College London, London, UK.
| |
Collapse
|
109
|
Vitamin D deficiency contributes to the reduction and impaired function of naïve CD45RA⁺ regulatory T cell in chronic heart failure. J Immunol Res 2015; 2015:547697. [PMID: 25984539 PMCID: PMC4423006 DOI: 10.1155/2015/547697] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/02/2015] [Accepted: 03/02/2015] [Indexed: 01/29/2023] Open
Abstract
The effect of vitamin D pertinent to cardiovascular health on the heart itself is considered to shift toward an anti-inflammatory response in chronic heart failure (CHF); however, its underlying mechanism is not completely understood. In this study, we demonstrated that plasma 25(OH)D level, negatively associated with NT-ProBNP, correlated with the decreased Treg in CHF compared to the patients with other cardiovascular diseases and healthy and older donors. Naïve Treg cell (CD4(+)CD45RA(+)Foxp3(lo)T) subset, rather than whole Treg cells, contributes to the reduction of Treg in CHF. 1,25(OH)2D treatment maintained partial expression of CD45RA on CD4(+)T cell after αCD3/CD28 monoclonal antibodies activation and ameliorated the impaired CD4(+)CD45RA(+)T cell function from CHF patients through upregulating Foxp3 expression and IL-10 secretion in vitro. Low level of vitamin D receptor (VDR) was detected in CD4(+)CD45RA(+)T cell of CHF than control, while 1,25(OH)2D treatment increased the VDR expression to exert its immunosuppression on T cell. The results of this study might provide tangible evidence to our knowledge of the impact of vitamin D supplementation on naïve Tregs, which may offer new means of preventing and treating CHF.
Collapse
|
110
|
Derhovanessian E, Chen S, Maier AB, Hähnel K, de Craen AJM, Roelofs H, Westendorp R, Pawelec G. CCR4+ Regulatory T Cells Accumulate in the Very Elderly and Correlate With Superior 8-Year Survival. J Gerontol A Biol Sci Med Sci 2015; 70:917-23. [PMID: 25852090 DOI: 10.1093/gerona/glu128] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 07/01/2014] [Indexed: 12/27/2022] Open
Abstract
CD4(+) regulatory T cells (Tregs) are a distinct population of T cells involved in maintaining peripheral tolerance to self-antigens. Several studies have shown increased frequency and number of Tregs in the elderly. Whether such an increase has any clinical relevance has not been addressed. Here, we have analyzed circulating Tregs in 114 donors between the ages of 18 and 89 years and assessed their implications for survival of the very elderly. In line with previously published data, we observed higher proportions of Tregs in the elderly. Expression of chemokine receptor 4 (CCR4) by Tregs has been shown to characterize antigen-primed activated Tregs with immediate suppressive function. Thus we further analyzed Tregs expressing or lacking this chemokine receptor. There were more CCR4(+) and CCR4(-) Tregs in the elderly than the young. Finally, using a subset of 48 elderly donors participating in the Leiden 85-plus study we documented that people with greater median frequencies of CCR4(+) Tregs enjoyed a better 8-year survival rate than those with lower frequencies of these cells. Our data, demonstrating for the first time a positive correlation between increased frequency of Tregs and survival in the elderly, imply an increasing importance of controlling inappropriate immune responses and inflammation as we grew old.
Collapse
Affiliation(s)
- Evelyna Derhovanessian
- Department of Internal Medicine II, Centre for Medical Research, University of Tübingen, Tübingen, Germany.
| | - Sijia Chen
- Department of Gerontology and Geriatrics and
| | - Andrea B Maier
- Department of Internal Medicine, Section of Gerontology and Geriatrics, VU University Medical Centre, Amsterdam, the Netherlands
| | - Karin Hähnel
- Department of Internal Medicine II, Centre for Medical Research, University of Tübingen, Tübingen, Germany
| | | | - Helene Roelofs
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, the Netherlands
| | - Rudi Westendorp
- Department of Gerontology and Geriatrics and Netherlands Consortium for Health Aging, Leiden University Medical Centre, Leiden, the Netherlands
| | - Graham Pawelec
- Department of Internal Medicine II, Centre for Medical Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
111
|
Divergent effects of type-I interferons on regulatory T cells. Cytokine Growth Factor Rev 2015; 26:133-41. [DOI: 10.1016/j.cytogfr.2014.10.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 10/22/2014] [Indexed: 02/07/2023]
|
112
|
Zhang Q, Jia Q, Deng T, Song B, Li L. Heterogeneous expansion of CD4+ tumor-infiltrating T-lymphocytes in clear cell renal cell carcinomas. Biochem Biophys Res Commun 2015; 458:70-6. [PMID: 25637538 DOI: 10.1016/j.bbrc.2015.01.069] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 01/16/2015] [Indexed: 11/18/2022]
Abstract
Aberrant expression of tumor-associated antigens (TAAs) mediates the effective mounting of adaptive immunity in human solid tumors. The foundations of this tumor-host interaction strongly depend on specific recognition via TAA-cognate-receptors in T-cell repertoires. Previous studies focused on the phenotypic and functional properties of CD4+/CD8+ tumor-infiltrating T-lymphocytes (TILs), but the detailed composition of T-cell repertoires of these fundamental subsets remains largely unknown. This study recruited 10 clear cell renal cell carcinoma (ccRCC) patients and obtained samples from various tissues, including tumors, adjacent healthy renal tissue and peripheral blood. We utilized deep sequencing of T-cell receptor beta chains (TCRB), which serve as a unique identifier for each T clonotype, to characterize the CD4+/CD8+ TIL repertoire in ccRCC patients, assess the diversity and clonality of infiltrated T-cells in distinct tissues from patients and depict the clonal expansion events that occur in anti-tumor immune responses. We found that the CD4+ TIL repertoire exhibited signatures of heterogeneous T-cell expansion, which were characterized by divergent TRBV/J usage and an enrichment of expanded dominant clones. Taken together, our findings provide additional evidence of CD4+ T-cell-mediated anti-tumor immunity. The identification of the underlying molecular mechanisms of this process may provide novel avenues for targeted immunotherapeutic interventions.
Collapse
MESH Headings
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- CD4-Positive T-Lymphocytes/physiology
- CD8-Positive T-Lymphocytes/pathology
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/pathology
- Humans
- Kidney Neoplasms/immunology
- Kidney Neoplasms/pathology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Lymphocytes, Tumor-Infiltrating/physiology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Tumor Microenvironment/genetics
Collapse
Affiliation(s)
- Qian Zhang
- Department of Urology, XinQiao Hospital, Third Military Medical University, Chongqing, China; Department of Urology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qingzhu Jia
- Biomedical Analysis Center, Third Military Medical University, Chongqing, China
| | - Tianxing Deng
- Department of Urology, XinQiao Hospital, Third Military Medical University, Chongqing, China
| | - Bo Song
- Department of Urology, Southwest Hospital, Third Military Medical University, Chongqing, China.
| | - Longkun Li
- Department of Urology, XinQiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
113
|
Braza F, Dugast E, Panov I, Paul C, Vogt K, Pallier A, Chesneau M, Baron D, Guerif P, Lei H, Laplaud DA, Volk HD, Degauque N, Giral M, Soulillou JP, Sawitzki B, Brouard S. Central Role of CD45RA- Foxp3hi Memory Regulatory T Cells in Clinical Kidney Transplantation Tolerance. J Am Soc Nephrol 2015; 26:1795-805. [PMID: 25556168 DOI: 10.1681/asn.2014050480] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/19/2014] [Indexed: 12/21/2022] Open
Abstract
The role of Foxp3(+) regulatory T cells (Tregs) in operational tolerance remains elusive, as initial results revealed an increased frequency of this subset in tolerant patients but no functional differences compared with immunosuppressed recipients. In addition, recent studies of regulatory B cells strongly suggest that Tregs may not have a central role in kidney transplantation tolerance. However, recent investigations of the crucial role of Foxp3 demethylation in Treg function and the possibility of identifying distinct Foxp3 T cell subsets prompted us to more thoroughly characterize Tregs in operationally tolerant patients. Thus, we studied the level of demethylation of the Foxp3 Treg-specific demethylated region (TSDR) in circulating CD4(+) T cells and analyzed Treg subset frequency in tolerant patients, healthy volunteers, patients with stable graft function under immunosuppression, and chronically rejecting recipients. We observed a higher proportion of CD4(+) T cells with demethylated Foxp3 and a specific expansion of CD4(+) CD45RA(-) Foxp3(hi) memory Tregs exclusively in tolerant patients. The memory Tregs of tolerant recipients exhibited increased Foxp3 TSDR demethylation, expressed higher levels of CD39 and glucocorticoid-induced TNF-related receptor, and harbored greater suppressive properties than memory Tregs from patients with stable graft function. Taken together, our data demonstrate that operationally tolerant patients mobilize an array of potentially suppressive cells, including not only regulatory B cells but also Tregs. Our results also indicate that tolerant patients have potent CD4(+)CD45RA(-) Foxp3(hi) memory Tregs with a specific Foxp3 TSDR demethylation pattern, which may contribute to the maintenance of graft tolerance.
Collapse
Affiliation(s)
- Faouzi Braza
- Faculty of Medicine, University of Nantes, Nantes, France; French Institute of Health and Medical Research Unit 1064, Research Institute on Urology, Nephrology, and Transplantation, and
| | - Emilie Dugast
- French Institute of Health and Medical Research Unit 1064, Research Institute on Urology, Nephrology, and Transplantation, and Biotherapy Clinical Investigation Center, Hôtel Dieu University Hospital, Nantes, France; and
| | - Ivo Panov
- Institute of Medical Immunology and Berlin Brandenburg Center for Regenerative Therapies, Charité Medical University, Berlin, Germany
| | - Chloé Paul
- French Institute of Health and Medical Research Unit 1064, Research Institute on Urology, Nephrology, and Transplantation, and Biotherapy Clinical Investigation Center, Hôtel Dieu University Hospital, Nantes, France; and
| | | | - Annaick Pallier
- French Institute of Health and Medical Research Unit 1064, Research Institute on Urology, Nephrology, and Transplantation, and Biotherapy Clinical Investigation Center, Hôtel Dieu University Hospital, Nantes, France; and
| | - Mélanie Chesneau
- Faculty of Medicine, University of Nantes, Nantes, France; French Institute of Health and Medical Research Unit 1064, Research Institute on Urology, Nephrology, and Transplantation, and
| | - Daniel Baron
- Faculty of Medicine, University of Nantes, Nantes, France; French Institute of Health and Medical Research Unit 1064, Research Institute on Urology, Nephrology, and Transplantation, and
| | - Pierrick Guerif
- Biotherapy Clinical Investigation Center, Hôtel Dieu University Hospital, Nantes, France; and
| | - Hong Lei
- Institute of Medical Immunology and Berlin Brandenburg Center for Regenerative Therapies, Charité Medical University, Berlin, Germany
| | - David-Axel Laplaud
- French Institute of Health and Medical Research Unit 1064, Research Institute on Urology, Nephrology, and Transplantation, and Biotherapy Clinical Investigation Center, Hôtel Dieu University Hospital, Nantes, France; and
| | - Hans-Dieter Volk
- Institute of Medical Immunology and Berlin Brandenburg Center for Regenerative Therapies, Charité Medical University, Berlin, Germany
| | - Nicolas Degauque
- French Institute of Health and Medical Research Unit 1064, Research Institute on Urology, Nephrology, and Transplantation, and Biotherapy Clinical Investigation Center, Hôtel Dieu University Hospital, Nantes, France; and
| | - Magali Giral
- French Institute of Health and Medical Research Unit 1064, Research Institute on Urology, Nephrology, and Transplantation, and Biotherapy Clinical Investigation Center, Hôtel Dieu University Hospital, Nantes, France; and
| | - Jean-Paul Soulillou
- French Institute of Health and Medical Research Unit 1064, Research Institute on Urology, Nephrology, and Transplantation, and Biotherapy Clinical Investigation Center, Hôtel Dieu University Hospital, Nantes, France; and
| | - Birgit Sawitzki
- Institute of Medical Immunology and Berlin Brandenburg Center for Regenerative Therapies, Charité Medical University, Berlin, Germany
| | - Sophie Brouard
- French Institute of Health and Medical Research Unit 1064, Research Institute on Urology, Nephrology, and Transplantation, and Biotherapy Clinical Investigation Center, Hôtel Dieu University Hospital, Nantes, France; and
| |
Collapse
|
114
|
Yu K, Dong Q, Mao X, Meng K, Zhao X, Ji Q, Wu B, Zhong Y, Zhu Z, Liu Y, Zhang W, Tony H, Shi H, Zeng Q. Disruption of the TSLP-TSLPR-LAP signaling between epithelial and dendritic cells through hyperlipidemia contributes to regulatory T-Cell defects in atherosclerotic mice. Atherosclerosis 2014; 238:278-88. [PMID: 25544178 DOI: 10.1016/j.atherosclerosis.2014.12.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/09/2014] [Accepted: 12/09/2014] [Indexed: 01/08/2023]
Abstract
Regulatory T-Cells (Tregs) play a protective role against the development of atherosclerosis. Moreover, thymic stromal lymphopoietin (TSLP)/thymic stromal lymphopoietin receptor (TSLPR) signaling in myeloid dendritic cells (DCs) promote Treg differentiation. Here, we examined the potential role of TSLP/TSLPR on Treg homeostasis in atherosclerosis. The frequencies of both latency-associated peptide (LAP)(+) and Foxp3(+) Tregs were reduced in the thymus and spleen of ApoE(-/-) mice compared with C57BL/6 mice, and this effect was associated with decreased thymic output. The tolerogenic function of DCs obtained from ApoE(-/-) mice was compromised compared with those from C57BL/6 mice. The expression of TSLP and TSLPR was also inhibited in ApoE(-/-) mice. In addition, we found that ox-LDL attenuated TSLP expression in cultured thymic epithelial cells (TECs) through the activation of retinoid X receptor alpha (RXRA) and IL-1β and decreased LAP and PD-L1 expression in oxLDL-activated DCs while both were up-regulated in TSLP-activated DCs. We also observed that the TSLP-DCs mediated differentiation of Tregs was abrogated through LAP neutralization. Furthermore, TSLP injection rescued Treg defects in ApoE(-/-) mice. These findings suggest that Treg defects in ApoE(-/-) mice might partially be attributed to the disruption of TSLP-TSLPR-LAP signaling in epithelial cells (ECs) and DCs.
Collapse
Affiliation(s)
- Kunwu Yu
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Dong
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobo Mao
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Meng
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqi Zhao
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qingwei Ji
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Bangwei Wu
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yucheng Zhong
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengfeng Zhu
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yuzhou Liu
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhang
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hasahya Tony
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Huairui Shi
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qiutang Zeng
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
115
|
Pesenacker AM, Broady R, Levings MK. Control of tissue-localized immune responses by human regulatory T cells. Eur J Immunol 2014; 45:333-43. [PMID: 25378065 DOI: 10.1002/eji.201344205] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 09/30/2014] [Accepted: 10/31/2014] [Indexed: 12/25/2022]
Abstract
Treg cells control immune responses to self and nonharmful foreign antigens. Emerging data from animal models indicate that Treg cells function in both secondary lymphoid organs and tissues, and that these different microenvironments may contain specialized subsets of Treg cells with distinct mechanisms of action. The design of therapies for the restoration of tissue-localized immune homeostasis is dependent upon understanding how local immune responses are influenced by Treg cells in health versus disease. Here we review the current state of knowledge about human Treg cells in four locations: the skin, lung, intestine, and joint. Despite the distinct biology of these tissues, there are commonalities in the biology of their resident Treg cells, including phenotypic and functional differences from circulating Treg cells, and the presence of cytokine-producing (e.g. IL-17(+)) FOXP3(+) cells. We also highlight the challenges to studying tissue Treg cells in humans, and opportunities to use new technologies for the detailed analysis of Treg cells at the single-cell level. As emerging biological therapies are increasingly targeted toward tissue-specific effects, it is critical to understand their potential impact on local immune regulation.
Collapse
Affiliation(s)
- Anne M Pesenacker
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; Child & Family Research Institute, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
116
|
Moore C, Tejon G, Fuentes C, Hidalgo Y, Bono MR, Maldonado P, Fernandez R, Wood KJ, Fierro JA, Rosemblatt M, Sauma D, Bushell A. Alloreactive regulatory T cells generated with retinoic acid prevent skin allograft rejection. Eur J Immunol 2014; 45:452-63. [PMID: 25381698 DOI: 10.1002/eji.201444743] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 10/06/2014] [Accepted: 11/06/2014] [Indexed: 12/24/2022]
Abstract
CD4(+) CD25(+) Foxp3(+) regulatory T (Treg) cells mediate immunological self-tolerance and suppress immune responses. Retinoic acid (RA), a natural metabolite of vitamin A, has been reported to enhance the differentiation of Treg cells in the presence of TGF-β. In this study, we show that the co-culture of naive T cells from C57BL/6 mice with allogeneic antigen-presenting cells (APCs) from BALB/c mice in the presence of TGF-β, RA, and IL-2 resulted in a striking enrichment of Foxp3(+) T cells. These RA in vitro-induced regulatory T (RA-iTreg) cells did not secrete Th1-, Th2-, or Th17-related cytokines, showed a nonbiased homing potential, and expressed several cell surface molecules related to Treg-cell suppressive potential. Accordingly, these RA-iTreg cells suppressed T-cell proliferation and inhibited cytokine production by T cells in in vitro assays. Moreover, following adoptive transfer, RA-iTreg cells maintained Foxp3 expression and their suppressive capacity. Finally, RA-iTreg cells showed alloantigen-specific immunosuppressive capacity in a skin allograft model in immunodeficient mice. Altogether, these data indicate that functional and stable allogeneic-specific Treg cells may be generated using TGF-β, RA, and IL-2. Thus, RA-iTreg cells may have a potential use in the development of more effective cellular therapies in clinical transplantation.
Collapse
Affiliation(s)
- Carolina Moore
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile; Facultad de Ciencias Biologicas, Universidad Andres Bello, Santiago, Chile
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Coe DJ, Kishore M, Marelli-Berg F. Metabolic regulation of regulatory T cell development and function. Front Immunol 2014; 5:590. [PMID: 25477880 PMCID: PMC4235430 DOI: 10.3389/fimmu.2014.00590] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/04/2014] [Indexed: 11/26/2022] Open
Abstract
It is now well established that the effector T cell (Teff) response is regulated by a series of metabolic switches. Quiescent T cells predominantly require adenosine triphosphate-generating processes, whereas proliferating Teff require high metabolic flux through growth-promoting pathways, such as glycolysis. Pathways that control metabolism and immune cell function are intimately linked, and changes in cell metabolism at both the cell and system levels have been shown to enhance or suppress specific T cell effector functions. Furthermore, functionally distinct T cell subsets require distinct energetic and biosynthetic pathways to support their specific functional needs. In particular, naturally occurring regulatory T cells (Treg) are characterized by a unique metabolic signature distinct to that of conventional Teff cells. We here briefly review the signaling pathways that control Treg metabolism and how this metabolic phenotype integrates their differentiation and function. Ultimately, these metabolic features may provide new opportunities for the therapeutic modulation of unwanted immune responses.
Collapse
Affiliation(s)
- David John Coe
- Department of Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University , London , UK
| | - Madhav Kishore
- Department of Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University , London , UK
| | - Federica Marelli-Berg
- Department of Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University , London , UK
| |
Collapse
|
118
|
Clinical Impact of Regulatory T cells (Treg) in Cancer and HIV. CANCER MICROENVIRONMENT 2014; 8:201-7. [PMID: 25385463 DOI: 10.1007/s12307-014-0159-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 10/06/2014] [Indexed: 12/31/2022]
Abstract
The role of regulatory T cells, (Treg) in human cancer and HIV-1 infections has been under intense scrutiny. While the lack of a marker specific for human Treg has made it challenging to phenotype these cells, combinations of several markers and functional attributes of Treg have made it possible to assess their contributions to immune homeostasis in health and disease. Treg diversity and their plasticity create a challenge in deciding whether they are beneficial to the host by down-regulating excessive immune activation or are responsible for adverse effects such as suppression of anti-tumor immune responses resulting in promotion of tumor growth. Treg are emerging as active participants in several biochemical pathways involved in immune regulation. This review attempts to integrate current information about human Treg in respect to their activities in cancer and HIV-1. The goal is to evaluate the potential of Treg as targets for future immune or pharmacologic therapies for cancer or HIV-1 infections.
Collapse
|
119
|
Mota C, Nunes-Silva V, Pires AR, Matoso P, Victorino RMM, Sousa AE, Caramalho I. Delta-like 1-mediated Notch signaling enhances the in vitro conversion of human memory CD4 T cells into FOXP3-expressing regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:5854-62. [PMID: 25367118 DOI: 10.4049/jimmunol.1400198] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
FOXP3-expressing regulatory T cells (Treg) are essential for the prevention of autoimmunity and were shown to be reduced and/or dysfunctional in several autoimmune diseases. Although Treg-based adoptive transfer represents a promising therapy, the large cell number required to achieve clinical efficacy constitutes an important limitation. Therefore, novel strategies to generate bona fide in vitro-induced Treg (iTreg) are critical. In this study, we report that human memory CD4 T cells can be efficiently converted into iTreg, and that Delta-like 1 (DL1)-mediated Notch signaling significantly enhances this process. The iTreg generated in the presence of DL1 featured higher levels of Treg function-associated molecules and were efficient suppressors. Importantly, these iTreg displayed a stable phenotype in long-term cultures, even in the presence of proinflammatory cytokines. Additionally, DL1 potentiated FOXP3 acquisition by memory CD4 cells through the modulation of the TGF-β signaling pathway and of Foxp3 transcription. Our data demonstrate that iTreg can be efficiently induced from memory CD4 cells, a subset enriched in relevant specificities for targeting in autoimmune diseases, and that DL1 enhances this process. DL1 also enhanced the proliferation and Treg function-associated marker expression of ex vivo-stimulated human circulating FOXP3(+) cells. Manipulation of the Notch signaling pathway constitutes a promising approach to boost the in vitro generation of iTreg and ex vivo Treg expansion, thus facilitating the establishment of effective Treg-based adoptive therapy in autoimmune diseases.
Collapse
Affiliation(s)
- Catarina Mota
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; and Hospital Universitário de Santa Maria, Centro Hospitalar Lisboa Norte, 1649-035 Lisbon, Portugal
| | - Vânia Nunes-Silva
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; and
| | - Ana R Pires
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; and
| | - Paula Matoso
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; and
| | - Rui M M Victorino
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; and Hospital Universitário de Santa Maria, Centro Hospitalar Lisboa Norte, 1649-035 Lisbon, Portugal
| | - Ana E Sousa
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; and
| | - Iris Caramalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; and
| |
Collapse
|
120
|
Taubert R, Hardtke-Wolenski M, Noyan F, Wilms A, Baumann AK, Schlue J, Olek S, Falk CS, Manns MP, Jaeckel E. Intrahepatic regulatory T cells in autoimmune hepatitis are associated with treatment response and depleted with current therapies. J Hepatol 2014; 61:1106-14. [PMID: 24882050 DOI: 10.1016/j.jhep.2014.05.034] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 05/20/2014] [Accepted: 05/22/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Autoimmune hepatitis (AIH) is a chronic autoimmune liver disease usually requiring life-long immunosuppression. The mechanisms for disease initiation and chronicity are largely unknown. A contribution of deficient regulatory T cells (Tregs) in the blood was controversially discussed recently. So far investigations in the target organ have been limited to single parameter analysis in untreated AIH. METHODS We retrospectively analysed the pattern of liver infiltrating T, B and regulatory T cells quantitatively with simultaneous multicolour immunofluorescence before (n=45) and under (n=31) therapy in adult AIH type 1 (AIH-1) patients. RESULTS Intrahepatic CD4(+) cells dominate over CD8(+) at diagnosis, but with increasing disease activity the CD4(+)/CD8(+) ratio approached one. While there is no change of Tregs in the blood, they are enriched with effector T cells (Teffs) within the liver of patients with untreated AIH-1 with a constant Treg/Teff ratio. Even more importantly, immunosuppression mostly with steroids and azathioprine caused a disproportional loss of intrahepatic Tregs. Patients reaching biochemical remission had higher intrahepatic Treg/Teff and Treg/B cell ratios compared to patients failing to reach remission. In vitro proliferation of Tregs seemed to be more suppressed by prednisolone than expansion of Teffs. Furthermore, intraportal B cells correlated with serum IgG suggesting an autochthonous intrahepatic IgG production. CONCLUSIONS Intrahepatic Tregs are rather enriched than numerically deficient in untreated AIH-1. The disproportional decrease of intrahepatic Tregs during therapy might explain high relapse rates after discontinuation of immunosuppression. Thus, future therapies increasing intrahepatic immunoregulation might be better suited for long-term control of AIH.
Collapse
Affiliation(s)
- Richard Taubert
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
| | | | - Fatih Noyan
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
| | - Artur Wilms
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
| | - Anna K Baumann
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
| | - Jerome Schlue
- Department of Pathology, Hannover Medical School, Germany
| | - Sven Olek
- Ivana Türbachová Laboratory for Epigenetics, Epiontis GmbH, Berlin, Germany
| | - Christine S Falk
- Institute of Transplantation Immunology and Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
| | - Michael P Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
| | - Elmar Jaeckel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany.
| |
Collapse
|
121
|
Liao KL, Bai XF, Friedman A. Mathematical modeling of Interleukin-35 promoting tumor growth and angiogenesis. PLoS One 2014; 9:e110126. [PMID: 25356878 PMCID: PMC4214702 DOI: 10.1371/journal.pone.0110126] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 09/17/2014] [Indexed: 01/18/2023] Open
Abstract
Interleukin-35 (IL-35), a cytokine from the Interleukin-12 cytokine family, has been considered as an anti-inflammatory cytokine which promotes tumor progression and tumor immune evasion. It has also been demonstrated that IL-35 is secreted by regulatory T cells. Recent mouse experiments have shown that IL-35 produced by cancer cells promotes tumor growth via enhancing myeloid cell accumulation and angiogenesis, and reducing the infiltration of activated CD8[Formula: see text] T cells into tumor microenvironment. In the present paper we develop a mathematical model based on these experimental results. We include in the model an anti-IL-35 drug as treatment. The extended model (with drug) is used to design protocols of anti-IL-35 injections for treatment of cancer. We find that with a fixed total amount of drug, continuous injection has better efficacy than intermittent injections in reducing the tumor load while the treatment is ongoing. We also find that the percentage of tumor reduction under anti-IL-35 treatment improves when the production of IL-35 by cancer is increased.
Collapse
Affiliation(s)
- Kang-Ling Liao
- Mathematical Biosciences Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Xue-Feng Bai
- Department of Pathology and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Avner Friedman
- Mathematical Biosciences Institute, The Ohio State University, Columbus, Ohio, United States of America
- Department of Mathematics, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
122
|
d’Hennezel E, Piccirillo CA. Functional plasticity in human FOXP3+regulatory T cells. Hum Vaccin Immunother 2014; 8:1001-5. [DOI: 10.4161/hv.20203] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
123
|
Weed DT, Vella JL, Reis IM, De la Fuente AC, Gomez C, Sargi Z, Nazarian R, Califano J, Borrello I, Serafini P. Tadalafil reduces myeloid-derived suppressor cells and regulatory T cells and promotes tumor immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res 2014; 21:39-48. [PMID: 25320361 DOI: 10.1158/1078-0432.ccr-14-1711] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg) play a key role in the progression of head and neck squamous cell carcinoma (HNSCC). On the basis of our preclinical data demonstrating that phosphodiesterase-5 (PDE5) inhibition can modulate these cell populations, we evaluated whether the PDE5 inhibitor tadalafil can revert tumor-induced immunosuppression and promote tumor immunity in patients with HNSCC. EXPERIMENTAL DESIGN First, we functionally and phenotypically characterized MDSCs in HNSCCs and determined, retrospectively, whether their presence at the tumor site correlates with recurrence. Then, we performed a prospective single-center, double-blinded, randomized, three-arm study in which patients with HNSCC undergoing definitive surgical resection of oral and oropharyngeal tumors were treated with tadalafil 10 mg/day, 20 mg/day, or placebo for at least 20 days preoperatively. Blood and tumor MDSC and Treg presence and CD8(+) T-cell reactivity to tumor antigens were evaluated before and after treatment. RESULTS MDSCs were characterized in HNSCC and their intratumoral presence significantly correlates with recurrence. Tadalafil treatment was well tolerated and significantly reduced both MDSCs and Treg concentrations in the blood and in the tumor (P < 0.05). In addition, the concentration of blood CD8(+) T cells reactive to autologous tumor antigens significantly increased after treatment (P < 0.05). Tadalafil immunomodulatory activity was maximized at an intermediate dose but not at higher doses. Mechanistic analysis suggests a possible off-target effect on PDE11 at high dosages that, by increasing intracellular cAMP, may negatively affect antitumor immunity. CONCLUSIONS Tadalafil seems to beneficially modulate the tumor micro- and macro-environment in patients with HNSCC by lowering MDSCs and Tregs and increasing tumor-specific CD8(+) T cells in a dose-dependent fashion.
Collapse
Affiliation(s)
- Donald T Weed
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Jennifer L Vella
- Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Isildinha M Reis
- Department of Public Health Sciences and Sylvester Biostatistics and Bioinformatics Core Resource, University of Miami, Miller School of Medicine, Miami, Florida
| | - Adriana C De la Fuente
- Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Carmen Gomez
- Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Zoukaa Sargi
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Ronen Nazarian
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Joseph Califano
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, Maryland.,Milton J. Dance Head and Neck Center, Greater Baltimore Medical Center, Baltimore, Maryland
| | - Ivan Borrello
- Oncology Department, Johns Hopkins University, Baltimore, Maryland
| | - Paolo Serafini
- Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| |
Collapse
|
124
|
Kelderman S, Schumacher TNM, Haanen JBAG. Acquired and intrinsic resistance in cancer immunotherapy. Mol Oncol 2014; 8:1132-9. [PMID: 25106088 PMCID: PMC5528612 DOI: 10.1016/j.molonc.2014.07.011] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 07/13/2014] [Indexed: 01/08/2023] Open
Abstract
A number of immunotherapies, in particular immune checkpoint targeting antibodies and adoptive T-cell therapies, are starting to transform the treatment of advanced cancers. The likelihood to respond to these immunotherapies differs strongly across tumor types, with response rates for checkpoint targeting being the highest in advanced melanoma, renal cell cancer and non-small cell lung cancer. However, also non-responsiveness is observed, indicating the presence of intrinsic resistance or naturally acquired resistance. In addition, a subgroup of patients that do initially respond to immunotherapy will later recur, thereby also pointing towards a role of therapy-induced acquired resistance. Here, we review our current understanding of both intrinsic and acquired resistance mechanisms in cancer immunotherapy, and discuss potential strategies to overcome them.
Collapse
Affiliation(s)
- Sander Kelderman
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Ton N M Schumacher
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - John B A G Haanen
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
125
|
Hanakawa H, Orita Y, Sato Y, Takeuchi M, Ohno K, Gion Y, Tsukahara K, Tamamura R, Ito T, Nagatsuka H, Nishizaki K, Yoshino T. Regulatory T-cell infiltration in tongue squamous cell carcinoma. Acta Otolaryngol 2014; 134:859-64. [PMID: 24921153 DOI: 10.3109/00016489.2014.918279] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONCLUSION In tongue squamous cell carcinoma (SCC), high levels of regulatory T-cell (Treg) infiltration in tumor nests are observed in the cases with poor prognosis. OBJECTIVES The role of Tregs in head and neck cancers remains unclear. The aim of this study was to observe the distribution of Tregs in different stages of tongue SCC and estimate the effects on prognosis. METHODS Thirty-four cases with tongue SCC were examined immunohistochemically for CD4, CD8, and Forkhead box P3 (Foxp3). Immunoreactive cells were counted in cancer stroma and nest regions, and relationships between cell numbers and disease-free survival rates were analyzed. RESULTS In the 34 cases, univariate analysis for disease-free survival indicated high-level infiltration of Tregs (CD4(+)Foxp3+) into both cancer nests and stroma and presence of helper T (CD4(+)Foxp3-) cells in cancer stroma as potential predictors of significantly worse prognosis. In early-stage cases (stage I/II), high-level infiltration of Tregs in cancer nests correlated significantly with poor disease-free survival rate. Multivariate analysis for disease-free survival found no independent variables.
Collapse
|
126
|
Im KI, Park MJ, Kim N, Lim JY, Park HS, Lee SH, Nam YS, Lee ES, Lee JH, Cho ML, Cho SG. Induction of mixed chimerism using combinatory cell-based immune modulation with mesenchymal stem cells and regulatory T cells for solid-organ transplant tolerance. Stem Cells Dev 2014; 23:2364-76. [PMID: 24804993 DOI: 10.1089/scd.2013.0617] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Establishment of mixed chimerism is an ideal approach to induce donor-specific tolerance while expanding its potential in various clinical settings. Despite the developments in partial conditioning regimens, improvements are still needed in reducing toxicity and bone marrow transplantation-related complications. Recently, cell-based therapies, including mesenchymal stem cells (MSCs), have been incorporated in establishing noncytoreductive mixed chimerism protocols; however, its efficacy is only partial and shows reversed immunosuppressive properties. This study demonstrates a novel approach to induce mixed chimerism and tolerance through combinatory cell-based immune modulation (CCIM) of MSCs and regulatory T cells (Tregs). We hypothesize that the interaction between these cells may lead to greater inhibition of host immune responses. Compared with single cell therapy, CCIM induced a higher engraftment rate and robust donor-specific tolerance to skin allografts across full major histocompatibility complex barriers. These regulatory effects were associated with inhibition of natural killer cell cytotoxic activity, CD4(+)IL-17(+) cells, memory B cells, plasma cells, and immunoglobulin production levels along with increased frequencies of CD4(+)Foxp3(+) cells, IL-10-producing mature B cells, and myeloid-derived suppressor cells. Furthermore, CCIM was able to regulate mortality in a graft-versus-host disease model through reciprocal regulation of Treg/Th17. Taken together, we suggest CCIM as a clinically applicable strategy for facilitating the induction of mixed chimerism and permanent tolerance.
Collapse
Affiliation(s)
- Keon-Il Im
- 1 Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, The Catholic University of Korea College of Medicine , Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Whiteside TL. Induced regulatory T cells in inhibitory microenvironments created by cancer. Expert Opin Biol Ther 2014; 14:1411-25. [PMID: 24934899 DOI: 10.1517/14712598.2014.927432] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Regulatory T cells (Tregs) accumulating in the peripheral circulation and tumor sites of patients contribute to tumor escape from the host immune system. Tregs encompass subsets of immune cells with distinct phenotypic and functional properties. Whereas natural (n) or thymic-derived (t) Tregs regulate responses to self-antigens, inducible (i) or peripheral (p) Tregs generated and expanded in regulatory microenvironments control immune responses to a broad variety of antigens. AREAS COVERED Tregs accumulating in the tumor microenvironment (TME) are contextually regulated. They acquire phenotypic and functional attributes imposed by the inhibitory molecular pathways operating in situ. Several molecular pathways active in human cancer are reviewed. The pathways may differ from one tumor to another, and environmentally induced Tregs may be functionally distinct. Potential therapeutic strategies for selective silencing of iTregs are considered in the light of the newly acquired understanding of their phenotypic and functional diversity. EXPERT OPINION Human Tregs accumulating in cancer comprise 'bad' subsets, which inhibit antitumor immunity, and 'good' anti-inflammatory subsets, which maintain tolerance to self and benefit the host. Future therapeutic strategies targeting Tregs will need to discriminate between these Treg subsets and will need to consider reprogramming strategies instead of Treg elimination. Re-establishment of effective antitumor immune responses in cancer patients without disturbing a normal homeostatic T-cell balance will greatly benefit from insights into inhibitory pathways engaged by human tumors.
Collapse
Affiliation(s)
- Theresa L Whiteside
- University of Pittsburgh Cancer Institute , 5117 Centre Avenue, Pittsburgh, PA 15213 , USA +1 412 624 0096 ; +1 412 624 0264 ;
| |
Collapse
|
128
|
Regulatory T cells and the risk of CMV end-organ disease in patients with AIDS. J Acquir Immune Defic Syndr 2014; 66:25-32. [PMID: 24378728 DOI: 10.1097/qai.0000000000000095] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Cytomegalovirus (CMV)-specific T-cell effectors (CMV-Teff) protect against CMV end-organ disease (EOD). In HIV-infected individuals, their numbers and function vary with CD4 cell numbers and HIV load. The role of regulatory T cells (Treg) in CMV-EOD has not been extensively studied. We investigated the contribution of Treg and Teff toward CMV-EOD in HIV-infected individuals independently of CD4 cell numbers and HIV load and controlling for CMV reactivations. DESIGN We matched 43 CMV-EOD cases to 93 controls without CMV-EOD, but with similar CD4 cell numbers and HIV plasma RNA. CMV reactivation was investigated by blood DNA polymerase chain reaction over 32 weeks preceding the CMV-EOD in cases and preceding the matching point in controls. METHODS CMV-Teff and Treg were characterized by the expression of interferon-γ (IFN-γ), interleukin 2, tumor necrosis factor α (TNFα), MIP1β, granzyme B (GrB), CD107a, TNFα, FOXP3, and CD25. RESULTS Sixty-five percent cases and 20% controls had CMV reactivations. In multivariate analyses that controlled for CMV reactivations, none of the CMV-Teff subsets correlated with protection, but high CMV-GrB enzyme-linked immunosorbent spot responses and CMV-specific CD4FOXP3+%, CD4TNFα+%, and CD8CD107a% were significant predictors of CMV-EOD. CONCLUSIONS Because both FOXP3 and GrB have been previously associated with Treg activity, we conclude that CMV-Treg may play an important role in the development of CMV-EOD in advanced HIV disease. We were not able to identify a CMV-Teff subset that could be used as a surrogate of protection against CMV-EOD in this highly immunocompromised population.
Collapse
|
129
|
Pinto MT, Malta TM, Rodrigues ES, Pinheiro DG, Panepucci RA, Malmegrim de Farias KCR, Sousa ADP, Takayanagui OM, Tanaka Y, Covas DT, Kashima S. Genes related to antiviral activity, cell migration, and lysis are differentially expressed in CD4(+) T cells in human t cell leukemia virus type 1-associated myelopathy/tropical spastic paraparesis patients. AIDS Res Hum Retroviruses 2014; 30:610-22. [PMID: 24041428 DOI: 10.1089/aid.2013.0109] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human T cell leukemia virus type 1 (HTLV-1) preferentially infects CD4(+) T cells and these cells play a central role in HTLV-1 infection. In this study, we investigated the global gene expression profile of circulating CD4(+) T cells from the distinct clinical status of HTLV-1-infected individuals in regard to TAX expression levels. CD4(+) T cells were isolated from asymptomatic HTLV-1 carrier (HAC) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) patients in order to identify genes involved in HAM/TSP development using a microarray technique. Hierarchical clustering analysis showed that healthy control (CT) and HTLV-1-infected samples clustered separately. We also observed that the HAC and HAM/TSP groups clustered separately regardless of TAX expression. The gene expression profile of CD4(+) T cells was compared among the CT, HAC, and HAM/TSP groups. The paxillin (Pxn), chemokine (C-X-C motif ) receptor 4 (Cxcr4), interleukin 27 (IL27), and granzyme A (Gzma) genes were differentially expressed between the HAC and HAM/TSP groups, regardless of TAX expression. The perforin 1 (Prf1) and forkhead box P3 (Foxp3) genes were increased in the HAM/TSP group and presented a positive correlation to the expression of TAX and the proviral load (PVL). The frequency of CD4(+)FOXP3(+) regulatory T cells (Treg) was higher in HTLV-1-infected individuals. Foxp3 gene expression was positively correlated with cell lysis-related genes (Gzma, Gzmb, and Prf1). These findings suggest that CD4(+) T cell activity is distinct between the HAC and HAM/TSP groups.
Collapse
Affiliation(s)
- Mariana Tomazini Pinto
- National Institute of Science and Technology in Stem Cell and Cell Therapy, Center for Cell-Based Therapy and Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, Brazil
| | - Tathiane Maistro Malta
- National Institute of Science and Technology in Stem Cell and Cell Therapy, Center for Cell-Based Therapy and Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, Brazil
| | - Evandra Strazza Rodrigues
- National Institute of Science and Technology in Stem Cell and Cell Therapy, Center for Cell-Based Therapy and Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniel Guariz Pinheiro
- National Institute of Science and Technology in Stem Cell and Cell Therapy, Center for Cell-Based Therapy and Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Rodrigo Alexandre Panepucci
- National Institute of Science and Technology in Stem Cell and Cell Therapy, Center for Cell-Based Therapy and Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Kelen Cristina Ribeiro Malmegrim de Farias
- National Institute of Science and Technology in Stem Cell and Cell Therapy, Center for Cell-Based Therapy and Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, Brazil
| | - Alessandra De Paula Sousa
- National Institute of Science and Technology in Stem Cell and Cell Therapy, Center for Cell-Based Therapy and Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
| | | | - Yuetsu Tanaka
- Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Dimas Tadeu Covas
- National Institute of Science and Technology in Stem Cell and Cell Therapy, Center for Cell-Based Therapy and Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Simone Kashima
- National Institute of Science and Technology in Stem Cell and Cell Therapy, Center for Cell-Based Therapy and Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
130
|
Abstract
Regulatory T cells are the central element for the maintenance of peripheral tolerance. Several subtypes of regulatory T (Treg) cells have been described, and most of them belong to the CD4(+) T-helper (Th) cell lineage. These specific subtypes can be discriminated according to phenotype and function. Forkhead box protein 3 (FoxP3)-expressing natural Treg cells (Tregs) and IL-10-producing, T-regulatory type 1 cells (Tr1) are the best-studied types of CD4(+) regulatory T cells in humans and experimental animal models. It was shown that they play a crucial role during autoimmune neuroinflammation. Both cells types seem to be particularly important for multiple sclerosis (MS). Here, we discuss the role of CD4(+) regulatory T cells in autoimmune neuroinflammation with an emphasis on Tregs and Tr1 cells in MS.
Collapse
Affiliation(s)
- Markus Kleinewietfeld
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
- Faculty of Medicine, Dresden University of Technology (TUD), Dresden, Germany
| | - David A. Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
131
|
Scurr M, Ladell K, Besneux M, Christian A, Hockey T, Smart K, Bridgeman H, Hargest R, Phillips S, Davies M, Price D, Gallimore A, Godkin A. Highly prevalent colorectal cancer-infiltrating LAP⁺ Foxp3⁻ T cells exhibit more potent immunosuppressive activity than Foxp3⁺ regulatory T cells. Mucosal Immunol 2014; 7:428-39. [PMID: 24064667 PMCID: PMC3931584 DOI: 10.1038/mi.2013.62] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 06/13/2013] [Accepted: 07/09/2013] [Indexed: 02/04/2023]
Abstract
Although elevated CD4⁺Foxp3⁺ regulatory T cell (Treg) frequencies within tumors are well documented, the functional and phenotypic characteristics of CD4⁺Foxp3⁺ and CD4⁺Foxp3⁻ T cell subsets from matched blood, healthy colon, and colorectal cancer require in-depth investigation. Flow cytometry revealed that the majority of intratumoral CD4⁺Foxp3⁺ T cells (Tregs) were Helios⁺ and expressed higher levels of cytotoxic T-lymphocyte antigen 4 (CTLA-4) and CD39 than Tregs from colon and blood. Moreover, ∼30% of intratumoral CD4⁺Foxp3⁻ T cells expressed markers associated with regulatory functions, including latency-associated peptide (LAP), lymphocyte activation gene-3 (LAG-3), and CD25. This unique population of cells produced interleukin-10 (IL-10) and transforming growth factor-β (TGF-β), and was ∼50-fold more suppressive than Foxp3⁺ Tregs. Thus, intratumoral Tregs are diverse, posing multiple obstacles to immunotherapeutic intervention in colorectal malignancies.
Collapse
Affiliation(s)
- M Scurr
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - K Ladell
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - M Besneux
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - A Christian
- Department of Pathology, University Hospital of Wales, Cardiff, UK
| | - T Hockey
- Department of Pathology, University Hospital of Wales, Cardiff, UK
| | - K Smart
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - H Bridgeman
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - R Hargest
- Department of Surgery, University Hospital of Wales, Cardiff, UK
| | - S Phillips
- Department of Surgery, University Hospital of Wales, Cardiff, UK
| | - M Davies
- Department of Surgery, University Hospital of Wales, Cardiff, UK
| | - D Price
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - A Gallimore
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - A Godkin
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK,
| |
Collapse
|
132
|
Sanchez Rodriguez R, Pauli ML, Neuhaus IM, Yu SS, Arron ST, Harris HW, Yang SHY, Anthony BA, Sverdrup FM, Krow-Lucal E, MacKenzie TC, Johnson DS, Meyer EH, Löhr A, Hsu A, Koo J, Liao W, Gupta R, Debbaneh MG, Butler D, Huynh M, Levin EC, Leon A, Hoffman WY, McGrath MH, Alvarado MD, Ludwig CH, Truong HA, Maurano MM, Gratz IK, Abbas AK, Rosenblum MD. Memory regulatory T cells reside in human skin. J Clin Invest 2014; 124:1027-36. [PMID: 24509084 DOI: 10.1172/jci72932] [Citation(s) in RCA: 261] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 11/21/2013] [Indexed: 01/07/2023] Open
Abstract
Regulatory T cells (Tregs), which are characterized by expression of the transcription factor Foxp3, are a dynamic and heterogeneous population of cells that control immune responses and prevent autoimmunity. We recently identified a subset of Tregs in murine skin with properties typical of memory cells and defined this population as memory Tregs (mTregs). Due to the importance of these cells in regulating tissue inflammation in mice, we analyzed this cell population in humans and found that almost all Tregs in normal skin had an activated memory phenotype. Compared with mTregs in peripheral blood, cutaneous mTregs had unique cell surface marker expression and cytokine production. In normal human skin, mTregs preferentially localized to hair follicles and were more abundant in skin with high hair density. Sequence comparison of TCRs from conventional memory T helper cells and mTregs isolated from skin revealed little homology between the two cell populations, suggesting that they recognize different antigens. Under steady-state conditions, mTregs were nonmigratory and relatively unresponsive; however, in inflamed skin from psoriasis patients, mTregs expanded, were highly proliferative, and produced low levels of IL-17. Taken together, these results identify a subset of Tregs that stably resides in human skin and suggest that these cells are qualitatively defective in inflammatory skin disease.
Collapse
|
133
|
Abstract
Regulatory T (TReg) cells constitute an essential counterbalance to adaptive immune responses. Failure to maintain appropriate TReg cell numbers or function leads to autoimmune, malignant and immunodeficient conditions. Dynamic homeostatic processes preserve the number of forkhead box P3-expressing (FOXP3(+)) TReg cells within a healthy range, with high rates of cell division being offset by apoptosis under steady-state conditions. Recent studies have shown that TReg cells become specialized for different environmental contexts, tailoring their functions and homeostatic properties to a wide range of tissues and immune conditions. In this Review, we describe new insights into the molecular controls that maintain the steady-state homeostasis of TReg cells and the cues that drive TReg cell adaptation to inflammation and/or different locations. We highlight how differing local milieu might drive context-specific TReg cell function and restoration of immune homeostasis, and how dysregulation of these processes can precipitate disease.
Collapse
Affiliation(s)
- Adrian Liston
- 1] Autoimmune Genetics Laboratory, VIB, Leuven 3000, Belgium. [2] Department of Microbiology and Immunology, University of Leuven, Leuven 3000, Belgium
| | - Daniel H D Gray
- 1] The Walter and Eliza Hall Institute of Medical Research, Melbourne 3053, Australia. [2] Department of Medical Biology, University of Melbourne, Melbourne 3052, Australia
| |
Collapse
|
134
|
Farber DL, Yudanin NA, Restifo NP. Human memory T cells: generation, compartmentalization and homeostasis. Nat Rev Immunol 2014; 14:24-35. [PMID: 24336101 PMCID: PMC4032067 DOI: 10.1038/nri3567] [Citation(s) in RCA: 619] [Impact Index Per Article: 61.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Memory T cells constitute the most abundant lymphocyte population in the body for the majority of a person's lifetime; however, our understanding of memory T cell generation, function and maintenance mainly derives from mouse studies, which cannot recapitulate the exposure to multiple pathogens that occurs over many decades in humans. In this Review, we discuss studies focused on human memory T cells that reveal key properties of these cells, including subset heterogeneity and diverse tissue residence in multiple mucosal and lymphoid tissue sites. We also review how the function and the adaptability of human memory T cells depend on spatial and temporal compartmentalization.
Collapse
Affiliation(s)
- Donna L Farber
- 1] Columbia Center for Translational Immunology and Department of Microbiology and Immunology, Columbia University Medical Center, 650 West 168th Street, BB1501, New York, New York 10032, USA. [2] Department of Surgery, Columbia University Medical Center, 650 West 168th Street, BB1501, New York 10032, USA
| | - Naomi A Yudanin
- Columbia Center for Translational Immunology and Department of Microbiology and Immunology, Columbia University Medical Center, 650 West 168th Street, BB1501, New York, New York 10032, USA
| | - Nicholas P Restifo
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
135
|
Matarese G, Colamatteo A, De Rosa V. Metabolic fuelling of proper T cell functions. Immunol Lett 2013; 161:174-8. [PMID: 24365064 DOI: 10.1016/j.imlet.2013.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 12/13/2013] [Indexed: 11/29/2022]
Abstract
The interplay of the immune system with other aspects of physiology is one of the hottest topics of the recent literature. A crucial example is the influence of metabolic cues on immune responses. It is now well accepted that upon activation, T lymphocytes take on a metabolic profile profoundly distinct from that of their quiescent and anergic counterparts; in these sense, T cell metabolism is highly dynamic and has a serious impact on the ability of T cell to grow, activate and differentiate. Specific metabolic pathways provide energy and biosynthetic precursors able to support specific T cell functions, such as effector, regulatory and memory. Here, we review the main signaling pathways that control metabolism and how the metabolic phenotypes of T cell subtypes integrate with their specific function.
Collapse
Affiliation(s)
- Giuseppe Matarese
- Dipartimento di Medicina e Chirurgia, Università di Salerno, Baronissi Campus, Baronissi 84081, Salerno, Italy; IRCCS-MultiMedica, Milano 20138, Italy.
| | | | - Veronica De Rosa
- Unità di NeuroImmunologia, IRCCS-Fondazione Santa Lucia, Roma 00143, Italy; Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli 80131, Italy
| |
Collapse
|
136
|
Carbone F, De Rosa V, Carrieri PB, Montella S, Bruzzese D, Porcellini A, Procaccini C, La Cava A, Matarese G. Regulatory T cell proliferative potential is impaired in human autoimmune disease. Nat Med 2013; 20:69-74. [DOI: 10.1038/nm.3411] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 10/22/2013] [Indexed: 12/15/2022]
|
137
|
HIV-1 Vpr accelerates viral replication during acute infection by exploitation of proliferating CD4+ T cells in vivo. PLoS Pathog 2013; 9:e1003812. [PMID: 24339781 PMCID: PMC3855622 DOI: 10.1371/journal.ppat.1003812] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 10/22/2013] [Indexed: 11/28/2022] Open
Abstract
The precise role of viral protein R (Vpr), an HIV-1-encoded protein, during HIV-1 infection and its contribution to the development of AIDS remain unclear. Previous reports have shown that Vpr has the ability to cause G2 cell cycle arrest and apoptosis in HIV-1-infected cells in vitro. In addition, vpr is highly conserved in transmitted/founder HIV-1s and in all primate lentiviruses, which are evolutionarily related to HIV-1. Although these findings suggest an important role of Vpr in HIV-1 pathogenesis, its direct evidence in vivo has not been shown. Here, by using a human hematopoietic stem cell-transplanted humanized mouse model, we demonstrated that Vpr causes G2 cell cycle arrest and apoptosis predominantly in proliferating CCR5+ CD4+ T cells, which mainly consist of regulatory CD4+ T cells (Tregs), resulting in Treg depletion and enhanced virus production during acute infection. The Vpr-dependent enhancement of virus replication and Treg depletion is observed in CCR5-tropic but not CXCR4-tropic HIV-1-infected mice, suggesting that these effects are dependent on the coreceptor usage by HIV-1. Immune activation was observed in CCR5-tropic wild-type but not in vpr-deficient HIV-1-infected humanized mice. When humanized mice were treated with denileukin diftitox (DD), to deplete Tregs, DD-treated humanized mice showed massive activation/proliferation of memory T cells compared to the untreated group. This activation/proliferation enhanced CCR5 expression in memory CD4+ T cells and rendered them more susceptible to CCR5-tropic wild-type HIV-1 infection than to vpr-deficient virus. Taken together, these results suggest that Vpr takes advantage of proliferating CCR5+ CD4+ T cells for enhancing viremia of CCR5-tropic HIV-1. Because Tregs exist in a higher cycling state than other T cell subsets, Tregs appear to be more vulnerable to exploitation by Vpr during acute HIV-1 infection. HIV-1 encodes nine genes, five of which (gag, pol, env, tat, and rev) are essential for viral replication, and four, termed accessory genes (vif, vpu, nef, and vpr), appear to aid virus infection. Of the four accessory proteins, Vpr is the most enigmatic. It is well known that Vpr has the potential to cause G2 cell cycle arrest and apoptosis in vitro. Moreover, it has been reported that Vpr-mediated G2 arrest increases HIV-1 production in vitro. However, the role of Vpr in HIV-1 propagation in vivo remains unclear. Here, by using a humanized mouse model, we demonstrate that Vpr enhances CCR5-tropic but not CXCR4-tropic HIV-1 replication in vivo by exploiting Tregs during acute infection. In CCR5-tropic HIV-1-infected humanized mice, Vpr-dependent G2 cell cycle arrest and apoptosis are predominantly observed in infected Tregs, and wild-type but not vpr-deficient HIV-1-infected mice displayed acute Treg depletion. This Vpr-dependent Treg depletion may lead to immune activation and provide a pool of activated/proliferating CD4+ T cells, which supports subsequent HIV-1 expansion in vivo. This is the first report demonstrating the role of Vpr in HIV-1 infection in vivo.
Collapse
|
138
|
Abstract
In recent years, there have been many new developments in the field of regulatory T cells (Treg), challenging the consensus on their behaviour, classification and role(s) in disease. The role Treg might play in autoimmune disease appears to be more complex than previously thought. Here, we discuss the current knowledge of regulatory T cells through animal and human research and illustrate the recent developments in childhood autoimmune arthritis (juvenile idiopathic arthritis (JIA)). Furthermore, this review summarises our understanding of the fields and assesses current and future implications for Treg in the treatment of JIA.
Collapse
|
139
|
Xu G, Gong Y. Deregulation from CD4+ memory T cells to regulatory cells in patients with chronic renal failure: a pilot study. J Clin Lab Anal 2013; 27:423-6. [PMID: 24218122 DOI: 10.1002/jcla.21622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 03/28/2013] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The aim of this study was to elucidate whether the CD4+ memory T (Tm) cells differentiation to regulatory T cells (Tregs) play a role in the immunological defects in patients with chronic kidney disease (CKD), and if the oxidized low-density lipoprotein (oxLDL) had affect on on CD4+ Tm cells and Tregs apoptosis in these subjects. METHODS CD4+ Tm cells and Tregs were detected by flow cytometry in each group of ten subjects. Apoptosis was measured by flow cytometry and confirmed by Western blotting. RESULTS The oxLDL concentration was significantly higher in CKD stage 4 (CKD4) patients than in controls, particularly in hemodialysis (HD) subjects (P < 0.001, respectively). In total, 100 μg/ml oxLDL significantly inhibited the CD4+ Tm cell proliferation. oxLDL-induced Tm generated Tregs apoptosis in controls and CKD4 patients, especially in HD patients (P < 0.001, respectively). CONCLUSION Dysregulation of CD4+ Tm cells converting into Tregs played a role in the immune defects of CKD patients, and oxLDL induced the apoptosis of Tm generating Tregs in these subjects. Larger size of sample should be investigated to confirm the findings in further studies.
Collapse
Affiliation(s)
- Gaosi Xu
- Department of Nephrology, Second Affiliated Hospital, Nanchang University, Jiangxi, P. R. China; Department of Immunology, Shangrao Branch of Jiangxi Medical College, Shangrao, P. R. China
| | | |
Collapse
|
140
|
Zhang X, Chang Li X, Xiao X, Sun R, Tian Z, Wei H. CD4(+)CD62L(+) central memory T cells can be converted to Foxp3(+) T cells. PLoS One 2013; 8:e77322. [PMID: 24155942 PMCID: PMC3796486 DOI: 10.1371/journal.pone.0077322] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 09/09/2013] [Indexed: 12/21/2022] Open
Abstract
The peripheral Foxp3(+) Treg pool consists of naturally arising Treg (nTreg) and adaptive Treg cells (iTreg). It is well known that naive CD4(+) T cells can be readily converted to Foxp3(+) iTreg in vitro, and memory CD4(+) T cells are resistant to conversion. In this study, we investigated the induction of Foxp3(+) T cells from various CD4(+) T-cell subsets in human peripheral blood. Though naive CD4(+) T cells were readily converted to Foxp3(+) T cells with TGF-β and IL-2 treatment in vitro, such Foxp3(+) T cells did not express the memory marker CD45RO as do Foxp3(+) T cells induced in the peripheral blood of Hepatitis B Virus (HBV) patients. Interestingly, a subset of human memory CD4(+) T cells, defined as CD62L(+) central memory T cells, could be induced by TGF-β to differentiate into Foxp3(+) T cells. It is well known that Foxp3(+) T cells derived from human CD4(+)CD25(-) T cells in vitro are lack suppressive functions. Our data about the suppressive functions of CD4(+)CD62L(+) central memory T cell-derived Foxp3(+) T cells support this conception, and an epigenetic analysis of these cells showed a similar methylation pattern in the FOXP3 Treg-specific demethylated region as the naive CD4(+) T cell-derived Foxp3(+) T cells. But further research showed that mouse CD4(+) central memory T cells also could be induced to differentiate into Foxp3(+) T cells, such Foxp3(+) T cells could suppress the proliferation of effector T cells. Thus, our study identified CD4(+)CD62L(+) central memory T cells as a novel potential source of iTreg.
Collapse
Affiliation(s)
- Xiaolong Zhang
- Institute of Immunology, School of Life Sciences, University of Science & Technology of China, Hefei, Anhui, China
| | - Xian Chang Li
- Transplant Research Center, Brigham and Women's Hospital & Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Xiang Xiao
- Transplant Research Center, Brigham and Women's Hospital & Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rui Sun
- Institute of Immunology, School of Life Sciences, University of Science & Technology of China, Hefei, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhigang Tian
- Institute of Immunology, School of Life Sciences, University of Science & Technology of China, Hefei, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
- * E-mail: (HW); (ZT)
| | - Haiming Wei
- Institute of Immunology, School of Life Sciences, University of Science & Technology of China, Hefei, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
- * E-mail: (HW); (ZT)
| |
Collapse
|
141
|
Selective depletion of FOXP3(high) cells by Fas-Fas-L-induced apoptosis occurs in CD4(+)CD25(+)-enriched populations during repeated expansion. Cytotherapy 2013; 15:1286-96. [PMID: 23993302 DOI: 10.1016/j.jcyt.2013.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/20/2013] [Accepted: 05/28/2013] [Indexed: 11/23/2022]
Abstract
BACKGROUND AIMS Expansion of anti-CD25 bead-isolated human Tregs culture has paradoxically resulted in reduced suppressive activity, but the mechanism(s) responsible for these observations are poorly defined. METHODS Magnetic-bead isolated human CD25(+) cells were expanded with anti-CD3/CD28 beads and high doses of rhIL-2. Detection of Fas and Fas ligand (Fas-L) expression, activation of Caspase 8, cell proliferation and cytokine production was evaluated by multi-color fluorescence-activated cell sorting analysis. The role of Fas-Fas-L-mediated cell death was dissected through the use of agonist or antagonist monoclonal antibodies directed at Fas and Fas-L. RESULTS Repeated expansion of bead-enriched CD4(+)CD25(+) cells generated a cellular product with markedly reduced suppressive activity and with significantly increased CD8(+) T cells and CD4(+) T cells producing interferon-γ and/or interleukin-2. We showed that Fas-Fas-L-mediated apoptosis of CD4(+)FOXP3(high) cells and rapid cell-cycling of CD8(+) T cells were collectively responsible for the reduced proportion of CD4(+)FOXP3(high) cells in expanded cultures. The depletion of CD4(+)FOXP3(high) cells and activation of Caspase 8 in CD4(+)FOXP3(high) cells was attenuated by Fas antagonist antibody, ZB4, in short-term culture. However, the loss of CD4(+)FOXP3(high) cells during expansion was not prevented by either Fas or Fas-L antagonist antibodies. CONCLUSIONS Taken together, the data show that Fas-Fas-L-mediated apoptosis may limit the expansion of anti-CD25 bead-isolated cells in vitro.
Collapse
|
142
|
p53 regulates mesenchymal stem cell-mediated tumor suppression in a tumor microenvironment through immune modulation. Oncogene 2013; 33:3830-8. [PMID: 23975435 DOI: 10.1038/onc.2013.355] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 07/13/2013] [Accepted: 07/24/2013] [Indexed: 01/14/2023]
Abstract
p53 is one of the most studied genes in cancer biology, and mutations in this gene may be predictive for the development of many types of cancer in humans and in animals. However, whether p53 mutations in non-tumor stromal cells can affect tumor development has received very little attention. In this study, we show that B16F0 melanoma cells form much larger tumors in p53-deficient mice than in wild-type mice, indicating a potential role of p53 deficiency in non-tumor cells of the microenvironment. As mesenchymal stem cells (MSCs) are attracted to tumors and form a major component of the tumor microenvironment, we examined the potential role of p53 status in MSCs in tumor development. We found that larger tumors resulted when B16F0 melanoma cells were co-injected with bone marrow MSCs derived from p53-deficient mice rather than MSCs from wild-type mice. Interestingly, this tumor-promoting effect by p53-deficient MSCs was not observed in non-obese diabetic/severe combined immunodeficiency mice, indicating the immune response has a critical role. Indeed, in the presence of inflammatory cytokines, p53-deficient MSCs expressed more inducible nitric oxide synthase (iNOS) and exhibited greater immunosuppressive capacity. Importantly, tumor promotion by p53-deficient MSCs was abolished by administration of S-methylisothiourea, an iNOS inhibitor. Therefore, our data demonstrate that p53 status in tumor stromal cells has a key role in tumor development by modulating immune responses.
Collapse
|
143
|
Fox M, Knapp LA, Andrews PW, Fincher CL. Hygiene and the world distribution of Alzheimer's disease: Epidemiological evidence for a relationship between microbial environment and age-adjusted disease burden. EVOLUTION MEDICINE AND PUBLIC HEALTH 2013; 2013:173-86. [PMID: 24481197 PMCID: PMC3868447 DOI: 10.1093/emph/eot015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
People living in sanitized environments may be at greater Alzheimer's risk. We compare Alzheimer's rates in different countries in light of countries' historical and contemporary pathogen prevalence, sanitation, and urbanization. We find that countries that are less urbanized, with more pathogens and lower degree of sanitation have lower Alzheimer's rates. Background and objectives: Alzheimer’s disease (AD) shares certain etiological features with autoimmunity. Prevalence of autoimmunity varies between populations in accordance with variation in environmental microbial diversity. Exposure to microorganisms may improve individuals’ immunoregulation in ways that protect against autoimmunity, and we suggest that this may also be the case for AD. Here, we investigate whether differences in microbial diversity can explain patterns of age-adjusted AD rates between countries. Methodology: We use regression models to test whether pathogen prevalence, as a proxy for microbial diversity, across 192 countries can explain a significant amount of the variation in age-standardized AD disability-adjusted life-year (DALY) rates. We also review and assess the relationship between pathogen prevalence and AD rates in different world populations. Results: Based on our analyses, it appears that hygiene is positively associated with AD risk. Countries with greater degree of sanitation and lower degree of pathogen prevalence have higher age-adjusted AD DALY rates. Countries with greater degree of urbanization and wealth exhibit higher age-adjusted AD DALY rates. Conclusions and implications: Variation in hygiene may partly explain global patterns in AD rates. Microorganism exposure may be inversely related to AD risk. These results may help predict AD burden in developing countries where microbial diversity is rapidly diminishing. Epidemiological forecasting is important for preparing for future healthcare needs and research prioritization.
Collapse
Affiliation(s)
- Molly Fox
- Division of Biological Anthropology, Department of Anthropology and Archaeology, University of Cambridge, Pembroke Street, Cambridge CB2 3QY, UK, Department of Anthropology, University of Utah, 270 S 1400 E, Salt Lake City, UT 84112, USA, Department of Psychology, Neuroscience & Behaviour, McMaster University, 1280 Main Street W, Hamilton, ON L8S 4K1, Canada and Institute of Neuroscience and Psychology, University of Glasgow, 58 Hillhead Street, Glasgow G12 8QB, UK
| | | | | | | |
Collapse
|
144
|
Simonetta F, Bourgeois C. CD4+FOXP3+ Regulatory T-Cell Subsets in Human Immunodeficiency Virus Infection. Front Immunol 2013; 4:215. [PMID: 23908654 PMCID: PMC3727053 DOI: 10.3389/fimmu.2013.00215] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 07/13/2013] [Indexed: 01/29/2023] Open
Abstract
The role of CD4+FOXP3+ regulatory T cells (Treg) in human immunodeficiency virus (HIV) infection has been an area of intensive investigation and remains a matter of ardent debate. Investigation and interpretation suffered from uncertainties concerning Treg quantification. Firstly, Treg quantification and function in HIV infection remain controversial in part because of the lack of reliable and specific markers to identify human Tregs. Secondly, analyzing Treg percentages or absolute numbers led to apparent discrepancies that are now solved: it is now commonly accepted that Treg are targets of HIV infection, but are preferentially preserved compared to conventional CD4 T cells. Moreover, the duality of immune defects associated to HIV infection, i.e., low grade chronic inflammation and defects in HIV-specific responses also casts doubts on the potential impact of Treg on HIV infection. Tregs may be beneficial or/and detrimental to the control of HIV infection by suppressing chronic inflammation or HIV-specific responses respectively. Indeed both effects of Treg suppression have been described in HIV infection. The discovery in recent years of the existence of phenotypically and functionally distinct human CD4+FOXP3+ Treg subsets may provide a unique opportunity to reconcile these contrasting results. It is tempting to speculate that different Treg subsets exert these different suppressive effects. This review summarizes available data concerning Treg fate during HIV infection when considering Treg globally or as subsets. We discuss how the identification of naïve and effector Treg subsets modulates our understanding of Treg biology during HIV infection and the potential impact of HIV infection on mechanisms governing peripheral differentiation of adaptive Tregs.
Collapse
Affiliation(s)
- Federico Simonetta
- INSERM, U1012 , Le Kremlin-Bicêtre , France ; Université Paris-SUD, UMR-S1012 , Le Kremlin-Bicêtre , France ; Division of Immunology and Allergy, Department of Internal Medicine, Geneva University Hospitals , Geneva , Switzerland
| | | |
Collapse
|
145
|
Sjåheim TB, Bjørtuft Ø, Drabløs PA, Kongerud J, Halstensen TS. Increased bronchial density of CD25+Foxp3+ regulatory T cells in occupational asthma: relationship to current smoking. Scand J Immunol 2013; 77:398-404. [PMID: 23421612 DOI: 10.1111/sji.12035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 01/21/2013] [Indexed: 11/30/2022]
Abstract
To identify activated T cell subset in the asthmatic bronchia, we developed a triple-colour immunohistofluorescence labelling technique on cryo-section to discriminate activated CD4+CD25+ T cells, (effector T cells) from Foxp3+ regulatory T cells (Treg). Additional coexpression of activation and proliferation markers was also examined in situ. Bronchial biopsies were taken from 20 aluminium potroom workers (12 smokers) with asthma (>12% reversibility), 15 non-asthmatic potroom workers (7 smokers) and 10 non-smoking, non-exposed controls. Non-smoking asthmatics had significantly higher subepithelial density of both Tregs, effector T cells, activated (HLA-DR+) CD8+ and activated CD4+ T cells. Moreover, both Tregs, effector T cells and CD8+ T cells proliferated in the non-smoking asthmatics, only. Although smoking asthmatics had no asthma-associated increase in bronchial T cell, both had a significantly increase in effector T cell to Treg ratios. The significantly increased bronchial density of Tregs, effector T cells, proliferative T cells and activated CD8+ T cells in non-smoking asthmatics clearly showed that both the effector T cells and the inhibitory Treg system were activated in asthma.
Collapse
Affiliation(s)
- T B Sjåheim
- Laboratory for mucosal immunology LMI, IOB, University of Oslo, 0316 Oslo, Norway
| | | | | | | | | |
Collapse
|
146
|
Bollyky JB, Long SA, Fitch M, Bollyky PL, Rieck M, Rogers R, Samuels PL, Sanda S, Buckner JH, Hellerstein MK, Greenbaum CJ. Evaluation of in vivo T cell kinetics: use of heavy isotope labelling in type 1 diabetes. Clin Exp Immunol 2013; 172:363-74. [PMID: 23600824 DOI: 10.1111/cei.12064] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2013] [Indexed: 01/27/2023] Open
Abstract
CD4(+) memory cell development is dependent upon T cell receptor (TCR) signal strength, antigen dose and the cytokine milieu, all of which are altered in type 1 diabetes (T1D). We hypothesized that CD4(+) T cell turnover would be greater in type 1 diabetes subjects compared to controls. In vitro studies of T cell function are unable to evaluate dynamic aspects of immune cell homoeostasis. Therefore, we used deuterium oxide ((2) H(2)O) to assess in vivo turnover of CD4(+) T cell subsets in T1D (n = 10) and control subjects (n = 10). Serial samples of naive, memory and regulatory (T(reg)) CD4(+) T cell subsets were collected and enrichment of deoxyribose was determined by gas chromatography-mass spectrometry (GC-MS). Quantification of T cell turnover was performed using mathematical models to estimate fractional enrichment (f, n = 20), turnover rate (k, n = 20), proliferation (p, n = 10) and disappearance (d*, n = 10). Although turnover of T(regs) was greater than memory and naive cells in both controls and T1D subjects, no differences were seen between T1D and controls in T(reg) or naive kinetics. However, turnover of CD4(+) memory T cells was faster in those with T1D compared to control subjects. Measurement and modelling of incorporated deuterium is useful for evaluating the in vivo kinetics of immune cells in T1D and could be incorporated into studies of the natural history of disease or clinical trials designed to alter the disease course. The enhanced CD4(+) memory T cell turnover in T1D may be important in understanding the pathophysiology and potential treatments of autoimmune diabetes.
Collapse
Affiliation(s)
- J B Bollyky
- Benaroya Research Institute, Seattle, WA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Kallapur SG, Presicce P, Senthamaraikannan P, Alvarez M, Tarantal AF, Miller LM, Jobe AH, Chougnet CA. Intra-amniotic IL-1β induces fetal inflammation in rhesus monkeys and alters the regulatory T cell/IL-17 balance. THE JOURNAL OF IMMUNOLOGY 2013; 191:1102-9. [PMID: 23794628 DOI: 10.4049/jimmunol.1300270] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Very low birth weight preterm newborns are susceptible to the development of debilitating inflammatory diseases, many of which are associated with chorioamnionitis. To define the effects of chorioamnionitis on the fetal immune system, IL-1β was administered intra-amniotically at ~80% gestation in rhesus monkeys. IL-1β caused histological chorioamnionitis, as well as lung inflammation (infiltration of neutrophils or monocytes in the fetal airways). There were large increases in multiple proinflammatory cytokine mRNAs in the lungs at 24 h postadministration, which remained elevated relative to controls at 72 h. Intra-amniotic IL-1β also induced the sustained expression of the surfactant proteins in the lungs. Importantly, IL-1β significantly altered the balance between inflammatory and regulatory T cells. Twenty-four hours after IL-1β injection, the frequency of CD3(+)CD4(+)FOXP3(+) T cells was decreased in lymphoid organs. In contrast, IL-17A-producing cells (CD3(+)CD4(+), CD3(+)CD4(-), and CD3(-)CD4(-) subsets) were increased in lymphoid organs. The frequency of IFN-γ-expressing cells did not change. In this model of a single exposure to an inflammatory trigger, CD3(+)CD4(+)FOXP3(+) cells rebounded quickly, and their frequency was increased at 72 h compared with controls. IL-17 expression was also transient. Interestingly, the T cell profile alteration was confined to the lymphoid organs and not to circulating fetal T cells. Together, these results suggest that the chorioamnionitis-induced IL-1/IL-17 axis is involved in the severe inflammation that can develop in preterm newborns. Boosting regulatory T cells and/or controlling IL-17 may provide a means to ameliorate these abnormalities.
Collapse
Affiliation(s)
- Suhas G Kallapur
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | | | |
Collapse
|
148
|
Borg M, Brincat S, Camilleri G, Schembri-Wismayer P, Brincat M, Calleja-Agius J. The role of cytokines in skin aging. Climacteric 2013; 16:514-21. [PMID: 23659624 DOI: 10.3109/13697137.2013.802303] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cutaneous aging is one of the major noticeable menopausal complications that most women want to fight in their quest for an eternally youthful skin appearance. It may contribute to some maladies that occur in aging which, despite not being life-threatening, affect the well-being, psychological state and quality of life of aged women. Skin aging is mainly affected by three factors: chronological aging, decreased levels of estrogen after menopause, and environmental factors. Aged skin is characterized by a decrease in collagen content and skin thickness which result in dry, wrinkled skin that is easily bruised and takes a longer time to heal. Cytokines play a crucial role in the manifestation of these features of old skin. The pro-inflammatory cytokine tumor necrosis factor-alpha inhibits collagen synthesis and enhances collagen degradation by increasing the production of MMP-9. It also lowers the skin immunity and thus increases the risk of cutaneous infections in old age. Deranged levels of several interleukins and interferons also affect the aging process. The high level of CCN1 protein in aged skin gives dermal fibroblasts an 'age-associated secretory phenotype' that causes abnormal homeostasis of skin collagen and leads to the loss of the function and integrity of skin. Further research is required especially to establish the role of cytokines in the treatment of cutaneous aging.
Collapse
Affiliation(s)
- M Borg
- * Department of Anatomy, Faculty of Medicine and Surgery, University of Malta
| | | | | | | | | | | |
Collapse
|
149
|
Jenks JA, Seki S, Kanai T, Huang J, Morgan AA, Scalco RC, Nath R, Bucayu R, Wit JM, Al-Herz W, Ramadan D, Jorge AA, Bacchetta R, Hwa V, Rosenfeld R, Nadeau KC. Differentiating the roles of STAT5B and STAT5A in human CD4+ T cells. Clin Immunol 2013; 148:227-36. [PMID: 23773921 DOI: 10.1016/j.clim.2013.04.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 04/14/2013] [Accepted: 04/24/2013] [Indexed: 01/06/2023]
Abstract
STAT5A and STAT5B are highly homologous proteins whose distinctive roles in human immunity remain unclear. However, STAT5A sufficiency cannot compensate for STAT5B defects, and human STAT5B deficiency, a rare autosomal recessive primary immunodeficiency, is characterized by chronic lung disease, growth failure and autoimmunity associated with regulatory T cell (Treg) reduction. We therefore hypothesized that STAT5A and STAT5B play unique roles in CD4(+) T cells. Upon knocking down STAT5A or STAT5B in human primary T cells, we found differentially regulated expression of FOXP3 and IL-2R in STAT5B knockdown T cells and down-regulated Bcl-X only in STAT5A knockdown T cells. Functional ex vivo studies in homozygous STAT5B-deficient patients showed reduced FOXP3 expression with impaired regulatory function of STAT5B-null Treg cells, also of increased memory phenotype. These results indicate that STAT5B and STAT5A act partly as non-redundant transcription factors and that STAT5B is more critical for Treg maintenance and function in humans.
Collapse
Affiliation(s)
- Jennifer A Jenks
- Division of Immunology and Allergy, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Martinez-Pasamar S, Abad E, Moreno B, Velez de Mendizabal N, Martinez-Forero I, Garcia-Ojalvo J, Villoslada P. Dynamic cross-regulation of antigen-specific effector and regulatory T cell subpopulations and microglia in brain autoimmunity. BMC SYSTEMS BIOLOGY 2013; 7:34. [PMID: 23618467 PMCID: PMC3651362 DOI: 10.1186/1752-0509-7-34] [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: 11/23/2012] [Accepted: 04/23/2013] [Indexed: 12/28/2022]
Abstract
Background Multiple Sclerosis (MS) is considered a T-cell-mediated autoimmune disease with a prototypical oscillatory behavior, as evidenced by the presence of clinical relapses. Understanding the dynamics of immune cells governing the course of MS, therefore, has many implications for immunotherapy. Here, we used flow cytometry to analyze the time-dependent behavior of antigen-specific effector (Teff) and regulatory (Treg) T cells and microglia in mice model of MS, Experimental Autoimmune Encephalomyelitis (EAE), and compared the observations with a mathematical cross-regulation model of T-cell dynamics in autoimmune disease. Results We found that Teff and Treg cells specific to myelin olygodendrocyte glycoprotein (MOG) developed coupled oscillatory dynamics with a 4- to 5-day period and decreasing amplitude that was always higher for the Teff populations, in agreement with the mathematical model. Microglia activation followed the oscillations of MOG-specific Teff cells in the secondary lymphoid organs, but they were activated before MOG-specific T-cell peaks in the CNS. Finally, we assessed the role of B-cell depletion induced by anti-CD20 therapy in the dynamics of T cells in an EAE model with more severe disease after therapy. We observed that B-cell depletion decreases Teff expansion, although its oscillatory behavior persists. However, the effect of B cell depletion was more significant in the Treg population within the CNS, which matched with activation of microglia and worsening of the disease. Mathematical modeling of T-cell cross-regulation after anti-CD20 therapy suggests that B-cell depletion may influence the dynamics of T cells by fine-tuning their activation. Conclusions The oscillatory dynamics of T-cells have an intrinsic origin in the physiological regulation of the adaptive immune response, which influences both disease phenotype and response to immunotherapy.
Collapse
Affiliation(s)
- Sara Martinez-Pasamar
- Center of Neuroimmunology, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), Hospital Clinic of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|