1
|
Martínez-Vila C, González-Navarro EA, Teixido C, Martin R, Aya F, Juan M, Arance A. Lymphocyte T Subsets and Outcome of Immune Checkpoint Inhibitors in Melanoma Patients: An Oncologist's Perspective on Current Knowledge. Int J Mol Sci 2024; 25:9506. [PMID: 39273452 PMCID: PMC11394732 DOI: 10.3390/ijms25179506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/09/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Melanoma is the most aggressive and deadly form of skin cancer, and its incidence has been steadily increasing over the past few decades, particularly in the Caucasian population. Immune checkpoint inhibitors (ICI), anti-PD-1 monotherapy or in combination with anti-CTLA-4, and more recently, anti-PD-1 plus anti-LAG-3 have changed the clinical evolution of this disease. However, a significant percentage of patients do not benefit from these therapies. Therefore, to improve patient selection, it is imperative to look for novel biomarkers. Immune subsets, particularly the quantification of lymphocyte T populations, could contribute to the identification of ICI responders. The main purpose of this review is to thoroughly examine significant published data on the potential role of lymphocyte T subset distribution in peripheral blood (PB) or intratumorally as prognostic and predictive of response biomarkers in advanced melanoma patients treated with ICI regardless of BRAFV600 mutational status.
Collapse
Affiliation(s)
- Clara Martínez-Vila
- Department of Medical Oncology, Althaia Xarxa Assistencial Universitària de Manresa, Dr. Joan Soler, 1-3, 08243 Manresa, Spain
- Programa de Doctorat en Medicina i Recerca Translacional, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
- Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), Roda 70, 08500 Vic, Spain
| | - Europa Azucena González-Navarro
- Department of Immunology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
| | - Cristina Teixido
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
- Department of Pathology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
| | - Roberto Martin
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- Grupo Español de Terapias Inmunobiológicas en Cáncer (GETICA), Velázquez 7, 28001 Madrid, Spain
| | - Francisco Aya
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- Grupo Español de Terapias Inmunobiológicas en Cáncer (GETICA), Velázquez 7, 28001 Madrid, Spain
| | - Manel Juan
- Department of Immunology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
- Grupo Español de Terapias Inmunobiológicas en Cáncer (GETICA), Velázquez 7, 28001 Madrid, Spain
| | - Ana Arance
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- Grupo Español de Terapias Inmunobiológicas en Cáncer (GETICA), Velázquez 7, 28001 Madrid, Spain
| |
Collapse
|
2
|
Tripathi S, Tsang JS, Park K. Systems immunology of regulatory T cells: can one circuit explain it all? Trends Immunol 2023; 44:766-781. [PMID: 37690962 PMCID: PMC10543564 DOI: 10.1016/j.it.2023.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 09/12/2023]
Abstract
Regulatory T (Treg) cells play vital roles in immune homeostasis and response, including discrimination between self- and non-self-antigens, containment of immunopathology, and inflammation resolution. These diverse functions are orchestrated by cellular circuits involving Tregs and other cell types across space and time. Despite dramatic progress in our understanding of Treg biology, a quantitative framework capturing how Treg-containing circuits give rise to these diverse functions is lacking. Here, we propose that different facets of Treg function can be interpreted as distinct operating regimes of the same underlying circuit. We discuss how a systems immunology approach, involving quantitative experiments, computational modeling, and machine learning, can advance our understanding of Treg function, and help identify general operating and design principles underlying immune regulation.
Collapse
Affiliation(s)
- Shubham Tripathi
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA.
| | - John S Tsang
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA; Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA.
| | - Kyemyung Park
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA; Graduate School of Health Science and Technology and Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea.
| |
Collapse
|
3
|
Hall BM, Verma ND, Tran GT, Hodgkinson SJ. Transplant Tolerance, Not Only Clonal Deletion. Front Immunol 2022; 13:810798. [PMID: 35529847 PMCID: PMC9069565 DOI: 10.3389/fimmu.2022.810798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
The quest to understand how allogeneic transplanted tissue is not rejected and how tolerance is induced led to fundamental concepts in immunology. First, we review the research that led to the Clonal Deletion theory in the late 1950s that has since dominated the field of immunology and transplantation. At that time many basic mechanisms of immune response were unknown, including the role of lymphocytes and T cells in rejection. These original observations are reassessed by considering T regulatory cells that are produced by thymus of neonates to prevent autoimmunity. Second, we review "operational tolerance" induced in adult rodents and larger animals such as pigs. This can occur spontaneously especially with liver allografts, but also can develop after short courses of a variety of rejection inhibiting therapies. Over time these animals develop alloantigen specific tolerance to the graft but retain the capacity to reject third-party grafts. These animals have a "split tolerance" as peripheral lymphocytes from these animals respond to donor alloantigen in graft versus host assays and in mixed lymphocyte cultures, indicating there is no clonal deletion. Investigation of this phenomenon excludes many mechanisms, including anti-donor antibody blocking rejection as well as anti-idiotypic responses mediated by antibody or T cells. This split tolerance is transferred to a second immune-depleted host by T cells that retain the capacity to effect rejection of third-party grafts by the same host. Third, we review research on alloantigen specific inhibitory T cells that led to the first identification of the CD4+CD25+T regulatory cell. The key role of T cell derived cytokines, other than IL-2, in promoting survival and expansion of antigen specific T regulatory cells that mediate transplant tolerance is reviewed. The precise methods for inducing and diagnosing operational tolerance remain to be defined, but antigen specific T regulatory cells are key mediators.
Collapse
Affiliation(s)
- Bruce M. Hall
- Immune Tolerance Laboratory, School of Medicine, University of New South Wales (UNSW) Sydney, Ingham Institute, and Renal Service and Multiple Sclerosis Clinic, Liverpool Hospital, Liverpool, NSW, Australia
| | | | | | | |
Collapse
|
4
|
Scott DW. Driving CARs to BARs: The Winding Road to Specific Regulatory T Cells for Tolerance. Front Immunol 2021; 12:742719. [PMID: 34552599 PMCID: PMC8450509 DOI: 10.3389/fimmu.2021.742719] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/16/2021] [Indexed: 02/05/2023] Open
Abstract
Chimeric antigen receptor (CAR) transduced T cells have significantly improved cancer immunotherapy. Similarly, engineering regulatory T cells (Treg) with specific receptors to endow specificity and increase efficacy of Tregs holds great promise for therapy of a variety of adverse immune responses. In this review, we focus on our approaches using retroviral transduction of specific T-cell receptors, single chain variable fragments (scFv) or antigen in models of monogenic diseases, autoimmunity and allergy. The advantages of each of these for different targets diseases are discussed as well as their potential for clinical translation.
Collapse
Affiliation(s)
- David W Scott
- Department of Medicine (MED), Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
5
|
Verma D, Chan ED, Ordway DJ. The double-edged sword of Tregs in M tuberculosis, M avium, and M absessus infection. Immunol Rev 2021; 301:48-61. [PMID: 33713043 DOI: 10.1111/imr.12959] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
Immunity against different Mycobacteria species targeting the lung requires distinctly different pulmonary immune responses for bacterial clearance. Many parameters of acquired and regulatory immune responses differ quantitatively and qualitatively from immunity during infection with Mycobacteria species. Nontuberculosis Mycobacteria species (NTM) Mycobacterium avium- (M avium), Mycobacterium abscessus-(M abscessus), and the Mycobacteria species Mycobacterium tuberculosis-(Mtb). Herein, we discuss the potential implications of acquired and regulatory immune responses in the context of animal and human studies, as well as future directions for efforts to treat Mycobacteria diseases.
Collapse
Affiliation(s)
- Deepshikha Verma
- Mycobacteria Research Laboratory, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Edward D Chan
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Denver, CO, USA.,Departments of Medicine and Academic Affairs, National Jewish Health, Denver, CO, USA.,Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Diane J Ordway
- Mycobacteria Research Laboratory, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
6
|
Ramsdell F, Rudensky AY. Foxp3: a genetic foundation for regulatory T cell differentiation and function. Nat Immunol 2020; 21:708-709. [PMID: 32577010 DOI: 10.1038/s41590-020-0694-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
MESH Headings
- Allergy and Immunology/history
- Animals
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Diabetes Mellitus, Type 1/congenital
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diarrhea/genetics
- Diarrhea/immunology
- Disease Models, Animal
- Female
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Gene Expression Regulation/immunology
- Genetic Diseases, X-Linked/genetics
- Genetic Diseases, X-Linked/immunology
- History, 20th Century
- History, 21st Century
- Humans
- Immune System Diseases/congenital
- Immune System Diseases/genetics
- Immune System Diseases/immunology
- Male
- Mice, Transgenic
- Mutation
- Sex Factors
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Fred Ramsdell
- Sonoma Biotherapeutics, South San Francisco, CA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| | - Alexander Y Rudensky
- Howard Hughes Medical Institute and Immunology Program, Sloan Kettering Institute and Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
7
|
Del Río-Araiza VH, Palacios-Arreola MI, Nava-Castro KE, Pérez-Sánchez NY, Ruíz-Manzano R, Segovia-Mendoza M, Girón-Pérez MI, Navidad-Murrieta MS, Morales-Montor J. Perinatal exposure to bisphenol A increases in the adulthood of the offspring the susceptibility to the human parasite Toxocara canis. ENVIRONMENTAL RESEARCH 2020; 184:109381. [PMID: 32199324 DOI: 10.1016/j.envres.2020.109381] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 02/26/2020] [Accepted: 03/10/2020] [Indexed: 06/10/2023]
Abstract
Bisphenol A, a very widespread environmental pollutant and endocrine disruptor compound, can interact with several steroid receptors, particularly with estrogen ones. In different studies, it has observed that the endocrine disruption during critical periods of development can trigger alterations in the immune response during the adult life. Male Wistar rats were exposed indirectly to BPA at a dose of 250 μg/kg day during the perinatal period (from day 5 of pregnancy until day 21 postnatal), At the 60 days of age, the adulthood, animals were infected with larvated eggs of the Toxocara canis, and were sacrificed at 7 days post-infection. Parasitic loads in the lung and in the liver were analyzed by artificial digestion. Furthermore, immune cell subpopulations (macrophages, NK cells, Tγδ, total T cells, T helper, T cytotoxic, and B lymphocytes) present in spleen, peripheral and mesenteric lymph nodes were analyzed by flow cytometry. The expression of Th1 and Th2 cytokines at the splenic level was determined by real-time quantitative PCR. Finally, the titers of specific antibodies against to the parasite were analyzed by ELISA. The BPA treatment administrated in the perinatally stage favors a significant increase of the percentage of Toxocara canis larvae in the lungs and liver in the adulthood. Additionally, the exposure to this compound caused a dramatically decrease in the production of specific antibodies against to this parasite, downregulating together Th2 cytokines (IL-4, IL-5 and IL-13), meanwhile upregulated Th1 cytokines (IFN-γ and TNF-α). Perinatal exposure to BPA affects the performance of the immune response during adult life, modifying both cytokines and antibodies production by these cells, which favors the susceptibility to infections, specifically toxocariosis.
Collapse
Affiliation(s)
- Víctor H Del Río-Araiza
- Departamento de Parasitología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, CP 04510, Ciudad de México, Mexico
| | - Margarita I Palacios-Arreola
- Departamento de Ciencias Ambientales, Laboratorio de Genotoxicología y Mutagénesis Ambientales, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, CP 04510, Ciudad de México, Mexico
| | - Karen E Nava-Castro
- Departamento de Ciencias Ambientales, Laboratorio de Genotoxicología y Mutagénesis Ambientales, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, CP 04510, Ciudad de México, Mexico
| | - Nashla Y Pérez-Sánchez
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CP 04510, Ciudad de México, Mexico
| | - Rocío Ruíz-Manzano
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CP 04510, Ciudad de México, Mexico
| | - Mariana Segovia-Mendoza
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CP 04510, Ciudad de México, Mexico
| | - Manuel Iván Girón-Pérez
- Universidad Autónoma de Nayarit, Secretaría de Investigación y Posgrado, Laboratorio de Inmunotoxicología, Cd. de la Cultura s/n, Tepic, Nayarit, Mexico; Centro Nayarita de Innovación y Transferencia de Tecnología, Unidad Especializada Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA-Unidad Nayarit), Calle Tres s/n, Tepic, Nayarit, Mexico
| | - Migdalia Sarahy Navidad-Murrieta
- Centro Nayarita de Innovación y Transferencia de Tecnología, Unidad Especializada Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA-Unidad Nayarit), Calle Tres s/n, Tepic, Nayarit, Mexico
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CP 04510, Ciudad de México, Mexico.
| |
Collapse
|
8
|
Sharif K, Watad A, Bridgewood C, Kanduc D, Amital H, Shoenfeld Y. Insights into the autoimmune aspect of premature ovarian insufficiency. Best Pract Res Clin Endocrinol Metab 2019; 33:101323. [PMID: 31606343 DOI: 10.1016/j.beem.2019.101323] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Premature ovarian insufficiency (POI) refers to a continuum of decreasing ovarian function in women before the age of 40. To date, the cause of POI in the majority of cases remain unresolved. Many cases has been linked to genetic, toxic, infections, enzymatic and iatrogenic causes. A key function of the immune system is to identify and differentiate "self" and "non self" i.e. tolerance. Loss of self-tolerance results in an immune response against self-tissues and thus autoimmunity. Various investigations have highlighted the role of autoimmunity and its pertinence to POI. Several potential immune antigenic targets in the ovary have been reported to be involved in autoantibody induced autoimmune attack. The presence of lymphocytic oöphorits in ovarian samples of patients with POI provides histopathological evidence of autoimmune ovarian involvement. Finally, POI is strongly associated with other autoimmune conditions including for instance Addison disease, autoimmune polyglandular syndrome (APS) -1, APS-4, hypothyroidism, and diabetes mellitus among other autoimmune diseases. Taken together, these lines of evidence provide strong basis that support the role of autoimmunity as a potential cause of disease etiopathogenesis. Continuing research is increasingly providing more insight into the complex disease process. The aim of this review is to summarize the current literature related to the autoimmune nature of POI.
Collapse
Affiliation(s)
- Kassem Sharif
- Department of Medicine 'B', Tel-Hashomer, Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Leeds Institute of Rheumatic and Musculoskeletal Medicine, Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Abdulla Watad
- Department of Medicine 'B', Tel-Hashomer, Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Charlie Bridgewood
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Darja Kanduc
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Howard Amital
- Department of Medicine 'B', Tel-Hashomer, Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Russia.
| |
Collapse
|
9
|
Kisielow P. How does the immune system learn to distinguish between good and evil? The first definitive studies of T cell central tolerance and positive selection. Immunogenetics 2019; 71:513-518. [PMID: 31418051 PMCID: PMC6790186 DOI: 10.1007/s00251-019-01127-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 08/03/2019] [Indexed: 12/12/2022]
Abstract
Demonstration that immature CD4 + 8+ thymocytes contain T cell precursors that are subjected to positive and negative selection was the major step towards understanding how the adaptive immune system acquires the ability to distinguish foreign or abnormal (mutated or infected) self-cells from normal (healthy) cells. In the present review, the roles of TCR, CD4, CD8, and MHC molecules in intrathymic selection and some of the crucial experiments that contributed to the solution of the great immunological puzzle of self/nonself discrimination are described in an historical perspective. Recently, these experiments were highlighted by the immunological community by awarding the 2016 Novartis Prize for Immunology to Philippa Marrack, John Kappler, and Harald von Boehmer.
Collapse
Affiliation(s)
- Paweł Kisielow
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolf Weigl St. 12, 53-114, Wroclaw, Poland.
| |
Collapse
|
10
|
Williams AC, Hill LJ. Nicotinamide and Demographic and Disease transitions: Moderation is Best. Int J Tryptophan Res 2019; 12:1178646919855940. [PMID: 31320805 PMCID: PMC6610439 DOI: 10.1177/1178646919855940] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 05/03/2019] [Indexed: 12/13/2022] Open
Abstract
Good health and rapid progress depend on an optimal dose of nicotinamide. Too little meat triggers the neurodegenerative condition pellagra and tolerance of symbionts such as tuberculosis (TB), risking dysbioses and impaired resistance to acute infections. Nicotinamide deficiency is an overlooked diagnosis in poor cereal-dependant economies masquerading as 'environmental enteropathy' or physical and cognitive stunting. Too much meat (and supplements) may precipitate immune intolerance and autoimmune and allergic disease, with relative infertility and longevity, via the tryptophan-nicotinamide pathway. This switch favours a dearth of regulatory T (Treg) and an excess of T helper cells. High nicotinamide intake is implicated in cancer and Parkinson's disease. Pro-fertility genes, evolved to counteract high-nicotinamide-induced infertility, may now be risk factors for degenerative disease. Moderation of the dose of nicotinamide could prevent some common diseases and personalised doses at times of stress or, depending on genetic background or age, may treat some other conditions.
Collapse
Affiliation(s)
- Adrian C Williams
- Department of Neurology, University
Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Lisa J Hill
- School of Biomedical Sciences, Institute
of Clinical Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
11
|
Geenen V, Trussart C, Michaux H, Halouani A, Jaïdane H, Collée C, Renard C, Daukandt M, Ledent P, Martens H. The presentation of neuroendocrine self-peptides in the thymus: an essential event for individual life and vertebrate survival. Ann N Y Acad Sci 2019; 1455:113-125. [PMID: 31008523 PMCID: PMC6899491 DOI: 10.1111/nyas.14089] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/27/2019] [Accepted: 03/10/2019] [Indexed: 12/12/2022]
Abstract
Confirming Burnet's early hypothesis, elimination of self‐reactive T cells in the thymus was demonstrated in the late 1980s, and an important question immediately arose about the nature of the self‐peptides expressed in the thymus. Many genes encoding neuroendocrine‐related and tissue‐restricted antigens (TRAs) are transcribed in thymic epithelial cells (TECs). They are then processed for presentation by proteins of the major histocompatibility complex (MHC) expressed by TECs and thymic dendritic cells. MHC presentation of self‐peptides in the thymus programs self‐tolerance by two complementary mechanisms: (1) negative selection of self‐reactive “forbidden” T cell clones starting already in fetal life, and (2) generation of self‐specific thymic regulatory T lymphocytes (tTreg cells), mainly after birth. Many studies, including the discovery of the transcription factors autoimmune regulator (AIRE) and fasciculation and elongation protein zeta family zinc finger (FEZF2), have shown that a defect in thymus central self‐tolerance is the earliest event promoting autoimmunity. AIRE and FEZF2 control the level of transcription of many neuroendocrine self‐peptides and TRAs in the thymic epithelium. Furthermore, AIRE and FEZF2 mutations are associated with the development of autoimmunity in peripheral organs. The discovery of the intrathymic presentation of self‐peptides has revolutionized our knowledge of immunology and is opening novel avenues for prevention/treatment of autoimmunity.
Collapse
Affiliation(s)
- Vincent Geenen
- GIGA Institute, University of Liège, Liège-Sart Tilman, Belgium
| | | | - Hélène Michaux
- GIGA Institute, University of Liège, Liège-Sart Tilman, Belgium
| | - Aymen Halouani
- GIGA Institute, University of Liège, Liège-Sart Tilman, Belgium.,Faculty of Sciences and Faculty of Pharmacy, University of Tunis El Manar, Monastir, Tunisia
| | - Hela Jaïdane
- Faculty of Sciences and Faculty of Pharmacy, University of Tunis El Manar, Monastir, Tunisia
| | - Caroline Collée
- GIGA Institute, University of Liège, Liège-Sart Tilman, Belgium
| | - Chantal Renard
- GIGA Institute, University of Liège, Liège-Sart Tilman, Belgium
| | - Marc Daukandt
- X-Press Biologics, Industrial Park of Milmort, Liège, Belgium
| | - Philippe Ledent
- X-Press Biologics, Industrial Park of Milmort, Liège, Belgium
| | - Henri Martens
- GIGA Institute, University of Liège, Liège-Sart Tilman, Belgium
| |
Collapse
|
12
|
Abstract
The thymus is a primary lymphoid organ essential for the development of T lymphocytes, which orchestrate adaptive immune responses. T-cell development in the thymus is spatially regulated; key checkpoints in T-cell maturation and selection occur in cortical and medullary regions to eliminate self-reactive T cells, establish central tolerance, and export naïve T cells to the periphery with the potential to recognize diverse pathogens. Thymic output is also temporally regulated due to age-related involution of the thymus accompanied by loss of epithelial cells. This review discusses the structural and age-related control of thymus function in humans.
Collapse
Affiliation(s)
- Puspa Thapa
- Columbia Center for Translational Immunology, Columbia University Medical Center, 650 West 168th Street, BB1501, New York, NY 10032, USA
| | - Donna L Farber
- Department of Surgery, Columbia Center for Translational Immunology, Columbia University Medical Center, 650 West 168th Street, BB1501, New York, NY 10032, USA.
| |
Collapse
|
13
|
Campbell-Tofte J, Vrahatis A, Josefsen K, Mehlsen J, Winther K. Investigating the aetiology of adverse events following HPV vaccination with systems vaccinology. Cell Mol Life Sci 2019; 76:67-87. [PMID: 30324425 PMCID: PMC11105185 DOI: 10.1007/s00018-018-2925-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 09/10/2018] [Accepted: 09/20/2018] [Indexed: 12/18/2022]
Abstract
In contrast to the insidious and poorly immunogenic human papillomavirus (HPV) infections, vaccination with the HPV virus-like particles (vlps) is non-infectious and stimulates a strong neutralizing-antibody response that protects HPV-naïve vaccinees from viral infection and associated cancers. However, controversy about alleged adverse events following immunization (AEFI) with the vlps have led to extensive reductions in vaccine acceptance, with countries like Japan dropping it altogether. The AEFIs are grouped into chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME). In this review, we present a hypothesis that the AEFIs might arise from malfunctions within the immune system when confronted with the unusual antigen. In addition, we outline how the pathophysiology of the AEFIs can be cost-effectively investigated with the holistic principles of systems vaccinology in a two-step process. First, comprehensive immunological profiles of HPV vaccinees exhibiting the AEFIs are generated by integrating the data derived from serological profiling for prominent HPV antibodies and serum cytokines, with data from serum metabolomics, peripheral white blood cells transcriptomics and gut microbiome profiling. Next, the immunological profiles are compared with corresponding profiles generated for matched (a) HPV vaccinees without AEFIs; (b) non-HPV-vaccinated individuals with CFS/ME-like symptoms; and (c) non-HPV-vaccinated individuals without CFS/ME. In these comparisons, any causal links between HPV vaccine and the AEFIs, as well as the underlying molecular basis for the links will be revealed. Such a study should provide an objective basis for evaluating HPV vaccine safety and for identifying biomarkers for individuals at risk of developing AEFI with HPV vaccination.
Collapse
Affiliation(s)
| | | | - Knud Josefsen
- Bartholin Institute, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen Ø, Denmark
| | - Jesper Mehlsen
- Coordinating Research Centre, Bispebjerg and Frederiksberg Hospital, Nordre Fasanvej 57, 2000, Frederiksberg, Denmark
| | - Kaj Winther
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Nørre Allé 51, DK-2200, Copenhagen N, Denmark
| |
Collapse
|
14
|
Human T Cell Development, Localization, and Function throughout Life. Immunity 2018; 48:202-213. [PMID: 29466753 DOI: 10.1016/j.immuni.2018.01.007] [Citation(s) in RCA: 803] [Impact Index Per Article: 114.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/07/2017] [Accepted: 01/08/2018] [Indexed: 01/03/2023]
Abstract
Throughout life, T cells coordinate multiple aspects of adaptive immunity, including responses to pathogens, allergens, and tumors. In mouse models, the role of T cells is studied in the context of a specific type of pathogen, antigen, or disease condition over a limited time frame, whereas in humans, T cells control multiple insults simultaneously throughout the body and maintain immune homeostasis over decades. In this review, we discuss how human T cells develop and provide essential immune protection at different life stages and highlight tissue localization and subset delineation as key determinants of the T cell functional role in immune responses. We also discuss how anatomic compartments undergo distinct age-associated changes in T cell subset composition and function over a lifetime. It is important to consider age and tissue influences on human T cells when developing targeted strategies to modulate T cell-mediated immunity in vaccines and immunotherapies.
Collapse
|
15
|
Dominguez-Villar M, Hafler DA. Regulatory T cells in autoimmune disease. Nat Immunol 2018; 19:665-673. [PMID: 29925983 PMCID: PMC7882196 DOI: 10.1038/s41590-018-0120-4] [Citation(s) in RCA: 508] [Impact Index Per Article: 72.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 01/29/2018] [Indexed: 12/22/2022]
Abstract
In recent years, the understanding of regulatory T cell (Treg cell) biology has expanded considerably. Key observations have challenged the traditional definition of Treg cells and have provided insight into the underlying mechanisms responsible for the development of autoimmune diseases, with new therapeutic strategies that improve disease outcome. This Review summarizes the newer concepts of Treg cell instability, Treg cell plasticity and tissue-specific Treg cells, and their relationship to autoimmunity. Those three main concepts have changed the understanding of Treg cell biology: how they interact with other immune and non-immune cells; their functions in specific tissues; and the implications of this for the pathogenesis of autoimmune diseases.
Collapse
Affiliation(s)
| | - David A Hafler
- Department of Neurology, Yale School of Medicine, New Haven, CN, USA.
- Department of Immunobiology, Yale School of Medicine, New Haven, CN, USA.
| |
Collapse
|
16
|
Abstract
The microbiome can be defined as the sum of the microbial and host's genome. Recent information regarding this complex organ suggests that in animal models of multiple sclerosis (MS), the composition of the gut microbiome can be altered, giving rise to both the effector and regulatory phases of central nervous system (CNS) demyelination. Experimental findings during the past decade in animal models of MS have provided clear evidence for the significant role of gut microbes in both the effector and regulatory phase of this condition. There is mounting evidence in preliminary human studies suggesting that a dysbiotic MS gut microbiome could affect disease progression. We propose considering the gut microbiome as a key organ for the regulation of tolerance mechanisms and speculate that the gut microbiome is the major environmental risk factor for CNS demyelinating disease. Accordingly, we hypothesize that intervention of the gut microbiome could result in safer novel therapeutic strategies to treat MS.
Collapse
Affiliation(s)
| | - Trevor O Kirby
- Department of Biology, Eastern Washington University, Cheney, Washington 99004
| | - Lloyd H Kasper
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, New Hampshire 03756
| |
Collapse
|
17
|
Abstract
Regulatory T (Treg) cells are a distinct subset of CD4+ T cells. Instead of triggering adaptive immunity, they suppress immune responses. Small numbers of Treg cells reside within lymphoid organs and peripheral tissues, but their contribution to immune tolerance is so significant that defects in Treg cell function cause catastrophic immune disorders. Since they were first discovered 20 years ago, efforts have been made to understand the differences in developmental processes between Treg cells and conventional T cells that determine the ultimate fate of the overall T-cell population. Transcription factor Foxp3 is crucial for Treg cell differentiation, but it is not the whole story. Owing to recent advances in Treg cell research, we are now on the verge of appreciating the comprehensive mechanisms underlying Treg cell generation. Here, we discuss major discoveries, active study topics and remaining questions regarding Treg cell development.
Collapse
Affiliation(s)
- Wonyong Lee
- Department of Life Science, Sogang University, Mapo-gu, Seoul 04107, Korea
| | - Gap Ryol Lee
- Department of Life Science, Sogang University, Mapo-gu, Seoul 04107, Korea
| |
Collapse
|
18
|
Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors in cancer therapy: a focus on T-regulatory cells. Immunol Cell Biol 2017; 96:21-33. [PMID: 29359507 DOI: 10.1111/imcb.1003] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/13/2017] [Accepted: 09/06/2017] [Indexed: 01/02/2023]
Abstract
Regulatory T cells (Tregs) play essential roles in immune homeostasis; however, their role in tumor microenvironment (TME) is not completely evident. Several studies reported that infiltration of Tregs into various tumor tissues promotes tumor progression by limiting antitumor immunity and supporting tumor immune evasion. Furthermore, in TME, Tregs include heterogeneous subsets of cells expressing different immunosuppressive molecules favoring tumor progression. For an effective cancer therapy, it is critical to understand the Treg heterogeneity and biology in the TME. Recent studies have shown that immune checkpoint molecules promote cancer progression through various antitumor inhibitory mechanisms. Recent advances in cancer immunotherapy have shown the promising potentials of immune checkpoint inhibitors (ICIs) in inducing antitumor immune responses and clinical benefits in patients with cancer at late stages. Most studies revealed the effect of ICIs on T effector cells, and little is known about their effect on Tregs. In this review, we highlight the effects of the ICIs, including anti-CTLA-4, anti-PD-1/PD-L1, anti-LAG-3, anti-TIM-3, and anti-TIGIT, on tumor-infiltrating and peripheral Tregs to elicit effector T-cell functions against tumors. Additionally, we discuss how ICIs may target Tregs for cancer immunotherapy.
Collapse
Affiliation(s)
- Varun Sasidharan Nair
- Cancer Research Center, Qatar Biomedical Research Institute, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.,Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
19
|
Self-Transducible Bimodal PDX1-FOXP3 Protein Lifts Insulin Secretion and Curbs Autoimmunity, Boosting Tregs in Type 1 Diabetic Mice. Mol Ther 2017; 26:184-198. [PMID: 28988715 DOI: 10.1016/j.ymthe.2017.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 08/09/2017] [Accepted: 08/16/2017] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes (T1D) is characterized by massive destruction of insulin-producing β cells by autoreactive T lymphocytes, arising via defective immune tolerance. Therefore, effective anti-T1D therapeutics should combine autoimmunity-preventing and insulin production-restoring properties. We constructed a cell-permeable PDX1-FOXP3-TAT fusion protein (FP) composed of two transcription factors: forkhead box P3 (FOXP3), the master regulator of differentiation and functioning of self-tolerance-promoting Tregs, and pancreatic duodenal homeobox-1 (PDX1), the crucial factor supporting β cell development and maintenance. The FP was tested in vitro and in a non-obese diabetic mouse T1D model. In vitro, FP converted naive CD4+ T cells into a functional "Treg-like" subset, which suppressed cytokine secretion, downregulated antigen-specific responses, and curbed viability of diabetogenic effector cells. In hepatic stem-like cells, FP potentiated endocrine transdifferentiation, inducing expression of Insulin2 and other β lineage-specific genes. In vivo, FP administration to chronically diabetic mice triggered (1) a significant elevation of insulin and C-peptide levels, (2) the formation of insulin-containing cell clusters in livers, and (3) a systemic anti-inflammatory shift (higher Foxp3+CD4+CD25+ T cell frequencies, elevated rates of IL-10-producing cells, and reduced rates of IFN-γ-secreting cells). Overall, in accordance with its design, PDX1-FOXP3-TAT FP delivered both Treg-stabilizing anti-autoimmune and de novo insulin-producing effects, proving its anti-T1D therapeutic potential.
Collapse
|
20
|
Palacios-Arreola MI, Nava-Castro KE, Río-Araiza VHD, Pérez-Sánchez NY, Morales-Montor J. A single neonatal administration of Bisphenol A induces higher tumour weight associated to changes in tumour microenvironment in the adulthood. Sci Rep 2017; 7:10573. [PMID: 28874690 PMCID: PMC5585249 DOI: 10.1038/s41598-017-10135-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/27/2017] [Indexed: 01/29/2023] Open
Abstract
BPA is an oestrogenic endocrine disrupting chemical compound. Exposure to BPA in as early as pregnancy leads to lifelong effects. Since endocrine and immune systems interact in a bidirectional manner, endocrine disruption may cause permanent alterations of the immune system, affecting a future anti-tumoral response. Neonate (PND 3) female syngeneic BALB/c mice were exposed to a single dose of 250 µg/kg BPA. Once sexual maturity was reached, a mammary tumour was induced injecting 4T1 cells in situ, these cells are derived from a spontaneous adenocarcinoma in a BALB/c mouse and therefore allows for an immunocompetent recipient. After 25 days of injection, showing no major endocrine alterations, BPA-exposed mice developed larger tumours. Tumour leukocytic infiltrate analysis revealed a higher proportion of regulatory T lymphocytes in the BPA-exposed group. RT-PCR analysis of tumour samples showed a decreased expression of TNF-α and IFN-γ, as well as the M2 macrophage marker Fizz-1 in the BPA-exposed group. Flow cytometry analysis revealed differences in ERα expression by T lymphocytes, macrophages and NK cells, both associated to BPA exposure and tumour development. These findings show a new aspect whereby early life BPA exposure can contribute to breast cancer development and progression by modulating the anti-tumoral immune response.
Collapse
Affiliation(s)
- Margarita Isabel Palacios-Arreola
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70228, Ciudad de Mexico, CP, 04510, Mexico
| | - Karen Elizabeth Nava-Castro
- Laboratorio de Genotoxicología y Mutagénesis Ambientales, Departamento de Ciencias Ambientales, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de Mexico, CP 04510, Ciudad de Mexico, Mexico
| | - Víctor Hugo Del Río-Araiza
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70228, Ciudad de Mexico, CP, 04510, Mexico
| | - Nashla Yazmín Pérez-Sánchez
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70228, Ciudad de Mexico, CP, 04510, Mexico
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70228, Ciudad de Mexico, CP, 04510, Mexico.
| |
Collapse
|
21
|
Abstract
Immunity against Mycobacterium tuberculosis requires a balance between adaptive immune responses to constrain bacterial replication and the prevention of potentially damaging immune activation. Regulatory T (Treg) cells express the transcription factor Foxp3+ and constitute an essential counterbalance of inflammatory Th1 responses and are required to maintain immune homeostasis. The first reports describing the presence of Foxp3-expressing CD4+ Treg cells in tuberculosis (TB) emerged in 2006. Different Treg cell subsets, most likely specialized for different tissues and microenvironments, have been shown to expand in both human TB and animal models of TB. Recently, additional functional roles for Treg cells have been demonstrated during different stages and spectrums of TB disease. Foxp3+ regulatory cells can quickly expand during early infection and impede the onset of cellular immunity and persist during chronic TB infection. Increased frequencies of Treg cells have been associated with a detrimental outcome of active TB, and may be dependent on the M. tuberculosis strain, animal model, local environment, and the stage of infection. Some investigations also suggest that Treg cells are required together with effector T cell responses to obtain reduced pathology and sterilizing immunity. In this review, we will first provide an overview of the regulatory cells and mechanisms that control immune homeostasis. Then, we will review what is known about the phenotype and function of Treg cells from studies in human TB and experimental animal models of TB. We will discuss the potential role of Treg cells in the progression of TB disease and the relevance of this knowledge for future efforts to prevent, modulate, and treat TB.
Collapse
|
22
|
Dong Y, Li H, Li Y, Liu Y, Chen H, Xu P, Zhao T, He W. The role of regulatory T cells in thymectomy-induced autoimmune ovarian disease. Am J Reprod Immunol 2017; 78. [PMID: 28660639 DOI: 10.1111/aji.12683] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/14/2017] [Indexed: 12/28/2022] Open
Affiliation(s)
- Yajun Dong
- Reproductive Medical Center; Department of Obstetrics and Gynecology; Southwest Hospital; Third Military Medical University; Chongqing China
| | - Hongmei Li
- Reproductive Medical Center; Department of Obstetrics and Gynecology; Southwest Hospital; Third Military Medical University; Chongqing China
| | - Yuyan Li
- Reproductive Medical Center; Department of Obstetrics and Gynecology; Southwest Hospital; Third Military Medical University; Chongqing China
| | - Yonggang Liu
- Reproductive Medical Center; Department of Obstetrics and Gynecology; Southwest Hospital; Third Military Medical University; Chongqing China
| | - Huiling Chen
- Reproductive Medical Center; Department of Obstetrics and Gynecology; Southwest Hospital; Third Military Medical University; Chongqing China
| | - Pingping Xu
- Reproductive Medical Center; Department of Obstetrics and Gynecology; Southwest Hospital; Third Military Medical University; Chongqing China
| | - Tingting Zhao
- Institute of Immunology; Third Military Medical University; Chongqing China
| | - Wei He
- Reproductive Medical Center; Department of Obstetrics and Gynecology; Southwest Hospital; Third Military Medical University; Chongqing China
| |
Collapse
|
23
|
Penaloza-MacMaster P. CD8 T-cell regulation by T regulatory cells and the programmed cell death protein 1 pathway. Immunology 2017; 151:146-153. [PMID: 28375543 DOI: 10.1111/imm.12739] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/09/2017] [Accepted: 03/29/2017] [Indexed: 12/23/2022] Open
Abstract
The primary function of the immune system is to protect the host from infectious microorganisms and cancers. However, a major component of the immune response involves the direct elimination of cells in the body and the induction of systemic inflammation, which may result in life-threatening immunopathology. Therefore, the immune system has developed complex mechanisms to regulate itself with a specialized subset of CD4 T lymphocytes (referred to as regulatory T cells) and immune checkpoint pathways, such as the programmed cell death protein 1 pathway. These immune regulatory mechanisms can be exploited by pathogens and tumours to establish persistence in the host, warranting a deeper understanding of how to fine-tune immune responses during these chronic diseases. Here, I discuss various features of immune regulatory pathways and what important aspects must be considered in the next generation of therapies to reverse immune exhaustion, understanding that this process is a natural mechanism to prevent the host from destroying itself.
Collapse
Affiliation(s)
- Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
24
|
Sehrawat S, Rouse BT. Interplay of Regulatory T Cell and Th17 Cells during Infectious Diseases in Humans and Animals. Front Immunol 2017; 8:341. [PMID: 28421070 PMCID: PMC5377923 DOI: 10.3389/fimmu.2017.00341] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 03/09/2017] [Indexed: 12/14/2022] Open
Abstract
It is now clear that the outcome of an inflammatory process caused by infections depends on the balance of responses by several components of the immune system. Of particular relevance is the interplay between regulatory T cells (Tregs) and CD4+ T cells that produce IL-17 (Th17 cells) during immunoinflammatory events. In addition to discussing studies done in mice to highlight some unresolved issues in the biology of these cells, we emphasize the need to include outbred animals and humans in analyses. Achieving a balance between Treg and Th17 cells responses represents a powerful approach to control events during immunity and immunopathology.
Collapse
Affiliation(s)
- Sharvan Sehrawat
- Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Barry T Rouse
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
25
|
Rezende RM, Weiner HL. History and mechanisms of oral tolerance. Semin Immunol 2017; 30:3-11. [DOI: 10.1016/j.smim.2017.07.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 07/13/2017] [Indexed: 12/26/2022]
|
26
|
|
27
|
Karni A, Asmail A, Drory VE, Kolb H, Kesler A. Thymus involvement in myasthenia gravis: Epidemiological and clinical impacts of different self-tolerance breakdown mechanisms. J Neuroimmunol 2016; 298:58-62. [DOI: 10.1016/j.jneuroim.2016.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/02/2016] [Accepted: 07/06/2016] [Indexed: 01/24/2023]
|
28
|
Clark DA. The importance of being a regulatory T cell in pregnancy. J Reprod Immunol 2016; 116:60-9. [DOI: 10.1016/j.jri.2016.04.288] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 12/16/2022]
|
29
|
Hoeppli RE, Wu D, Cook L, Levings MK. The environment of regulatory T cell biology: cytokines, metabolites, and the microbiome. Front Immunol 2015; 6:61. [PMID: 25741338 PMCID: PMC4332351 DOI: 10.3389/fimmu.2015.00061] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/30/2015] [Indexed: 12/14/2022] Open
Abstract
Regulatory T cells (Tregs) are suppressive T cells that have an essential role in maintaining the balance between immune activation and tolerance. Their development, either in the thymus, periphery, or experimentally in vitro, and stability and function all depend on the right mix of environmental stimuli. This review focuses on the effects of cytokines, metabolites, and the microbiome on both human and mouse Treg biology. The role of cytokines secreted by innate and adaptive immune cells in directing Treg development and shaping their function is well established. New and emerging data suggest that metabolites, such as retinoic acid, and microbial products, such as short-chain fatty acids, also have a critical role in guiding the functional specialization of Tregs. Overall, the complex interaction between distinct environmental stimuli results in unique, and in some cases tissue-specific, tolerogenic environments. Understanding the conditions that favor Treg induction, accumulation, and function is critical to defining the pathophysiology of many immune-mediated diseases and to developing new therapeutic interventions.
Collapse
Affiliation(s)
- Romy E. Hoeppli
- Department of Surgery, Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Dan Wu
- Department of Surgery, Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Laura Cook
- Department of Surgery, Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Megan K. Levings
- Department of Surgery, Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
30
|
|
31
|
Abstract
Naturally occurring Foxp3(+)CD25(+)CD4(+) regulatory T (TREG) cells maintain immunological self-tolerance and prevent a variety of autoimmune diseases, including rheumatic diseases such as rheumatoid arthritis and systemic lupus erythematosus. In animal models of rheumatic disease, autoimmune responses can be controlled by re-establishing the T-cell balance in favour of TREG cells. Here we discuss three potential strategies for the clinical use of TREG cells to treat autoimmune rheumatic disease: expansion of self-antigen-specific natural TREG cells in vivo; propagation of antigen-specific natural TREG cells ex vivo, by in vitro antigenic stimulation, and subsequent transfer back into the host; or conversion of antigen-specific conventional T cells into TREG cells in vivo or ex vivo. These strategies require depletion of the effector T cells that mediate autoimmunity before initiating TREG-cell-based therapies. Immunotherapies that target TREG cells, and the balance of TREG cells and autoreactive T cells, are therefore an important modality for the treatment of autoimmune rheumatic disease.
Collapse
|
32
|
Bretscher PA. On the mechanism determining the TH1/TH2 phenotype of an immune response, and its pertinence to strategies for the prevention, and treatment, of certain infectious diseases. Scand J Immunol 2014; 79:361-76. [PMID: 24684592 PMCID: PMC4282429 DOI: 10.1111/sji.12175] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 03/27/2014] [Indexed: 12/31/2022]
Abstract
It is well recognized that the physiological/pathological consequences of an immune response, against a foreign or a self-antigen, are often critically dependent on the class of immunity generated. Here we focus on how antigen interacts with the cells of the immune system to determine whether antigen predominantly generates Th1 or Th2 cells. We refer to this mechanism as the 'decision criterion' controlling the Th1/Th2 phenotype of the immune response. A plausible decision criterion should account for the variables of immunization known to affect the Th1/Th2 phenotype of the ensuing immune response. Documented variables include the nature of the antigen, in terms of its degree of foreignness, the dose of antigen and the time after immunization at which the Th1/Th2 phenotype of the immune response is assessed. These are quantitative variables made at the level of the system. In addition, the route of immunization is also critical. I describe a quantitative hypothesis as to the nature of the decision criterion, referred to as the Threshold Hypothesis. This hypothesis accounts for the quantitative variables of immunization known to affect the Th1/Th2 phenotype of the immune response generated. I suggest and illustrate how this is not true of competing, contemporary hypotheses. I outline studies testing predictions of the hypothesis and illustrate its potential utility in designing strategies to prevent or treat medical situations where a predominant Th1 response is required to contain an infection, such as those caused by HIV-1 and by Mycobacterium tuberculosis, or to contain cancers.
Collapse
Affiliation(s)
- P A Bretscher
- University of SaskatchewanSaskatoon, Saskatchewan, Canada
| |
Collapse
|
33
|
Kisand K, Peterson P, Laan M. Lymphopenia-induced proliferation in aire-deficient mice helps to explain their autoimmunity and differences from human patients. Front Immunol 2014; 5:51. [PMID: 24592265 PMCID: PMC3923166 DOI: 10.3389/fimmu.2014.00051] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/29/2014] [Indexed: 12/23/2022] Open
Abstract
Studies on autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (APECED) and its mouse model – both caused by mutant AIRE – have greatly advanced the understanding of thymic processes that generate a self-tolerant T-cell repertoire. Much is now known about the molecular mechanisms by which AIRE induces tissue-specific antigen expression in thymic epithelium, and how this leads to negative selection of auto-reactive thymocytes. However, we still do not understand the processes that lead to the activation of any infrequent naïve auto-reactive T-cells exported by AIRE-deficient thymi. Also, the striking phenotypic differences between APECED and its mouse models have puzzled researchers for years. The aim of this review is to suggest explanations for some of these unanswered questions, based on a fresh view of published experiments. We review evidence that auto-reactive T-cells can be activated by the prolonged neonatal lymphopenia that naturally develops in young Aire-deficient mice due to delayed export of mature thymocytes. Lymphopenia-induced proliferation (LIP) helps to fill the empty space; by favoring auto-reactive T-cells, it also leads to lymphocyte infiltration in the same tissues as in day 3 thymectomized animals. The LIP becomes uncontrolled when loss of Aire is combined with defects in genes responsible for anergy induction and Treg responsiveness, or in signaling from the T-cell receptor and homeostatic cytokines. In APECED patients, LIP is much less likely to be involved in activation of naïve auto-reactive T-cells, as humans are born with a more mature immune system than in neonatal mice. We suggest that human AIRE-deficiency presents with different phenotypes because of additional precipitating factors that compound the defective negative selection of potentially autoaggressive tissue-specific thymocytes.
Collapse
Affiliation(s)
- Kai Kisand
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu , Tartu , Estonia
| | - Pärt Peterson
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu , Tartu , Estonia
| | - Martti Laan
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu , Tartu , Estonia
| |
Collapse
|
34
|
El Chami H, Hassoun PM. Inflammatory mechanisms in the pathogenesis of pulmonary arterial hypertension. Compr Physiol 2013; 1:1929-41. [PMID: 23733693 DOI: 10.1002/cphy.c100028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inflammation is a prominent feature of human and experimental pulmonary hypertension (PH) as suggested by infiltration of various inflammatory cells and increased expression of certain cytokines in remodeled pulmonary vessels. Macrophages, T and B lymphocytes, and dendritic cells are found in the vascular lesions of idiopathic pulmonary arterial hypertension (PAH) as well as in PAH associated with connective tissue diseases or infectious etiologies such as HIV. In addition, PAH is often characterized by the presence of circulating chemokines and cytokines, increased expression of growth (such as VEGF and PDGF) and transcriptional (e.g., nuclear factor of activated T cells or NFAT) factors, and viral protein components (e.g., HIV-1 Nef), which directly contribute to further recruitment of inflammatory cells and the pulmonary vascular remodeling process. These inflammatory pathways may thus serve as potential specific therapeutic targets. This article provides an overview of inflammatory pathways involving chemokines and cytokines as well as growth factors, highlighting their potential role in pulmonary vascular remodeling and the possibility of future targeted therapy.
Collapse
Affiliation(s)
- Hala El Chami
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | | |
Collapse
|
35
|
Foxp3(+) regulatory T cells in tuberculosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 783:165-80. [PMID: 23468109 DOI: 10.1007/978-1-4614-6111-1_9] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The immune response to Mycobacterium tuberculosis (Mtb) must be tightly regulated to mount a sufficient response to limit bacterial growth and dissemination while avoiding excessive inflammation that could damage host tissues. A wide variety of cell types, cell surface molecules, and cytokines are likely to contribute to this regulation, but recent studies have revealed that a subset of CD4 T cells expressing the transcription factor Foxp3, called regulatory T (reg) cells, play a critical role [1-3]. Although the first reports of T reg cells in tuberculosis (TB) occurred only recently (i.e., 2006) [4, 5], we have already gained many insights into their activity during TB. While it is likely that T reg cells do play some beneficial roles by preventing inflammation-mediated damage to host tissues during TB, this aspect of their function has not been well studied to date. What is clear, however, is that during the initial T cell response to Mtb infection, Mtb induces the expansions of T reg cells that delay the onset of adaptive immunity, suggesting that Mtb has hijacked T reg cell-mediated immune suppression to allow it to replicate unabated in the lung until T cells finally arrive [6]. In this chapter, we will first provide an overview of the delayed T cell response to Mtb and a brief introduction to regulatory T cells. We will then review what is known about T reg cells from observations in human populations, discuss mechanistic insights revealed in the mouse model, and speculate about the relevance of this understanding for future efforts to prevent and treat TB.
Collapse
|
36
|
Schmitt EG, Williams CB. Generation and function of induced regulatory T cells. Front Immunol 2013; 4:152. [PMID: 23801990 PMCID: PMC3685796 DOI: 10.3389/fimmu.2013.00152] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/04/2013] [Indexed: 12/17/2022] Open
Abstract
CD4+ CD25+ Foxp3+ regulatory T (Treg) cells are essential to the balance between pro- and anti-inflammatory responses. There are two major subsets of Treg cells, “natural” Treg (nTreg) cells that develop in the thymus, and “induced” Treg (iTreg) cells that arise in the periphery from CD4+ Foxp3− conventional T cells and can be generated in vitro. Previous work has established that both subsets are required for immunological tolerance. Additionally, in vitro-derived iTreg cells can reestablish tolerance in situations where Treg cells are decreased or defective. This review will focus on iTreg cells, drawing comparisons to nTreg cells when possible. We discuss the molecular mechanisms of iTreg cell induction, both in vivo and in vitro, review the Foxp3-dependent and -independent transcriptional landscape of iTreg cells, and examine the proposed suppressive mechanisms utilized by each Treg cell subset. We also compare the T cell receptor repertoire of the Treg cell subsets, discuss inflammatory conditions where iTreg cells are generated or have been used for treatment, and address the issue of iTreg cell stability.
Collapse
Affiliation(s)
- Erica G Schmitt
- Section of Rheumatology, Department of Pediatrics, Medical College of Wisconsin , Milwaukee, WI , USA
| | | |
Collapse
|
37
|
Banerjee A, Vasanthakumar A, Grigoriadis G. Modulating T regulatory cells in cancer: how close are we? Immunol Cell Biol 2013; 91:340-9. [PMID: 23567897 DOI: 10.1038/icb.2013.12] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Regulatory T cells (Tregs) are a specialized subset of CD4 T cells that have an indispensable role in maintaining immune homeostasis and tolerance. Although studies in mice and humans have clearly highlighted that the absence of these cells results in severe autoimmunity and inflammation, increased Treg numbers and/or function is not always beneficial. This is best exemplified in certain cancers where increased Tregs promote cancer progression by interfering with immune surveillance. Conversely, in other types of cancers that have an inflammatory component, Tregs can inhibit cancer progression by dampening inflammation. In this review article, we provide a historical perspective of the discovery of Tregs, followed by a summary of the existing literature on the role of Tregs in malignancy.
Collapse
Affiliation(s)
- Ashish Banerjee
- Centre for Inflammatory Diseases, Monash Medical Centre, Southern Clinical School, Monash University, Clayton, Victoria, Australia.
| | | | | |
Collapse
|
38
|
Quintana FJ. The aryl hydrocarbon receptor: a molecular pathway for the environmental control of the immune response. Immunology 2013. [PMID: 23190340 DOI: 10.1111/imm.12046] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Environmental factors have significant effects on the development of autoimmune diseases. The ligand-activated transcription factor aryl hydrocarbon receptor (AHR) is controlled by endogenous and environmental small molecules. Hence, AHR provides a molecular pathway by which endogenous and environmental signals can influence the immune response and the development of autoimmune diseases. AHR also provides a target for therapeutic intervention in immune-mediated disorders. In this review, we discuss the role of AHR in the regulation of T-cell differentiation and autoimmunity.
Collapse
Affiliation(s)
- Francisco J Quintana
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
39
|
Kurobe H, Tominaga T, Sugano M, Hayabuchi Y, Egawa Y, Takahama Y, Kitagawa T. Complete but not partial thymectomy in early infancy reduces T-cell-mediated immune response: three-year tracing study after pediatric cardiac surgery. J Thorac Cardiovasc Surg 2013; 145:656-62, 662.e1-2; discussion 662. [PMID: 23312343 DOI: 10.1016/j.jtcvs.2012.12.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 10/29/2012] [Accepted: 12/05/2012] [Indexed: 10/27/2022]
Abstract
OBJECTIVE Thymectomy is often performed to secure an operative field in surgery for congenital heart defects in early infancy. However, how neonatal thymectomy affects the subsequent development of the immune system in humans remains unclear. We monitored patients for 3 years from the time of thymectomy that was performed during cardiac surgery in early infancy. METHODS For up to 3 years, we monitored the number of circulating lymphocytes and the clinical course of the children who underwent complete (n = 17), partial, and no (n = 15) thymectomy during congenital heart defect surgery performed at less than 3 months of age. The titers of immunoglobulin-G produced in response to vaccinated viruses and phytohemagglutinin responses were also measured. RESULTS Six months after surgery, the number of T cells, including CD4(+) and CD8(+) subpopulations, decreased in patients with complete but not partial thymectomy. The reduction in T-cell number persisted for 3 years, whereas the number of B cells did not change. In patients with complete thymectomy, the titers of immunoglobulin-G produced in response to vaccinated measles and rubella viruses were reduced, whereas the phytohemagglutinin-induced proliferation of T cells was not impaired. In addition, hospitalization frequency associated with infectious diseases increased in patients with complete but not partial thymectomy. CONCLUSIONS The results revealed that complete thymectomy in early infancy reduces the number of circulating T cells and T-cell-mediated immune responses for at least 3 years, suggesting that the thymus should be at least partially preserved during surgery in early infancy to maintain protective immunity.
Collapse
Affiliation(s)
- Hirotsugu Kurobe
- Department of Cardiovascular Surgery, Institute for Health Biosciences, Graduate School of Medical Sciences, The University of Tokushima, Tokushima, Japan.
| | | | | | | | | | | | | |
Collapse
|
40
|
Smith KA. The molecular mechanisms of regulatory T cell immunosuppression. Front Immunol 2012; 3:379. [PMID: 23248628 PMCID: PMC3522108 DOI: 10.3389/fimmu.2012.00379] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 11/27/2012] [Indexed: 02/03/2023] Open
Affiliation(s)
- Kendall A Smith
- Division of Immunology, Department of Medicine, Weill Medical College, Cornell University New York, NY, USA
| |
Collapse
|
41
|
Predictors of immunosuppressive regulatory T lymphocytes in healthy women. J Cancer Epidemiol 2012; 2012:191090. [PMID: 22969801 PMCID: PMC3433139 DOI: 10.1155/2012/191090] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 07/12/2012] [Indexed: 01/04/2023] Open
Abstract
Immunosuppressive regulatory T (Treg) cells play an important role in antitumor immunity, self-tolerance, transplantation tolerance, and attenuation of allergic response. Higher proportion of Treg cells has been observed in peripheral blood of cancer cases compared to controls. Little is known about potential epidemiological predictors of Treg cell levels in healthy individuals. We conducted a cross-sectional study including 75 healthy women, between 20 and 80 years of age, who participated in the Data Bank and BioRepository (DBBR) program at Roswell Park Cancer Institute (RPCI), Buffalo, NY, USA. Peripheral blood levels of CD4+CD25+FOXP3+ Treg cells were measured using flow cytometric analysis. A range of risk factors was evaluated using Wilcoxon Rank-Sum test, Kruskal-Wallis test, and linear regression. Age, smoking, medications for treatment of osteoporosis, postmenopausal status, body mass index (BMI), and hormone replacement therapy (HRT) were found to be significant positive predictors of Treg cell levels in peripheral blood (P ≤ 0.05). Higher education, exercise, age at first birth, oral contraceptives, and use of Ibuprofen were found be significant (P < 0.05) negative predictors of Treg levels. Thus, various epidemiological risk factors might explain interindividual variation in immune response to pathological conditions, including cancer.
Collapse
|
42
|
Kim YH, Kim HJ, Kim JS, Park CG. Application of Regulatory T Cells in Transplantation Field. KOREAN JOURNAL OF TRANSPLANTATION 2012. [DOI: 10.4285/jkstn.2012.26.2.74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Yong-Hee Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
- Transplantation Research Institute SNUMRC, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute and TIMRC, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun-Je Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
- Transplantation Research Institute SNUMRC, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute and TIMRC, Seoul National University College of Medicine, Seoul, Korea
| | - Jung-Sik Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
- Transplantation Research Institute SNUMRC, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute and TIMRC, Seoul National University College of Medicine, Seoul, Korea
| | - Chung-Gyu Park
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
- Transplantation Research Institute SNUMRC, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute and TIMRC, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
43
|
Takahashi T, Shiku H. Cell surface antigens: invaluable landmarks reflecting the nature of cells. CANCER IMMUNITY 2012; 12:2. [PMID: 22896747 PMCID: PMC3380351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Affiliation(s)
- Toshitada Takahashi
- Comprehensive Health Science Center, Aichi Health Promotion Foundation, Higashiura Town, Aichi, Japan.
| | | |
Collapse
|
44
|
Collinge M, Burns-Naas LA, Chellman GJ, Kawabata TT, Komocsar WJ, Piccotti JR, Shenton J, Wierda D. Developmental immunotoxicity (DIT) testing of pharmaceuticals: Current practices, state of the science, knowledge gaps, and recommendations. J Immunotoxicol 2012; 9:210-30. [DOI: 10.3109/1547691x.2012.661486] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
45
|
Schmetterer KG, Neunkirchner A, Pickl WF. Naturally occurring regulatory T cells: markers, mechanisms, and manipulation. FASEB J 2012; 26:2253-76. [DOI: 10.1096/fj.11-193672] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Klaus G. Schmetterer
- Institute of ImmunologyCenter for Pathophysiology, Infectiology, and ImmunologyMedical University of ViennaViennaAustria
| | - Alina Neunkirchner
- Institute of ImmunologyCenter for Pathophysiology, Infectiology, and ImmunologyMedical University of ViennaViennaAustria
- Christian Doppler Laboratory for ImmunmodulationViennaAustria
| | - Winfried F. Pickl
- Institute of ImmunologyCenter for Pathophysiology, Infectiology, and ImmunologyMedical University of ViennaViennaAustria
- Christian Doppler Laboratory for ImmunmodulationViennaAustria
| |
Collapse
|
46
|
Abstract
The immune system has evolved to mount an effective defense against pathogens and to minimize deleterious immune-mediated inflammation caused by commensal microorganisms, immune responses against self and environmental antigens, and metabolic inflammatory disorders. Regulatory T (Treg) cell-mediated suppression serves as a vital mechanism of negative regulation of immune-mediated inflammation and features prominently in autoimmune and autoinflammatory disorders, allergy, acute and chronic infections, cancer, and metabolic inflammation. The discovery that Foxp3 is the transcription factor that specifies the Treg cell lineage facilitated recent progress in understanding the biology of regulatory T cells. In this review, we discuss cellular and molecular mechanisms in the differentiation and function of these cells.
Collapse
Affiliation(s)
- Steven Z Josefowicz
- Howard Hughes Medical Institute and Immunology Program, Sloan Kettering Institute, New York, NY 10021, USA
| | | | | |
Collapse
|
47
|
Kang Y, Zheng G, Chen A, Wang J, Hu Y, Li J, Zhang J, Gao W, Fu H, Xia G, Wang B. Tolerogenic DNA vaccine for prevention of autoimmune ovarian disease. Immunol Invest 2012; 41:249-60. [PMID: 22221010 DOI: 10.3109/08820139.2011.622828] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
DNA vaccines have been widely used to induce immune responses against molecular targets. In this study, we explored the possibility of using DNA vaccine combined with the immunosuppressant FK506 (tacrolimus) to antigen-specifically suppress unwanted immune responses and prevent autoimmune ovarian disease. To that end, we immunized C57BL/6 mice with a DNA vaccine encoding mouse zona pellucida 3 (ZP3) together with FK506. The immunization induced ZP3-specific CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg), which suppressed the induction of ZP3-specific delayed-type hypersensitivity in the animals. Significantly, the immunization also protected the animals from experimentally induced autoimmune ovarian disease. These results suggest that DNA vaccination in the presence of FK506 may be used to induce Treg cells and prevent AOD.
Collapse
Affiliation(s)
- Youmin Kang
- State Key Laboratory for Agro-Biotechnology, Key Laboratory of Microbiological Resources and Applications of the Ministry of Agriculture, College of Biological Science, China Agricultural University, Beijing 100193, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Otsuka N, Tong ZB, Vanevski K, Tu W, Cheng MH, Nelson LM. Autoimmune oophoritis with multiple molecular targets mitigated by transgenic expression of mater. Endocrinology 2011; 152:2465-73. [PMID: 21447630 PMCID: PMC3100611 DOI: 10.1210/en.2011-0022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 03/09/2011] [Indexed: 11/19/2022]
Abstract
Primary ovarian insufficiency (POI) resulting from ovarian autoimmunity is a poorly understood clinical condition lacking in effective treatments. Understanding the targets of the autoimmune response and induction of ovarian-specific tolerance would allow development of focused therapies to preserve fertility in an at-risk population. MATER (maternal antigen that embryos require) is a known ovarian autoantigen targeted in autoimmune syndromes of POI. We attempt to induce ovarian-specific tolerance via transgenic expression of the MATER antigen on potentially tolerogenic antigen-presenting cells (APC), which typically present antigen via the major histocompatibility complex (MHC) class II molecule. We hypothesize that expression of MATER in a MHC class II-dependent manner on APC can mediate induction of ovarian tolerance. We utilized a well-characterized murine model of ovarian autoimmunity, whereby oophoritis develops after d 3 neonatal thymectomy (NTx). Wild-type and transgenic mice, carrying an MHC Class II-driven Mater gene (IE-Mater), were subjected to NTx and assessed for evidence of autoimmune oophoritis. After disease induction by NTx, female mice carrying the IE-Mater transgene had significant reductions in histological oophoritis (56%) and circulating ovarian autoantibodies (28%) compared with wild-type females (94% and 82%, respectively). Incidence of other autoimmunity was unaffected as assessed by antinuclear autoantibodies. Transgenic expression of MATER in APC can induce antigen-specific tolerance with a significant reduction in ovarian autoimmunity. Lack of complete disease protection suggests that other antigens may also play a role in autoimmune oophoritis. As a known autoantigen in the human APS1 (autoimmune polyglandular syndrome type 1), which is associated with POI, MATER may represent a relevant target for future diagnostic and therapeutic clinical interventions.
Collapse
Affiliation(s)
- Noriyuki Otsuka
- University of California San Francisco Diabetes Center, San Francisco, California 94143, USA
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Despite the skepticism that once prevailed among immunologists, it is now widely accepted that the normal immune system harbors a T-cell population, called regulatory T cells (Treg cells), specialized for immune suppression. It was first shown that depletion of a T-cell subpopulation from normal rodents produced autoimmune disease. Search for a molecular marker specific for such autoimmune-preventive Treg cells has revealed that the majority, if not all, of them constitutively express the CD25 molecule as depletion of CD25(+)CD4(+) T cells spontaneously evokes autoimmune disease in otherwise normal rodents. The expression of CD25 by Treg cells has made it possible to delineate their developmental pathways, in particular their thymic development, and establish simple in vitro assay for assessing their suppressive activity. The marker and the in vitro assay have helped to identify human Treg cells with similar functional and phenotypic characteristics. Recent efforts have shown that natural Treg cells specifically express the transcription factor Foxp3 and that mutations of the Foxp3 gene produce a variety of immunological diseases in humans and rodents. Specific expression of Foxp3 in natural Treg cells has enabled their functional and developmental characterization by genetic approach. These studies altogether have provided firm evidence for Foxp3(+)CD25(+)CD4(+) Treg cells as an indispensable cellular constituent of the normal immune system for establishing and maintaining immunologic self-tolerance and immune homeostasis. Treg cells are now within the scope of clinical use to treat immunological diseases and control physiological and pathological immune responses.
Collapse
|
50
|
Hartigan-O'Connor DJ, Hirao LA, McCune JM, Dandekar S. Th17 cells and regulatory T cells in elite control over HIV and SIV. Curr Opin HIV AIDS 2011; 6:221-7. [PMID: 21399494 PMCID: PMC4079838 DOI: 10.1097/coh.0b013e32834577b3] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE OF REVIEW We present current findings about two subsets of CD4+ T cells that play an important part in the initial host response to infection with the HIV type 1: those producing IL-17 (Th17 cells) and those with immunosuppressive function (CD25+FoxP3+ regulatory T cells or T-reg). The role of these cells in the control of viral infection and immune activation as well as in the prevention of immune deficiency in HIV-infected elite controllers will be examined. We will also discuss the use of the simian immunodeficiency virus (SIV)-infected macaque model of AIDS to study the interplay between these cells and lentiviral infection in vivo. RECENT FINDINGS Study of Th17 cells in humans and nonhuman primates (NHPs) has shown that depletion of these cells is associated with the dissemination of microbial products from the infected gut, increased systemic immune activation, and disease progression. Most impressively, having a smaller Th17-cell compartment has been found to predict these outcomes. T-reg have been associated with the reduced antiviral T-cell responses but not with the suppression of generalized T cell activation. Both cell subsets influence innate immune responses and, in doing so, may shape the inflammatory milieu of the host at infection. SUMMARY Interactions between Th17 cells, T-reg, and cells of the innate immune system influence the course of HIV and SIV infection from its earliest stages, even before the appearance of adaptive immunity. Such interactions may be pivotal for elite control over disease progression.
Collapse
Affiliation(s)
- Dennis J Hartigan-O'Connor
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, USA.
| | | | | | | |
Collapse
|