151
|
Reading JL, Gálvez-Cancino F, Swanton C, Lladser A, Peggs KS, Quezada SA. The function and dysfunction of memory CD8 + T cells in tumor immunity. Immunol Rev 2018; 283:194-212. [PMID: 29664561 DOI: 10.1111/imr.12657] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The generation and maintenance of CD8+ T cell memory is crucial to long-term host survival, yet the basic tenets of CD8+ T cell immunity are still being established. Recent work has led to the discovery of tissue-resident memory cells and refined our understanding of the transcriptional and epigenetic basis of CD8+ T cell differentiation and dysregulation. In parallel, the unprecedented clinical success of immunotherapy has galvanized an intense, global research effort to decipher and de-repress the anti-tumor response. However, the progress of immunotherapy is at a critical juncture, since the efficacy of immuno-oncology agents remains confined to a fraction of patients and often fails to provide durable benefit. Unlocking the potential of immunotherapy requires the design of strategies that both induce a potent effector response and reliably forge stable, functional memory T cell pools capable of protecting from recurrence or relapse. It is therefore essential that basic and emerging concepts of memory T cell biology are rapidly and faithfully transposed to advance therapeutic development in cancer immunotherapy. This review highlights seminal and recent reports in CD8+ T cell memory and tumor immunology, and evaluates recent data from solid cancer specimens in the context of the key paradigms from preclinical models. We elucidate the potential significance of circulating effector cells poised downstream of neoantigen recognition and upstream of T cell dysfunction and propose that cells in this immunological 'sweet spot' may be key anti-tumor effectors.
Collapse
Affiliation(s)
- James L Reading
- Cancer Immunology Unit, University College London Cancer Institute, University College London, London, UK
- Research Department of Haematology, University College London Cancer Institute, University College London, London, UK
| | | | | | - Alvaro Lladser
- Laboratory of Gene Immunotherapy, Fundación Ciencia & Vida, Santiago, Chile
| | - Karl S Peggs
- Cancer Immunology Unit, University College London Cancer Institute, University College London, London, UK
- Research Department of Haematology, University College London Cancer Institute, University College London, London, UK
| | - Sergio A Quezada
- Cancer Immunology Unit, University College London Cancer Institute, University College London, London, UK
- Research Department of Haematology, University College London Cancer Institute, University College London, London, UK
| |
Collapse
|
152
|
Piergallini TJ, Turner J. Tuberculosis in the elderly: Why inflammation matters. Exp Gerontol 2018; 105:32-39. [PMID: 29287772 PMCID: PMC5967410 DOI: 10.1016/j.exger.2017.12.021] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/22/2017] [Accepted: 12/22/2017] [Indexed: 12/20/2022]
Abstract
Growing old is associated with an increase in the basal inflammatory state of an individual and susceptibility to many diseases, including infectious diseases. Evidence is growing to support the concept that inflammation and disease susceptibility in the elderly is linked. Our studies focus on the infectious disease tuberculosis (TB), which is caused by Mycobacterium tuberculosis (M.tb), a pathogen that infects approximately one fourth of the world's population. Aging is a major risk factor for developing TB, and inflammation has been strongly implicated. In this review we will discuss the relationship between inflammation in the lung and susceptibility to develop and succumb to TB in old age. Further understanding of the relationship between inflammation, age, and M.tb will lead to informed decisions about TB prevention and treatment strategies that are uniquely designed for the elderly.
Collapse
Affiliation(s)
- Tucker J Piergallini
- Texas Biomedical Research Institute, San Antonio, TX 78227, United States; College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - Joanne Turner
- Texas Biomedical Research Institute, San Antonio, TX 78227, United States.
| |
Collapse
|
153
|
Rudolph ME, McArthur MA, Barnes RS, Magder LS, Chen WH, Sztein MB. Differences Between Pediatric and Adult T Cell Responses to In Vitro Staphylococcal Enterotoxin B Stimulation. Front Immunol 2018; 9:498. [PMID: 29616025 PMCID: PMC5869216 DOI: 10.3389/fimmu.2018.00498] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 02/26/2018] [Indexed: 12/23/2022] Open
Abstract
Toxic shock syndrome (TSS) is capable of inducing life-threatening fever, rash, and systemic organ failure, though the specific mechanisms behind these symptoms remain poorly understood. Staphylococcal enterotoxin B (SEB) and other superantigens have shown to be important factors in TSS, capable of promoting cross-linking between T cell receptors and major histocompatibility complexes which results in overwhelming T cell activation, proliferation, and cytokine production. The resulting proinflammatory cytokine cascade, often referred to as the “cytokine storm,” seems to be critical to the development of disease. Interestingly, clinical studies have shown that children exhibit less severe TSS-associated morbidity than adults, though the mechanism behind this phenomenon has not been addressed. Indeed, despite the fact that most novel antigen exposure occurs early in life, be it from environmentally acquired pathogens or routine vaccination, normal pediatric T cell immune functions remain critically underexplored. This is largely due to difficulty in obtaining enough samples to explore more than a narrow sliver of the cell-mediated immune compartment. To address this limitation, we optimized a T effector (Teff)/circulating T follicular helper (cTFH) cell mass cytometry panel which allowed us to analyze a wide array of T cell populations and effector functions following in vitro SEB stimulation. We show that T cell activation—as measured by CD69 expression—following SEB stimulation is lower in pediatric participants, increasing throughout childhood, and reaching adult levels by around 15 years old. Further, while individual CD4+ effector memory T cell (TEM) effector molecules show limited age-associated differences following SEB stimulation, multifunctional CD4+ TEM are shown to positively correlate with increasing age through adolescence. Individual CD8+ TEM effectors and multifunctional phenotypes also show very strong age-associated increases following SEB stimulation. SEB stimulation has little impact on cTFH activation or functional cellular markers, regardless of age. These results, coupled with the fact that a robust proinflammatory cytokine response seems critical to developing severe TSS, suggest a possible connection between the significantly reduced T cell activation and multifunctional populations following in vitro SEB stimulation in our pediatric participants and clinical observations relating to reduced TSS mortality in children.
Collapse
Affiliation(s)
- Mark E Rudolph
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, United States.,Molecular Microbiology and Immunology Department, University of Maryland Graduate Program in Life Sciences, Baltimore, MD, United States
| | - Monica A McArthur
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Robin S Barnes
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Laurence S Magder
- Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Wilbur H Chen
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marcelo B Sztein
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, United States.,Molecular Microbiology and Immunology Department, University of Maryland Graduate Program in Life Sciences, Baltimore, MD, United States.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
154
|
Franzese O, Barbaccia ML, Bonmassar E, Graziani G. Beneficial and Detrimental Effects of Antiretroviral Therapy on HIV-Associated Immunosenescence. Chemotherapy 2018; 63:64-75. [PMID: 29533947 DOI: 10.1159/000487534] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 02/06/2018] [Indexed: 02/28/2024]
Abstract
Since the introduction of highly active antiretroviral therapy more than 2 decades ago, HIV-related deaths have dramatically decreased and HIV infection has become a chronic disease. Due to the inability of antiretroviral drugs to eradicate the virus, treatment of HIV infection requires a systemic lifelong therapy. However, even when successfully treated, HIV patients still show increased incidence of age-associated co-morbidities compared with uninfected individuals. Virus- induced immunosenescence, a process characterized by a progressive decline of immune system function, contributes to the premature ageing observed in HIV patients. Although antiretroviral therapy has significantly improved both the quality and length of patient lives, the life expectancy of treated patients is still shorter compared with that of uninfected individuals. In particular, while antiretroviral therapy can contrast some features of HIV-associated immunosenescence, several anti-HIV agents may themselves contribute to other aspects of immune ageing. Moreover, older HIV patients tend to have a worse immunological response to the antiviral therapy. In this review we will examine the available evidence on the role of antiretroviral therapy in the control of the main features regulating immunosenescence.
Collapse
|
155
|
Wiest M, Upchurch K, Yin W, Ellis J, Xue Y, Lanier B, Millard M, Joo H, Oh S. Clinical implications of CD4 + T cell subsets in adult atopic asthma patients. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2018; 14:7. [PMID: 29507584 PMCID: PMC5833086 DOI: 10.1186/s13223-018-0231-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/22/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND T cells play a central role in chronic inflammation in asthma. However, the roles of individual subsets of T cells in the pathology of asthma in patients remain to be better understood. METHODS We investigated the potential signatures of T cell subset phenotypes in asthma using fresh whole blood from adult atopic asthma patients (n = 43) and non-asthmatic control subjects (n = 22). We further assessed their potential clinical implications by correlating asthma severity. RESULTS We report four major features of CD4+ T cells in the blood of atopic asthma patients. First, patients had a profound increase of CCR7+ memory CD4+ T cells, but not CCR7- memory CD4+ T cells. Second, an increase in CCR4+ CD4+ T cells in patients was mainly attributed to the increase of CCR7+ memory CD4+ T cells. Accordingly, the frequency of CCR4+CCR7+ memory CD4+ T cells correlated with asthma severity. Current common asthma therapeutics (including corticosteroids) were not able to affect the frequency of CCR4+CCR7+ memory CD4+ T cell subsets. Third, patients had an increase of Tregs, as assessed by measuring CD25, Foxp3, IL-10 and CTLA-4 expression. However, asthma severity was inversely correlated only with the frequency of CTLA-4+ CD4+ T cells. Lastly, patients and control subjects have similar frequencies of CD4+ T cells that express CCR5, CCR6, CXCR3, CXCR5, CD11a, or α4 integrin. However, the frequency of α4+ CD4+ T cells in patients correlated with asthma severity. CONCLUSIONS CCR4+CCR7+ memory, but not CCR4+CCR7- memory, α4+, and CTLA4+ CD4+ T cells in patients show significant clinical implications in atopic asthma. Current common therapeutics cannot alter the frequency of such CD4+ T cell subsets in adult atopic asthma patients.
Collapse
Affiliation(s)
- Matthew Wiest
- Baylor Institute for Immunology Research, 3434 Live Oak St., Dallas, TX 75204 USA
- Institute for Biomedical Studies, Baylor University, Waco, TX USA
| | - Katherine Upchurch
- Baylor Institute for Immunology Research, 3434 Live Oak St., Dallas, TX 75204 USA
- Institute for Biomedical Studies, Baylor University, Waco, TX USA
| | - Wenjie Yin
- Baylor Institute for Immunology Research, 3434 Live Oak St., Dallas, TX 75204 USA
- Institute for Biomedical Studies, Baylor University, Waco, TX USA
| | - Jerome Ellis
- Baylor Institute for Immunology Research, 3434 Live Oak St., Dallas, TX 75204 USA
| | - Yaming Xue
- Baylor Institute for Immunology Research, 3434 Live Oak St., Dallas, TX 75204 USA
| | | | - Mark Millard
- Martha Foster Lung Care Center, Baylor University Medical Center, Dallas, TX USA
| | - HyeMee Joo
- Baylor Institute for Immunology Research, 3434 Live Oak St., Dallas, TX 75204 USA
- Institute for Biomedical Studies, Baylor University, Waco, TX USA
| | - SangKon Oh
- Baylor Institute for Immunology Research, 3434 Live Oak St., Dallas, TX 75204 USA
- Institute for Biomedical Studies, Baylor University, Waco, TX USA
| |
Collapse
|
156
|
Cao J, Xu X, Zhang Y, Zeng Z, Hylkema MN, Huo X. Increased memory T cell populations in Pb-exposed children from an e-waste-recycling area. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 616-617:988-995. [PMID: 29096958 DOI: 10.1016/j.scitotenv.2017.10.220] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 10/21/2017] [Accepted: 10/21/2017] [Indexed: 02/05/2023]
Abstract
Chronic exposure to heavy metals could affect cell-mediated immunity. The aim of this study was to explore the status of memory T cell development in preschool children from an e-waste recycling area. Blood lead (Pb) levels, peripheral T cell subpopulations, and serum levels of cytokines (IL-2/IL-7/IL-15), relevant to generation and homeostasis of memory T cells were evaluated in preschool children from Guiyu (e-waste-exposed group) and Haojiang (reference group). The correlations between blood Pb levels and percentages of memory T cell subpopulations were also evaluated. Guiyu children had higher blood Pb levels and increased percentages of CD4+ central memory T cells and CD8+ central memory T cells than in the Haojiang group. Moreover, blood Pb levels were positively associated with the percentages of CD4+ central memory T cells. In contrast, Pb exposure contributed marginally in the change of percentages of CD8+ central memory T cells in children. There was no significant difference in the serum cytokine levels between the e-waste-exposed and reference children. Taken together, preschool children from an e-waste recycling area suffer from relatively higher levels of Pb exposure, which might facilitate the development of CD4+ central memory T cells in these children.
Collapse
Affiliation(s)
- Junjun Cao
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou 515041, Guangdong, China; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen 9713, GZ, The Netherlands; GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen 9713, GZ, The Netherlands
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou 515041, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, Guangdong, China.
| | - Yu Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou 515041, Guangdong, China; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen 9713, GZ, The Netherlands; GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen 9713, GZ, The Netherlands
| | - Zhijun Zeng
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou 515041, Guangdong, China; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen 9713, GZ, The Netherlands; GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen 9713, GZ, The Netherlands
| | - Machteld N Hylkema
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen 9713, GZ, The Netherlands; GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen 9713, GZ, The Netherlands
| | - Xia Huo
- School of Environment, Guangzhou Key Laboratory of Environmental Exposure and Health, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China.
| |
Collapse
|
157
|
Stephen B, Hajjar J. Overview of Basic Immunology and Translational Relevance for Clinical Investigators. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 995:1-41. [DOI: 10.1007/978-3-030-02505-2_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
158
|
Lee F, Lawrence DA. From Infections to Anthropogenic Inflicted Pathologies: Involvement of Immune Balance. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2017; 21:24-46. [PMID: 29252129 DOI: 10.1080/10937404.2017.1412212] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A temporal trend can be seen in recent human history where the dominant causes of death have shifted from infectious to chronic diseases in industrialized societies. Human influences in the current "Anthropocene" epoch are exponentially impacting the environment and consequentially health. Changing ecological niches are suggested to have created health transitions expressed as modifications of immune balance from infections inflicting pathologies in the Holocene epoch (12,000 years ago) to human behaviors inflicting pathologies beginning in the Anthropocene epoch (300 years ago). A review of human immune health and adaptations responding to environmental (biological, chemical, physical, and psychological) stresses, which are influenced by social conditions, emphasize the involvement of fluctuations in immune cell subsets affecting influential gene-environment interactions. The literature from a variety of fields (anthropological, immunological, and environmental) is incorporated to present an expanded perspective on shifts in diseases within the context of immune balance and function and environmental immunology. The influences between historical and contemporary human ecology are examined in relation to human immunity. Several examples of shifts in human physiology and immunity support the premise that increased incidences of chronic diseases are a consequence of human modification of environment and lifestyle. Although the development of better health care and a broader understanding of human health have helped with better life quality and expectancy, the transition of morbidity and mortality rates from infections to chronic diseases is a cause for concern. Combinations of environmental stressors/pollutants and human behaviors and conditions are modulating the immune-neuroendocrine network, which compromises health benefits.
Collapse
Affiliation(s)
- Florence Lee
- a Department of Anthropology , University at Albany , Albany , NY , USA
| | - David A Lawrence
- b Wadsworth Center/New York State Department of Health , Albany , NY , USA
- c Biomedical Sciences and Environmental Health Sciences , University at Albany, School of Public Health , Albany , NY , USA
| |
Collapse
|
159
|
Khan S, Telwatte S, Trapecar M, Yukl S, Sanjabi S. Differentiating Immune Cell Targets in Gut-Associated Lymphoid Tissue for HIV Cure. AIDS Res Hum Retroviruses 2017; 33:S40-S58. [PMID: 28882067 PMCID: PMC5685216 DOI: 10.1089/aid.2017.0153] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The single greatest challenge to an HIV cure is the persistence of latently infected cells containing inducible, replication-competent proviral genomes, which constitute only a small fraction of total or infected cells in the body. Although resting CD4+ T cells in the blood are a well-known source of viral rebound, more than 90% of the body's lymphocytes reside elsewhere. Many are in gut tissue, where HIV DNA levels per million CD4+ T cells are considerably higher than in the blood. Despite the significant contribution of gut tissue to viral replication and persistence, little is known about the cell types that support persistence of HIV in the gut; importantly, T cells in the gut have phenotypic, functional, and survival properties that are distinct from T cells in other tissues. The mechanisms by which latency is established and maintained will likely depend on the location and cytokine milieu surrounding the latently infected cells in each compartment. Therefore, successful HIV cure strategies require identification and characterization of the exact cell types that support viral persistence, particularly in the gut. In this review, we describe the seeding of the latent HIV reservoir in the gut mucosa; highlight the evidence for compartmentalization and depletion of T cells; summarize the immunologic consequences of HIV infection within the gut milieu; propose how the damaged gut environment may promote the latent HIV reservoir; and explore several immune cell targets in the gut and their place on the path toward HIV cure.
Collapse
Affiliation(s)
- Shahzada Khan
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California
| | - Sushama Telwatte
- San Francisco VA Health Care System and University of California, San Francisco (UCSF), San Francisco, California
| | - Martin Trapecar
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California
| | - Steven Yukl
- San Francisco VA Health Care System and University of California, San Francisco (UCSF), San Francisco, California
| | - Shomyseh Sanjabi
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
160
|
Krenzien F, Quante M, Heinbokel T, Seyda M, Minami K, Uehara H, Biefer HRC, Schuitenmaker JM, Gabardi S, Splith K, Schmelzle M, Petrides AK, Azuma H, Pratschke J, Li XC, ElKhal A, Tullius SG. Age-Dependent Metabolic and Immunosuppressive Effects of Tacrolimus. Am J Transplant 2017; 17:1242-1254. [PMID: 27754593 PMCID: PMC5395364 DOI: 10.1111/ajt.14087] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 09/15/2016] [Accepted: 10/11/2016] [Indexed: 01/25/2023]
Abstract
Immunosuppression in elderly recipients has been underappreciated in clinical trials. Here, we assessed age-specific effects of the calcineurin inhibitor tacrolimus (TAC) in a murine transplant model and assessed its clinical relevance on human T cells. Old recipient mice exhibited prolonged skin graft survival compared with young animals after TAC administration. More important, half of the TAC dose was sufficient in old mice to achieve comparable systemic trough levels. TAC administration was able to reduce proinflammatory interferon-γ cytokine production and promote interleukin-10 production in old CD4+ T cells. In addition, TAC administration decreased interleukin-2 secretion in old CD4+ T cells more effectively while inhibiting the proliferation of CD4+ T cells in old mice. Both TAC-treated murine and human CD4+ T cells demonstrated an age-specific suppression of intracellular calcineurin levels and Ca2+ influx, two critical pathways in T cell activation. Of note, depletion of CD8+ T cells did not alter allograft survival outcome in old TAC-treated mice, suggesting that TAC age-specific effects were mainly CD4+ T cell mediated. Collectively, our study demonstrates age-specific immunosuppressive capacities of TAC that are CD4+ T cell mediated. The suppression of calcineurin levels and Ca2+ influx in both old murine and human T cells emphasizes the clinical relevance of age-specific effects when using TAC.
Collapse
Affiliation(s)
- Felix Krenzien
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA,Department of Surgery, Campus Virchow-Klinikum and Campus Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Quante
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA,Department of General, Visceral and Transplant Surgery, Tuebingen Unversity Hospital, Tuebingen, Germany
| | - Timm Heinbokel
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA,Department of Nephrology, Charité Campus Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Midas Seyda
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA,Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Koichiro Minami
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA,Department of Urology, Osaka Medical College, Osaka, Japan
| | - Hirohito Uehara
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA,Department of Urology, Osaka Medical College, Osaka, Japan
| | - Hector Rodriguez Cetina Biefer
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA,Division of Cardiovascular Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Jeroen M. Schuitenmaker
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Steven Gabardi
- Renal and Pharmacy Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Katrin Splith
- Department of Surgery, Campus Virchow-Klinikum and Campus Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Moritz Schmelzle
- Department of Surgery, Campus Virchow-Klinikum and Campus Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Athena K. Petrides
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Haruhito Azuma
- Department of General, Visceral and Transplant Surgery, Tuebingen Unversity Hospital, Tuebingen, Germany
| | - Johann Pratschke
- Department of Surgery, Campus Virchow-Klinikum and Campus Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Xian C. Li
- Center for Immunobiology & Transplant Science, Houston Methodist Research Institute, Texas Medical Center, Houston, USA
| | - Abdallah ElKhal
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Stefan G. Tullius
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
161
|
Gustafson MP, DiCostanzo AC, Wheatley CM, Kim CH, Bornschlegl S, Gastineau DA, Johnson BD, Dietz AB. A systems biology approach to investigating the influence of exercise and fitness on the composition of leukocytes in peripheral blood. J Immunother Cancer 2017; 5:30. [PMID: 28428879 PMCID: PMC5394617 DOI: 10.1186/s40425-017-0231-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/10/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Exercise immunology has become a growing field in the past 20 years, with an emphasis on understanding how different forms of exercise affect immune function. Mechanistic studies are beginning to shed light on how exercise may impair the development of cancer or be used to augment cancer treatment. The beneficial effects of exercise on the immune system may be exploited to improve patient responses to cancer immunotherapy. METHODS We investigated the effects of acute exercise on the composition of peripheral blood leukocytes over time in a male population of varying fitness. Subjects performed a brief maximal intensity cycling regimen and a longer less intense cycling regimen at separate visits. Leukocytes were measured by multi-parameter flow cytometry of more than 50 immunophenotypes for each collection sample. RESULTS We found a differential induction of leukocytosis dependent on exercise intensity and duration. Cytotoxic natural killer cells demonstrated the greatest increase (average of 5.6 fold) immediately post-maximal exercise whereas CD15+ granulocytes demonstrated the largest increase at 3 h post-maximal exercise (1.6 fold). The longer, less intense endurance exercise resulted in an attenuated leukocytosis. Induction of leukocytosis did not differ in our limited study of active (n = 10) and sedentary (n = 5) subjects to exercise although we found that in baseline samples, sedentary individuals had elevated percentages of CD45RO+ memory CD4+ T cells and elevated proportions of CD4+ T cells expressing the negative immune regulator programmed death-1 (PD-1). Finally, we identified several leukocytes whose presence correlated with obesity related fitness parameters. CONCLUSIONS Our data suggests that leukocytes subsets are differentially mobilized into the peripheral blood and dependent on the intensity and duration of exercise. Pre-existing compositional differences of leukocytes were associated with various fitness parameters.
Collapse
Affiliation(s)
- Michael P Gustafson
- Human Cellular Therapy Laboratory, Department of Laboratory Medicine and Pathology, Division of Transfusion Medicine, Mayo Clinic, Rochester, MN USA.,Laboratory Medicine and Pathology, Mayo Clinic, Hilton 2-74B, Rochester, MN 55905 USA
| | | | | | - Chul-Ho Kim
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN USA
| | - Svetlana Bornschlegl
- Human Cellular Therapy Laboratory, Department of Laboratory Medicine and Pathology, Division of Transfusion Medicine, Mayo Clinic, Rochester, MN USA
| | - Dennis A Gastineau
- Human Cellular Therapy Laboratory, Department of Laboratory Medicine and Pathology, Division of Transfusion Medicine, Mayo Clinic, Rochester, MN USA
| | - Bruce D Johnson
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN USA
| | - Allan B Dietz
- Human Cellular Therapy Laboratory, Department of Laboratory Medicine and Pathology, Division of Transfusion Medicine, Mayo Clinic, Rochester, MN USA.,Department of Immunology, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
162
|
Abstract
Globally, as a leading agent of acute respiratory tract infections in children <5 years of age and the elderly, the human metapneumovirus (HMPV) has gained considerable attention. As inferred from studies comparing vaccinated and experimentally infected mice, the acquired immune response elicited by this pathogen fails to efficiently clear the virus from the airways, which leads to an exaggerated inflammatory response and lung damage. Furthermore, after disease resolution, there is a poor development of T and B cell immunological memory, which is believed to promote reinfections and viral spread in the community. In this article, we discuss the molecular mechanisms that shape the interactions of HMPV with host tissues that lead to pulmonary pathology and to the development of adaptive immunity that fails to protect against natural infections by this virus.
Collapse
|
163
|
Stephen B, Hajjar J. Overview of Basic Immunology for Clinical Investigators. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 995:1-31. [PMID: 28321810 DOI: 10.1007/978-3-319-53156-4_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Tumor exists as a complex network of structures with an ability to evolve and evade the host immune surveillance mechanism. The immune milieu which includes macrophages, dendritic cells, natural killer cells, neutrophils, mast cells, B cells, and T cells are found in the core, the invasive margin, or the adjacent stromal or lymphoid component of the tumor. The immune infiltrate is heterogeneous and varies within a patient and between patients of the same tumor histology. The location, density, functionality, and the cross talk between the immune cells in the tumor microenvironment influence the nature of immune response, prognosis, and treatment outcomes in cancer patients. Therefore, an understanding of the characteristics of the immune cells and their role in tumor immune surveillance is of paramount importance to identify immune targets and to develop novel immune therapeutics in the war against cancer. In this chapter, we provide an overview of the individual components of the human immune system and the translational relevance of predictive biomarkers.
Collapse
Affiliation(s)
- Bettzy Stephen
- The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Joud Hajjar
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
164
|
Esaki H, Brunner PM, Renert-Yuval Y, Czarnowicki T, Huynh T, Tran G, Lyon S, Rodriguez G, Immaneni S, Johnson DB, Bauer B, Fuentes-Duculan J, Zheng X, Peng X, Estrada YD, Xu H, de Guzman Strong C, Suárez-Fariñas M, Krueger JG, Paller AS, Guttman-Yassky E. Early-onset pediatric atopic dermatitis is T H 2 but also T H 17 polarized in skin. J Allergy Clin Immunol 2016; 138:1639-1651. [DOI: 10.1016/j.jaci.2016.07.013] [Citation(s) in RCA: 230] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/17/2016] [Accepted: 07/04/2016] [Indexed: 12/20/2022]
|
165
|
Siemeni T, Knöfel AK, Madrahimov N, Sommer W, Avsar M, Salman J, Ius F, Frank N, Büchler G, Jonigk D, Jansson K, Maus U, Tudorache I, Falk CS, Haverich A, Warnecke G. In Vivo Development of Transplant Arteriosclerosis in Humanized Mice Reflects Alloantigen Recognition and Peripheral Treg Phenotype of Lung Transplant Recipients. Am J Transplant 2016; 16:3150-3162. [PMID: 27273729 DOI: 10.1111/ajt.13905] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 05/24/2016] [Accepted: 05/27/2016] [Indexed: 01/25/2023]
Abstract
Experimentally, regulatory T cells inhibit rejection. In clinical transplantations, however, it is not known whether T cell regulation is the cause for, or an epiphenomenon of, long-term allograft survival. Here, we study naïve and alloantigen-primed T cell responses of clinical lung transplant recipients in humanized mice. The pericardiophrenic artery procured from human lung grafts was implanted into the aorta of NODrag-/- /IL-2rγc-/- mice reconstituted with peripheral blood mononuclear cells (PBMCs) from the respective lung recipient. Naïve or primed allogeneic PBMCs procured 21 days post-lung transplantation with or without enriching for CD4+ CD25high T cells were used. Transplant arteriosclerosis was assessed 28 days later by histology. Mice reconstituted with alloantigen-primed PBMCs showed significantly more severe transplant arteriosclerosis than did mice with naïve PBMCs (p = 0.005). Transplant arteriosclerosis was equally suppressed by enriching for autologous naïve (p = 0.012) or alloantigen-primed regulatory T cells (Tregs) (p = 0.009). Alloantigen priming in clinical lung recipients can be adoptively transferred into a humanized mouse model. Transplant arteriosclerosis elicited by naïve or alloantigen-primed PBMCs can be similarly controlled by potent autologous Tregs. Cellular therapy with expanded autologous Tregs in lung transplantation might be a promising future strategy.
Collapse
Affiliation(s)
- T Siemeni
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - A-K Knöfel
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany
| | - N Madrahimov
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - W Sommer
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany
| | - M Avsar
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - J Salman
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - F Ius
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - N Frank
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - G Büchler
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - D Jonigk
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - K Jansson
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - U Maus
- German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany.,Department of Experimental Pneumology, Hannover Medical School, Hannover, Germany
| | - I Tudorache
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - C S Falk
- Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany
| | - A Haverich
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany
| | - G Warnecke
- Division of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany. .,German Centre for Lung Research, BREATH Site, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
166
|
Johnstone J, Parsons R, Botelho F, Millar J, McNeil S, Fulop T, McElhaney JE, Andrew MK, Walter SD, Devereaux PJ, Malek M, Brinkman RR, Bramson J, Loeb M. T-Cell Phenotypes Predictive of Frailty and Mortality in Elderly Nursing Home Residents. J Am Geriatr Soc 2016; 65:153-159. [PMID: 27775813 DOI: 10.1111/jgs.14507] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To determine whether immune phenotypes associated with immunosenescence are predictive of frailty and mortality within 1-year in elderly nursing home residents. DESIGN Cross sectional study of frailty; prospective cohort study of mortality. SETTING Thirty-two nursing homes in four Canadian cities between September 2009 and October 2011. PARTICIPANTS Nursing home residents aged 65 and older (N = 1,072, median age 86, 72% female). MEASUREMENTS After enrollment, peripheral blood mononuclear cells were obtained and analyzed using flow cytometry for CD4+ and CD8+ T-cell subsets (naïve, memory (central, effector, terminally differentiated, senescent), and regulatory T-cells) and cytomegalovirus (CMV)-reactive CD4+ and CD8+ T-cells. Multilevel linear regression analysis was performed to determine the relationship between immune phenotypes and frailty; frailty was measured at the time of enrollment using the Frailty Index. A Cox proportional hazards model was used to determine the relationship between immune phenotypes and time to death (within 1 year). RESULTS Mean Frailty Index was 0.44 ± 0.13. Multilevel regression analysis showed that higher percentages of naïve CD4+ T-cells (P = .001) and effector memory CD8+ T-cells (P = .02) were associated with a lower mean Frailty Index, whereas a higher percentage of CD8+ central memory T-cells was associated with a higher mean Frailty Index score (P = .02). One hundred fifty one (14%) members of the cohort died within 1 year. Multivariable analysis showed a significant negative multiplicative interaction between age and percentage of CMV-reactive CD4+ T-cells (hazard ratio = 0.87, 95% confidence interval = 0.79-0.96). No other significant factors were identified. CONCLUSION Immune phenotypes found to be predictive of frailty and mortality in this study can help further understanding of immunosenescence and may provide a rationale for future intervention studies designed to modulate immunity.
Collapse
Affiliation(s)
- Jennie Johnstone
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada.,Public Health Ontario, Toronto, Ontario, Canada
| | - Robin Parsons
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Fernando Botelho
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Jamie Millar
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Shelly McNeil
- Canadian Center for Vaccinology, IWK Health Center and Capital Health, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Tamas Fulop
- Department of Medicine, Geriatrics Division, Research Center on Aging, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - Melissa K Andrew
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Stephen D Walter
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | - P J Devereaux
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Mehrnoush Malek
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Ryan R Brinkman
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan Bramson
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.,Department Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Mark Loeb
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Department Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
167
|
Luther N, Shahneh F, Brähler M, Krebs F, Jäckel S, Subramaniam S, Stanger C, Schönfelder T, Kleis-Fischer B, Reinhardt C, Probst HC, Wenzel P, Schäfer K, Becker C. Innate Effector-Memory T-Cell Activation Regulates Post-Thrombotic Vein Wall Inflammation and Thrombus Resolution. Circ Res 2016; 119:1286-1295. [PMID: 27707800 DOI: 10.1161/circresaha.116.309301] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/27/2016] [Accepted: 10/05/2016] [Indexed: 11/16/2022]
Abstract
RATIONALE Immune cells play an important role during the generation and resolution of thrombosis. T cells are powerful regulators of immune and nonimmune cell function, however, their role in sterile inflammation in venous thrombosis has not been systematically examined. OBJECTIVE This study investigated the recruitment, activation, and inflammatory activity of T cells in deep vein thrombosis and its consequences for venous thrombus resolution. METHODS AND RESULTS CD4+ and CD8+ T cells infiltrate the thrombus and vein wall rapidly on deep vein thrombosis induction and remain in the tissue throughout the thrombus resolution. In the vein wall, recruited T cells largely consist of effector-memory T (TEM) cells. Using T-cell receptor transgenic reporter mice, we demonstrate that deep vein thrombosis-recruited TEM receive an immediate antigen-independent activation and produce IFN-γ (interferon) in situ. Mapping inflammatory conditions in the thrombotic vein, we identify a set of deep vein thrombosis upregulated cytokines and chemokines that synergize to induce antigen-independent IFN-γ production in CD4+ and CD8+ TEM cells. Reducing the number of TEM cells through a depletion recovery procedure, we show that intravenous TEM activation determines neutrophil and monocyte recruitment and delays thrombus neovascularization and resolution. Examining T-cell recruitment in human venous stasis, we show that superficial varicose veins preferentially contain activated memory T cells. CONCLUSIONS TEM orchestrate the inflammatory response in venous thrombosis affecting thrombus resolution.
Collapse
Affiliation(s)
- Natascha Luther
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.)
| | - Fatemeh Shahneh
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.)
| | - Melanie Brähler
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.)
| | - Franziska Krebs
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.)
| | - Sven Jäckel
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.)
| | - Saravanan Subramaniam
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.)
| | - Christian Stanger
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.)
| | - Tanja Schönfelder
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.)
| | - Bettina Kleis-Fischer
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.)
| | - Christoph Reinhardt
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.)
| | - Hans Christian Probst
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.)
| | - Philip Wenzel
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.)
| | - Katrin Schäfer
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.)
| | - Christian Becker
- From the Department of Dermatology (N.L., F.S., M.B., F.K., C.S., B.K.-F., C.B.), Center for Thrombosis and Hemostasis (CTH) (S.J., S.S., T.S., C.R., P.W., C.B.), Institute for Immunology (H.C.P.), and Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany (P.W., K.S.).
| |
Collapse
|
168
|
The role of oxidative and inflammatory stress and persistent viral infections in immunosenescence. Mech Ageing Dev 2016; 158:27-37. [DOI: 10.1016/j.mad.2016.01.001] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/08/2015] [Accepted: 01/04/2016] [Indexed: 11/17/2022]
|
169
|
George RP, Mehta AK, Perez SD, Winterberg P, Cheeseman J, Johnson B, Kwun J, Monday S, Stempora L, Warshaw B, Kirk AD. Premature T Cell Senescence in Pediatric CKD. J Am Soc Nephrol 2016; 28:359-367. [PMID: 27413076 DOI: 10.1681/asn.2016010053] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 06/07/2016] [Indexed: 12/29/2022] Open
Abstract
An individual's immune function, susceptibility to infection, and response to immunosuppressive therapy are influenced in part by his/her T cell maturation state. Although childhood is the most dynamic period of immune maturation, scant information regarding the variability of T cell maturation in children with renal disease is available. In this study, we compared the T cell phenotype in children with renal failure (n=80) with that in healthy children (n=20) using multiparameter flow cytometry to detect markers of T cell maturation, exhaustion, and senescence known to influence immune function. We correlated data with the degree of renal failure (dialysis or nondialysis), prior immunosuppression use, and markers of inflammation (C-reactive protein and inflammatory cytokines) to assess the influence of these factors on T cell phenotype. Children with renal disease had highly variable and often markedly skewed maturation phenotypes, including CD4/CD8 ratio reversal, increased terminal effector differentiation in CD8+ T cells, reduction in the proportion of naïve T cells, evidence of T cell exhaustion and senescence, and variable loss of T cell CD28 expression. These findings were most significant in patients who had experienced major immune insults, particularly prior immunosuppressive drug exposure. In conclusion, children with renal disease have exceptional heterogeneity in the T cell repertoire. Cognizance of this heterogeneity might inform risk stratification with regard to the balance between infectious risk and response to immunosuppressive therapy, such as that required for autoimmune disease and transplantation.
Collapse
Affiliation(s)
- Roshan P George
- Division of Pediatric Nephrology, Department of Pediatrics, Children's Healthcare of Atlanta, Atlanta, Georgia; .,Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and
| | - Aneesh K Mehta
- Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and
| | - Sebastian D Perez
- Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and
| | - Pamela Winterberg
- Division of Pediatric Nephrology, Department of Pediatrics, Children's Healthcare of Atlanta, Atlanta, Georgia.,Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and
| | | | - Brandi Johnson
- Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and
| | - Jean Kwun
- Division of Surgery, Duke University, Durham, North Carolina
| | | | - Linda Stempora
- Division of Surgery, Duke University, Durham, North Carolina
| | - Barry Warshaw
- Division of Pediatric Nephrology, Department of Pediatrics, Children's Healthcare of Atlanta, Atlanta, Georgia.,Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and
| | - Allan D Kirk
- Division of Pediatric Nephrology, Department of Pediatrics, Children's Healthcare of Atlanta, Atlanta, Georgia.,Division of Pediatric Nephrology, Department of Pediatrics, Emory Transplant Center, Emory University, Atlanta, Georgia; and.,Division of Surgery, Duke University, Durham, North Carolina
| |
Collapse
|
170
|
Sinnott BD, Park B, Boer MC, Lewinsohn DA, Lancioni CL. Direct TLR-2 Costimulation Unmasks the Proinflammatory Potential of Neonatal CD4+ T Cells. THE JOURNAL OF IMMUNOLOGY 2016; 197:68-77. [PMID: 27194790 DOI: 10.4049/jimmunol.1501297] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 04/28/2016] [Indexed: 12/11/2022]
Abstract
Neonatal CD4(+) T cells have traditionally been viewed as deficient in their capacity to produce Th1 cytokines in response to polyclonal or Ag-specific stimuli. Thus, defining unique aspects of CD4(+) T cell activation and development into Th1 effector cells in neonates is essential to the successful development of novel vaccines and immunotherapies to protect infants from intracellular pathogens. Using highly purified naive CD4(+) T cells derived from cord and adult peripheral blood, we compared the impact of anti-CD3 stimulation plus costimulation through TLR-2 performed in the absence of APC on CD4(+) T cell cytokine production, proliferation, and expression of activation markers. In both age groups, TLR-2 costimulation elicited activation of naive CD4(+) T cells, characterized by robust production of IL-2 as well as key Th1-type cytokines IFN-γ and TNF-α. TLR-2 costimulation also dramatically reduced naive T cell production of the immunosuppressive cytokine IL-10. We observed that neonatal naive CD4(+) T cells are uniquely sensitive to TLR-2-mediated costimulation, which enabled them to produce equivalent amounts of IFN-γ and more IL-2 when compared with adult responses. Thus, neonatal CD4(+) T cells have a distinctive propensity to use TLR-2-mediated costimulation for development into proinflammatory Th1 effectors, and interventions that target CD4(+) T cell TLR-2-mediated responses may be exploited to enhance neonatal adaptive immunity.
Collapse
Affiliation(s)
- Brian D Sinnott
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239; and
| | - Byung Park
- Division of Biostatistics, Department of Public Health and Preventive Medicine, Oregon Health & Science University, Portland, OR 97239
| | - Mardi C Boer
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239; and
| | - Deborah A Lewinsohn
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239; and
| | - Christina L Lancioni
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239; and
| |
Collapse
|
171
|
Immune cell subsets at birth may help to predict risk of late-onset sepsis and necrotizing enterocolitis in preterm infants. Early Hum Dev 2016; 93:9-16. [PMID: 26704574 DOI: 10.1016/j.earlhumdev.2015.10.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 10/24/2015] [Accepted: 10/27/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND Parameters predicting late-onset sepsis (LOS) and necrotizing enterocolitis (NEC) in preterm infants would be valuable. Ten-color flow-cytometry enables the estimation of cellular immune status requiring only small sample volumes. AIMS Identifying predictive parameters for LOS and NEC in the cellular immune status of preterm infants. STUDY DESIGN AND SUBJECTS In this prospective study in 40 preterm infants (week 26+0 to 30+6) and 10 healthy full-term newborn infants (control group, week 37+0 to 40+6), flow cytometric analyses of lymphocyte subpopulations were performed between the 2nd and the 6th day of life, with a follow-up until the preterm infant reached the calculated gestational age of week 40. Patients' episodes of infections and NEC were analyzed according to the NEO-KISS criteria of the German National Reference Center. RESULTS Ten preterm infants showed events within the first week of life and were excluded from the analysis. Of the other 30, five developed NEC, twelve LOS. In patients with LOS, the proportion of double-negative (DN) T cells was significantly elevated compared to patients without LOS, while immune-regulatory CD56bright and CD56negCD16+ NK cells were significantly decreased (p<0.05). Patients with NEC showed a reduction in the NK cell proportion (<3.7%) and significantly decreased naïve cytotoxic CD45RA+CD62L+ T cells (p<0.05). CONCLUSION NK cells and DN-T cell counts within the first week of life may be predictors for NEC and LOS in preterm infants. In order to identify patients at risk early, further analysis of these populations might be of interest.
Collapse
|
172
|
Silva LCR, de Araújo AL, Fernandes JR, Matias MDST, Silva PR, Duarte AJS, Garcez Leme LE, Benard G. Moderate and intense exercise lifestyles attenuate the effects of aging on telomere length and the survival and composition of T cell subpopulations. AGE (DORDRECHT, NETHERLANDS) 2016; 38:24. [PMID: 26863877 PMCID: PMC5005879 DOI: 10.1007/s11357-016-9879-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/19/2016] [Indexed: 06/05/2023]
Abstract
Studies indicate that exercise might delay human biological aging, but the effects of long-term exercise on T cell function are not well known. We tested the hypothesis that moderate or intense exercise lifestyle may attenuate the effects of aging on the telomere length and the survival and composition of T cell subpopulations. Elderly (65-85 years) with intense training lifestyle (IT, n = 15), moderate training lifestyle (MT, n = 16), and who never trained (NT, n = 15) were studied. Although the three groups presented the age-associated contraction of the TCD4(+)/TCD8(+) naïve compartments and expansion of the memory compartments, both training modalities were associated with lower proportion of terminally differentiated (CD45RA(+)CCR7(neg)) TCD4(+) and TCD8(+) cells, although among the latter cells, the reduction reached statistical significance only with IT. MT was associated with higher proportion of central memory TCD4(+) cells, while IT was associated with higher proportion of effector memory TCD8(+) cells. However, both training lifestyles were unable to modify the proportion of senescent (CD28(neg)) TCD8(+) cells. Telomeres were longer in T cells in both training groups; with IT, telomere length increased mainly in TCD8(+) cells, whereas with MT, a modest increase in telomere length was observed in both TCD8(+) and TCD4(+) cells. Reduced commitment to apoptosis of resting T cells, as assessed by caspase-3 and Bcl-2 expression, was seen predominantly with IT. Measurement of pro-inflammatory cytokines in serum and peripheral blood mononuclear cell (PBMC)'s supernatants did not show chronic low-grade inflammation in any of the groups. In conclusion, MT and IT lifestyles attenuated some of the effects of aging on the immune system.
Collapse
Affiliation(s)
- Léia Cristina Rodrigues Silva
- Laboratory of Dermatology and Immunodeficiencies, Dermatology Division, Clinics Hospital, School of Medicine, USP, São Paulo, SP, Brazil
| | - Adriana Ladeira de Araújo
- Laboratory of Dermatology and Immunodeficiencies, Dermatology Division, Clinics Hospital, School of Medicine, USP, São Paulo, SP, Brazil
| | - Juliana Ruiz Fernandes
- Laboratory of Dermatology and Immunodeficiencies, Dermatology Division, Clinics Hospital, School of Medicine, USP, São Paulo, SP, Brazil
| | | | - Paulo Roberto Silva
- Laboratory of Movement Studies, Department of Orthopedics and Traumatology, School of Medicine, USP, São Paulo, SP, Brazil
| | - Alberto J S Duarte
- Laboratory of Dermatology and Immunodeficiencies, Dermatology Division, Clinics Hospital, School of Medicine, USP, São Paulo, SP, Brazil
| | | | - Gil Benard
- Laboratory of Dermatology and Immunodeficiencies, Dermatology Division, Clinics Hospital, School of Medicine, USP, São Paulo, SP, Brazil.
- Medical Mycology Laboratory, Institute of Tropical Medicine, University of São Paulo, São Paulo, Brazil.
- , R. Dr. Eneas de Carvalho Aguiar, 470, Cerqueira Cesar, São Paulo, CEP 05403-903, Brazil.
| |
Collapse
|
173
|
Abstract
Demographic changes are associated with a steady increase of older patients with end-stage organ failure in need for transplantation. As a result, the majority of transplant recipients are currently older than 50 years, and organs from elderly donors are more frequently used. Nevertheless, the benefit of transplantation in older patients is well recognized, whereas the most frequent causes of death among older recipients are potentially linked to side effects of their immunosuppressants.Immunosenescence is a physiological part of aging linked to higher rates of diabetes, bacterial infections, and malignancies representing the major causes of death in older patients. These age-related changes impact older transplant candidates and may have significant implications for an age-adapted immunosuppression. For instance, immunosenescence is linked to lower rates of acute rejections in older recipients, whereas the engraftment of older organs has been associated with higher rejection rates. Moreover, new-onset diabetes mellitus after transplantation is more frequent in the elderly, potentially related to corticosteroids, calcineurin inhibitors, and mechanistic target of rapamycin inhibitors.This review presents current knowledge for an age-adapted immunosuppression based on both, experimental and clinical studies in and beyond transplantation. Recommendations of maintenance and induction therapy may help to improve graft function and to design future clinical trials in the elderly.
Collapse
|
174
|
Nouatin O, Gbédandé K, Ibitokou S, Vianou B, Houngbegnon P, Ezinmegnon S, Borgella S, Akplogan C, Cottrell G, Varani S, Massougbodji A, Moutairou K, Troye-Blomberg M, Deloron P, Luty AJF, Fievet N. Infants' Peripheral Blood Lymphocyte Composition Reflects Both Maternal and Post-Natal Infection with Plasmodium falciparum. PLoS One 2015; 10:e0139606. [PMID: 26580401 PMCID: PMC4651557 DOI: 10.1371/journal.pone.0139606] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 09/14/2015] [Indexed: 11/19/2022] Open
Abstract
Maternal parasitoses modulate fetal immune development, manifesting as altered cellular immunological activity in cord blood that may be linked to enhanced susceptibility to infections in early life. Plasmodium falciparum typifies such infections, with distinct placental infection-related changes in cord blood exemplified by expanded populations of parasite antigen-specific regulatory T cells. Here we addressed whether such early-onset cellular immunological alterations persist through infancy. Specifically, in order to assess the potential impacts of P. falciparum infections either during pregnancy or during infancy, we quantified lymphocyte subsets in cord blood and in infants' peripheral blood during the first year of life. The principal age-related changes observed, independent of infection status, concerned decreases in the frequencies of CD4+, NKdim and NKT cells, whilst CD8+, Treg and Teff cells' frequencies increased from birth to 12 months of age. P. falciparum infections present at delivery, but not those earlier in gestation, were associated with increased frequencies of Treg and CD8+ T cells but fewer CD4+ and NKT cells during infancy, thus accentuating the observed age-related patterns. Overall, P. falciparum infections arising during infancy were associated with a reversal of the trends associated with maternal infection i.e. with more CD4+ cells, with fewer Treg and CD8+ cells. We conclude that maternal P. falciparum infection at delivery has significant and, in some cases, year-long effects on the composition of infants' peripheral blood lymphocyte populations. Those effects are superimposed on separate and independent age- as well as infant infection-related alterations that, respectively, either match or run counter to them.
Collapse
MESH Headings
- Adult
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Benin
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Female
- Fetal Blood/immunology
- Fetal Blood/parasitology
- Humans
- Immunophenotyping
- Infant
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Lymphocyte Count
- Malaria, Falciparum/immunology
- Malaria, Falciparum/parasitology
- Malaria, Falciparum/pathology
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/pathology
- Placenta/immunology
- Placenta/parasitology
- Placenta/pathology
- Plasmodium falciparum/immunology
- Pregnancy
- Pregnancy Complications, Parasitic/immunology
- Pregnancy Complications, Parasitic/parasitology
- Pregnancy Complications, Parasitic/pathology
- Retrospective Studies
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/pathology
Collapse
Affiliation(s)
- Odilon Nouatin
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Komi Gbédandé
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Samad Ibitokou
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Bertin Vianou
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Parfait Houngbegnon
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Sem Ezinmegnon
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Sophie Borgella
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Institut de Recherche pour le Développement, MERIT UMR D216 Mère et enfant face aux infections tropicales, Paris, France
| | - Carine Akplogan
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Gilles Cottrell
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Institut de Recherche pour le Développement, MERIT UMR D216 Mère et enfant face aux infections tropicales, Paris, France
- PRES Sorbonne Paris Cité, Université Paris Descartes, Faculté de Pharmacie, Paris, France
| | - Stefania Varani
- Unit of Microbiology, Department of Diagnostic, Experimental and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Achille Massougbodji
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
| | - Kabirou Moutairou
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou, Bénin
| | - Marita Troye-Blomberg
- Department of Molecular Biosciences, the Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Philippe Deloron
- Institut de Recherche pour le Développement, MERIT UMR D216 Mère et enfant face aux infections tropicales, Paris, France
- PRES Sorbonne Paris Cité, Université Paris Descartes, Faculté de Pharmacie, Paris, France
| | - Adrian J. F. Luty
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Nadine Fievet
- Centre d’Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l’Enfance (CERPAGE), Faculté des Sciences de la Santé, Université d’Abomey-Calavi, Cotonou, Benin
- Institut de Recherche pour le Développement, MERIT UMR D216 Mère et enfant face aux infections tropicales, Paris, France
- PRES Sorbonne Paris Cité, Université Paris Descartes, Faculté de Pharmacie, Paris, France
- * E-mail:
| |
Collapse
|
175
|
Immune Dysfunction in Children with CHARGE Syndrome: A Cross-Sectional Study. PLoS One 2015; 10:e0142350. [PMID: 26544072 PMCID: PMC4636349 DOI: 10.1371/journal.pone.0142350] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/19/2015] [Indexed: 12/31/2022] Open
Abstract
CHARGE syndrome is a variable, multiple congenital malformation syndrome. Patients with CHARGE syndrome have frequent infections that are presumed to be due to anatomical anomalies of the craniofacial region and upper airway, and cranial nerve problems resulting in swallowing difficulties and aspiration. The possible contribution of immunological abnormalities to these infections has not been systematically studied even though immune deficiencies have been described in patients with 22q11.2 deletion syndrome, a condition which shares remarkable clinical overlap with CHARGE syndrome. We assessed the frequency and nature of immune dysfunction in 24 children with genetically proven CHARGE syndrome. All patients, or their parents, completed a questionnaire on infectious history. Their immune system was extensively assessed through full blood counts, immunoglobulin levels, lymphocyte subpopulations, peripheral B- and T-cell differentiation, T-receptor excision circle (TREC) analysis, T-cell function, and vaccination responses. All CHARGE patients had a history of infections (often frequent), mainly otitis media and pneumonia, leading to frequent use of antibiotics and to hospital admissions. Decreased T-cell numbers were found in 12 (50%) patients, presumably caused by insufficient thymic output since TREC amounts were also diminished in CHARGE patients. Despite normal peripheral B-cell differentiation and immunoglobulin production in all patients, 83% of patients had insufficient antibody titers to one or more early childhood vaccinations. Based on our results, we recommend immunological evaluation of CHARGE patients with recurrent infections.
Collapse
|
176
|
Czarnowicki T, Esaki H, Gonzalez J, Malajian D, Shemer A, Noda S, Talasila S, Berry A, Gray J, Becker L, Estrada Y, Xu H, Zheng X, Suárez-Fariñas M, Krueger JG, Paller AS, Guttman-Yassky E. Early pediatric atopic dermatitis shows only a cutaneous lymphocyte antigen (CLA)(+) TH2/TH1 cell imbalance, whereas adults acquire CLA(+) TH22/TC22 cell subsets. J Allergy Clin Immunol 2015; 136:941-951.e3. [PMID: 26242300 PMCID: PMC4946641 DOI: 10.1016/j.jaci.2015.05.049] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/13/2015] [Accepted: 05/19/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Identifying differences and similarities between cutaneous lymphocyte antigen (CLA)(+) polarized T-cell subsets in children versus adults with atopic dermatitis (AD) is critical for directing new treatments toward children. OBJECTIVE We sought to compare activation markers and frequencies of skin-homing (CLA(+)) versus systemic (CLA(-)) "polar" CD4 and CD8 T-cell subsets in patients with early pediatric AD, adults with AD, and control subjects. METHODS Flow cytometry was used to measure CD69/inducible costimulator/HLA-DR frequency in memory cell subsets, as well as IFN-γ, IL-13, IL-9, IL-17, and IL-22 cytokines, defining TH1/cytotoxic T (TC) 1, TH2/TC2, TH9/TC9, TH17/TC17, and TH22/TC22 populations in CD4 and CD8 cells, respectively. We compared peripheral blood from 19 children less than 5 years old and 42 adults with well-characterized moderate-to-severe AD, as well as age-matched control subjects (17 children and 25 adults). RESULTS Selective inducible costimulator activation (P < .001) was seen in children. CLA(+) TH2 T cells were markedly expanded in both children and adults with AD compared with those in control subjects, but decreases in CLA(+) TH1 T-cell numbers were greater in children with AD (17% vs 7.4%, P = .007). Unlike in adults, no imbalances were detected in CLA(-) T cells from pediatric patients with AD nor were there altered frequencies of TH22 T cells within the CLA(+) or CLA(-) compartments. Adults with AD had increased frequencies of IL-22-producing CD4 and CD8 T cells within the skin-homing population, compared with controls (9.5% vs 4.5% and 8.6% vs 2.4%, respectively; P < .001), as well as increased HLA-DR activation (P < .01). CONCLUSIONS These data suggest that TH2 activation within skin-homing T cells might drive AD in children and that reduced counterregulation by TH1 T cells might contribute to excess TH2 activation. TH22 "spreading" of AD is not seen in young children and might be influenced by immune development, disease chronicity, or recurrent skin infections.
Collapse
Affiliation(s)
- Tali Czarnowicki
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Hitokazu Esaki
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Juana Gonzalez
- Translational Technology Core Laboratory, Rockefeller University, New York, NY
| | - Dana Malajian
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY; Columbia University College of Physicians and Surgeons, New York, NY
| | - Avner Shemer
- Department of Dermatology, Tel-Hashomer Hospital, Tel Aviv, Israel
| | - Shinji Noda
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Sreya Talasila
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Adam Berry
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Jayla Gray
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Lauren Becker
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Yeriel Estrada
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Hui Xu
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Xiuzhong Zheng
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Mayte Suárez-Fariñas
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY; Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - James G Krueger
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Amy S Paller
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Emma Guttman-Yassky
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
177
|
Biomolecular bases of the senescence process and cancer. A new approach to oncological treatment linked to ageing. Ageing Res Rev 2015; 23:125-38. [PMID: 25847820 DOI: 10.1016/j.arr.2015.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/30/2015] [Indexed: 01/07/2023]
Abstract
Human ageing is associated with a gradual decline in the physiological functions of the body at multiple levels and it is a key risk factor for many diseases, including cancer. Ageing process is intimately related to widespread cellular senescence, characterised by an irreversible loss of proliferative capacity and altered functioning associated with telomere attrition, accumulation of DNA damage and compromised mitochondrial and metabolic function. Tumour and senescent cells may be generated in response to the same stimuli, where either cellular senescence or transformation would constitute two opposite outcomes of the same degenerative process. This paper aims to review the state of knowledge on the biomolecular relationship between cellular senescence, ageing and cancer. Importantly, many of the cell signalling pathways that are found to be altered during both cellular senescence and tumourigenesis are regulated through shared epigenetic mechanisms and, therefore, they are potentially reversible. MicroRNAs are emerging as pivotal players linking ageing and cancer. These small RNA molecules have generated great interest from the point of view of future clinical therapy for cancer because successful experimental results have been obtained in animal models. Micro-RNA therapies for cancer are already being tested in clinical phase trials. These findings have potential importance in cancer treatment in aged people although further research-based knowledge is needed to convert them into an effective molecular therapies for cancer linked to ageing.
Collapse
|
178
|
Rozati M, Barnett J, Wu D, Handelman G, Saltzman E, Wilson T, Li L, Wang J, Marcos A, Ordovás JM, Lee YC, Meydani M, Meydani SN. Cardio-metabolic and immunological impacts of extra virgin olive oil consumption in overweight and obese older adults: a randomized controlled trial. Nutr Metab (Lond) 2015; 12:28. [PMID: 26251666 PMCID: PMC4527272 DOI: 10.1186/s12986-015-0022-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/21/2015] [Indexed: 12/13/2022] Open
Abstract
Background Both aging and obesity are related to dysregulated immune function, which may be responsible for increased risk of infection and also chronic non-infectious diseases. Dietary lipids have been shown to impact immune and inflammatory responses and cardio-metabolic risk factors. No information on the impact of olive oil on immune responses of overweight and obese older adults is available. Objective We aimed to determine the effect of replacing oils used in a typical American diet with extra virgin olive oil for 3 months on immune responses and cardio-metabolic risk factors in overweight and obese older adults. Methods This was a randomized, single-blinded and placebo-controlled trial in 41 overweight or obese participants (aged ≥ 65) who consumed a typical American diet. Participants in the control (CON, n = 21) group were provided with a mixture of corn, soybean oil and butter, and those in the olive oil (OO, n = 20) group, with extra virgin olive oil, to replace substitutable oils in their diet. At baseline and 3 months, we measured blood pressure, biochemical and immunological parameters using fasting blood, and delayed-type hypersensitivity (DTH) skin response. Results Compared to the CON group, the OO group showed decreased systolic blood pressure (P < 0.05), a strong trend toward increased plasma HDL-C concentrations (P = 0.06), and increased anti-CD3/anti-CD28 -stimulated T cell proliferation (P < 0.05). No differences were found in T cell phenotype, cytokine production, and DTH response between the two groups. Conclusions Our results indicate that substitution of oils used in a typical American diet with extra virgin olive oil in overweight and obese older adults may have cardio-metabolic and immunological health benefits. This trial was registered at clinicaltrials.gov as NCT01903304.
Collapse
Affiliation(s)
- Mitra Rozati
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111 USA ; Department of Health and Clinical Sciences, University of Massachusetts, Lowell 3 Solomont Way, Suite 4, Lowell, MA 01854 USA
| | - Junaidah Barnett
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111 USA
| | - Dayong Wu
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111 USA
| | - Garry Handelman
- Department of Health and Clinical Sciences, University of Massachusetts, Lowell 3 Solomont Way, Suite 4, Lowell, MA 01854 USA
| | - Edward Saltzman
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111 USA
| | - Thomas Wilson
- Department of Health and Clinical Sciences, University of Massachusetts, Lowell 3 Solomont Way, Suite 4, Lowell, MA 01854 USA
| | - Lijun Li
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111 USA
| | - Junpeng Wang
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111 USA
| | - Ascensión Marcos
- Institute of Food Science and Technology and Nutrition (ICTAN), Scientific National Research Council (CSIC), Madrid, Spain
| | - José M Ordovás
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111 USA
| | - Yu-Chi Lee
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111 USA
| | - Mohsen Meydani
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111 USA
| | - Simin Nikbin Meydani
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111 USA
| |
Collapse
|
179
|
Åkesson K, Tompa A, Rydén A, Faresjö M. Low expression of CD39(+) /CD45RA(+) on regulatory T cells (Treg ) cells in type 1 diabetic children in contrast to high expression of CD101(+) /CD129(+) on Treg cells in children with coeliac disease. Clin Exp Immunol 2015; 180:70-82. [PMID: 25421756 DOI: 10.1111/cei.12559] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2014] [Indexed: 12/19/2022] Open
Abstract
Type 1 diabetes (T1D) and coeliac disease are both characterized by an autoimmune feature. As T1D and coeliac disease share the same risk genes, patients risk subsequently developing the other disease. This study aimed to investigate the expression of T helper (Th), T cytotoxic (Tc) and regulatory T cells (Treg ) in T1D and/or coeliac disease children in comparison to healthy children. Subgroups of T cells (Th : CD4(+) or Tc : CD8(+) ); naive (CD27(+) CD28(+) CD45RA(+) CCR7(+) ), central memory (CD27(+) CD28(+) CD45RA(-) CCR7(+) ), effector memory (early differentiated; CD27(+) CD28(+) CD45RA(-) CCR7(-) and late differentiated; CD27(-) CD28(-) CD45RA(-) CCR7(-) ), terminally differentiated effector cells (TEMRA; CD27(-) CD28(-) CD45RA(+) CCR7(-) ) and Treg (CD4(+) CD25(+) FOXP3(+) CD127(-) ) cells, and their expression of CD39, CD45RA, CD101 and CD129, were studied by flow cytometry in T1D and/or coeliac disease children or without any of these diseases (reference group). Children diagnosed with both T1D and coeliac disease showed a higher percentage of TEMRA CD4(+) cells (P < 0·05), but lower percentages of both early and late effector memory CD8(+) cells (P < 0·05) compared to references. Children with exclusively T1D had lower median fluorescence intensity (MFI) of forkhead box protein 3 (FoxP3) (P < 0·05) and also a lower percentage of CD39(+) and CD45RA(+) within the Treg population (CD4(+) CD25(+) FOXP3(+) CD127(-) ) (P < 0·05). Children with exclusively coeliac disease had a higher MFI of CD101 (P < 0·01), as well as a higher percentage of CD129(+) (P < 0·05), in the CD4(+) CD25(hi) lymphocyte population, compared to references. In conclusion, children with combined T1D and coeliac disease have a higher percentage of differentiated CD4(+) cells compared to CD8(+) cells. T1D children show signs of low CD39(+) /CD45RA(+) Treg cells that may indicate loss of suppressive function. Conversely, children with coeliac disease show signs of CD101(+) /CD129(+) Treg cells that may indicate suppressor activity.
Collapse
Affiliation(s)
- K Åkesson
- Department of Pediatrics, Ryhov County Hospital, Jönköping, Sweden; Futurum - the Academy for Health and Care in Jönköping County Council, Jönköping, Sweden
| | | | | | | |
Collapse
|
180
|
Westera L, van Hoeven V, Drylewicz J, Spierenburg G, van Velzen JF, de Boer RJ, Tesselaar K, Borghans JAM. Lymphocyte maintenance during healthy aging requires no substantial alterations in cellular turnover. Aging Cell 2015; 14:219-27. [PMID: 25627171 PMCID: PMC4364834 DOI: 10.1111/acel.12311] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2014] [Indexed: 12/19/2022] Open
Abstract
In healthy humans, lymphocyte populations are maintained at a relatively constant size throughout life, reflecting a balance between lymphocyte production and loss. Given the profound immunological changes that occur during healthy aging, including a significant decline in T-cell production by the thymus, lymphocyte maintenance in the elderly is generally thought to require homeostatic alterations in lymphocyte dynamics. Surprisingly, using in vivo2H2O labeling, we find similar dynamics of most lymphocyte subsets between young adult and elderly healthy individuals. As the contribution of thymic output to T-cell production is only minor from young adulthood onward, compensatory increases in peripheral T-cell division rates are not required to maintain the T-cell pool, despite a tenfold decline in thymic output. These fundamental insights will aid the interpretation of further research into aging and clinical conditions related to disturbed lymphocyte dynamics.
Collapse
Affiliation(s)
- Liset Westera
- Laboratory of Translational Immunology; Department of Immunology; University Medical Center Utrecht; Lundlaan 6 3584 EA Utrecht The Netherlands
| | - Vera van Hoeven
- Laboratory of Translational Immunology; Department of Immunology; University Medical Center Utrecht; Lundlaan 6 3584 EA Utrecht The Netherlands
| | - Julia Drylewicz
- Laboratory of Translational Immunology; Department of Immunology; University Medical Center Utrecht; Lundlaan 6 3584 EA Utrecht The Netherlands
- Theoretical Biology and Bioinformatics; Department of Biology; Utrecht University; Padualaan 8 3584 CH Utrecht The Netherlands
| | - Gerrit Spierenburg
- Laboratory of Translational Immunology; Department of Immunology; University Medical Center Utrecht; Lundlaan 6 3584 EA Utrecht The Netherlands
| | - Jeroen F. van Velzen
- Laboratory of Translational Immunology; Department of Immunology; University Medical Center Utrecht; Lundlaan 6 3584 EA Utrecht The Netherlands
| | - Rob J. de Boer
- Theoretical Biology and Bioinformatics; Department of Biology; Utrecht University; Padualaan 8 3584 CH Utrecht The Netherlands
| | - Kiki Tesselaar
- Laboratory of Translational Immunology; Department of Immunology; University Medical Center Utrecht; Lundlaan 6 3584 EA Utrecht The Netherlands
| | - José A. M. Borghans
- Laboratory of Translational Immunology; Department of Immunology; University Medical Center Utrecht; Lundlaan 6 3584 EA Utrecht The Netherlands
| |
Collapse
|
181
|
Identification of pertussis-specific effector memory T cells in preschool children. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:561-9. [PMID: 25787136 DOI: 10.1128/cvi.00695-14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/11/2015] [Indexed: 02/07/2023]
Abstract
Whooping cough remains a problem despite vaccination, and worldwide resurgence of pertussis is evident. Since cellular immunity plays a role in long-term protection against pertussis, we studied pertussis-specific T-cell responses. Around the time of the preschool acellular pertussis (aP) booster dose at 4 years of age, T-cell memory responses were compared in children who were primed during infancy with either a whole-cell pertussis (wP) or an aP vaccine. Peripheral blood mononuclear cells (PBMCs) were isolated and stimulated with pertussis vaccine antigens for 5 days. T cells were characterized by flow-based analysis of carboxyfluorescein succinimidyl ester (CFSE) dilution and CD4, CD3, CD45RA, CCR7, gamma interferon (IFN-γ), and tumor necrosis factor alpha (TNF-α) expression. Before the aP preschool booster vaccination, both the proliferated pertussis toxin (PT)-specific CD4(+) and CD8(+) T-cell fractions (CFSE(dim)) were higher in aP- than in wP-primed children. Post-booster vaccination, more pertussis-specific CD4(+) effector memory cells (CD45RA(-) CCR7(-)) were induced in aP-primed children than in those primed with wP. The booster vaccination did not appear to significantly affect the T-cell memory subsets and functionality in aP-primed or wP-primed children. Although the percentages of Th1 cytokine-producing cells were alike in aP- and wP-primed children pre-booster vaccination, aP-primed children produced more Th1 cytokines due to higher numbers of proliferated pertussis-specific effector memory cells. At present, infant vaccinations with four aP vaccines in the first year of life result in pertussis-specific CD4(+) and CD8(+) effector memory T-cell responses that persist in children until 4 years of age and are higher than those in wP-primed children. The booster at 4 years of age is therefore questionable; this may be postponed to 6 years of age.
Collapse
|
182
|
Thome JJC, Yudanin N, Ohmura Y, Kubota M, Grinshpun B, Sathaliyawala T, Kato T, Lerner H, Shen Y, Farber DL. Spatial map of human T cell compartmentalization and maintenance over decades of life. Cell 2015; 159:814-28. [PMID: 25417158 DOI: 10.1016/j.cell.2014.10.026] [Citation(s) in RCA: 439] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 08/08/2014] [Accepted: 09/24/2014] [Indexed: 01/01/2023]
Abstract
Mechanisms for human memory T cell differentiation and maintenance have largely been inferred from studies of peripheral blood, though the majority of T cells are found in lymphoid and mucosal sites. We present here a multidimensional, quantitative analysis of human T cell compartmentalization and maintenance over six decades of life in blood, lymphoid, and mucosal tissues obtained from 56 individual organ donors. Our results reveal that the distribution and tissue residence of naive, central, and effector memory, and terminal effector subsets is contingent on both their differentiation state and tissue localization. Moreover, T cell homeostasis driven by cytokine or TCR-mediated signals is different in CD4+ or CD8+ T cell lineages, varies with their differentiation stage and tissue localization, and cannot be inferred from blood. Our data provide an unprecedented spatial and temporal map of human T cell compartmentalization and maintenance, supporting distinct pathways for human T cell fate determination and homeostasis.
Collapse
Affiliation(s)
- Joseph J C Thome
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Naomi Yudanin
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Yoshiaki Ohmura
- Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - Masaru Kubota
- Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - Boris Grinshpun
- Department of Systems Biology and Biomedical Informatics, and the JP Sulzberger Columbia Genome Center, Columbia University, New York, NY 10032, USA
| | - Taheri Sathaliyawala
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Tomoaki Kato
- Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA
| | - Harvey Lerner
- The New York Organ Donor Network (NYODN), New York, NY 10001, USA
| | - Yufeng Shen
- Department of Systems Biology and Biomedical Informatics, and the JP Sulzberger Columbia Genome Center, Columbia University, New York, NY 10032, USA
| | - Donna L Farber
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
183
|
Zeissig Y, Petersen BS, Milutinovic S, Bosse E, Mayr G, Peuker K, Hartwig J, Keller A, Kohl M, Laass MW, Billmann-Born S, Brandau H, Feller AC, Röcken C, Schrappe M, Rosenstiel P, Reed JC, Schreiber S, Franke A, Zeissig S. XIAP variants in male Crohn's disease. Gut 2015; 64:66-76. [PMID: 24572142 DOI: 10.1136/gutjnl-2013-306520] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The genetic basis of inflammatory bowel disease (IBD) is incompletely understood. The aim of this study was to identify rare genetic variants involved in the pathogenesis of IBD. DESIGN Exome sequencing and immunological profiling were performed in a patient with early onset Crohn's disease (CD). The coding region of the gene encoding X-linked inhibitor of apoptosis protein (XIAP) was sequenced in samples of 275 paediatric IBD and 1047 adult-onset CD patients. XIAP genotyping was performed in samples of 2680 IBD patients and 2864 healthy controls. Functional effects of the variants identified were investigated in primary cells and cultured cell lines. RESULTS Our results demonstrate the frequent occurrence of private variants in XIAP in about four percent of male patients with paediatric-onset CD. While XIAP mutations are known to be associated with the primary immunodeficiency (PID) X-linked lymphoproliferative disease type 2 (XLP2), CD patients described here exhibited intestinal inflammation in the absence of XLP2 and harboured a spectrum of mutations partially distinct from that observed in XLP2. The majority of XIAP variants identified was associated with a selective defect in NOD1/2 signalling, impaired NOD1/2-mediated activation of NF-κB, and altered NF-κB-dependent cytokine production. CONCLUSIONS This study reveals the unanticipated, frequent occurrence of XIAP variants in male paediatric-onset CD. The link between XIAP and NOD1/2, and the association of XIAP variants with XLP2, support the concept of PID in a subset of IBD patients. Moreover, these studies provide a rationale for the implementation of XIAP sequencing in clinical diagnostics in male patients with severe CD.
Collapse
Affiliation(s)
- Yvonne Zeissig
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany Department of General Pediatrics, University Medical Center Schleswig-Holstein, Kiel, Germany Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Britt-Sabina Petersen
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | | | - Esther Bosse
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Gabriele Mayr
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Kenneth Peuker
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Jelka Hartwig
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Andreas Keller
- Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Martina Kohl
- Department of General Pediatrics, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Martin W Laass
- Children's Hospital, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Susanne Billmann-Born
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Heide Brandau
- Department of General Pediatrics, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Alfred C Feller
- Institute of Pathology, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Christoph Röcken
- Institute of Pathology, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Martin Schrappe
- Department of General Pediatrics, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - John C Reed
- Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Stefan Schreiber
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Sebastian Zeissig
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
184
|
Serre-Miranda C, Roque S, Santos NC, Portugal-Nunes C, Costa P, Palha JA, Sousa N, Correia-Neves M. Effector memory CD4(+) T cells are associated with cognitive performance in a senior population. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2014; 2:e54. [PMID: 25566544 PMCID: PMC4277304 DOI: 10.1212/nxi.0000000000000054] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 11/18/2014] [Indexed: 01/24/2023]
Abstract
Objective: Immunosenescence and cognitive decline are common markers of the aging process. Taking into consideration the heterogeneity observed in aging processes and the recently described link between lymphocytes and cognition, we herein explored the possibility of an association between alterations in lymphocytic populations and cognitive performance. Methods: In a cohort of cognitively healthy adults (n = 114), previously characterized by diverse neurocognitive/psychological performance patterns, detailed peripheral blood immunophenotyping of both the innate and adaptive immune systems was performed by flow cytometry. Results: Better cognitive performance was associated with lower numbers of effector memory CD4+ T cells and higher numbers of naive CD8+ T cells and B cells. Furthermore, effector memory CD4+ T cells were found to be predictors of general and executive function and memory, even when factors known to influence cognitive performance in older individuals (e.g., age, sex, education, and mood) were taken into account. Conclusions: This is the first study in humans associating specific phenotypes of the immune system with distinct cognitive performance in healthy aging.
Collapse
Affiliation(s)
- Cláudia Serre-Miranda
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Susana Roque
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Nadine Correia Santos
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Carlos Portugal-Nunes
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Patrício Costa
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Joana Almeida Palha
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| |
Collapse
|
185
|
Johnstone J, Parsons R, Botelho F, Millar J, McNeil S, Fulop T, McElhaney J, Andrew MK, Walter SD, Devereaux PJ, Malekesmaeili M, Brinkman RR, Mahony J, Bramson J, Loeb M. Immune biomarkers predictive of respiratory viral infection in elderly nursing home residents. PLoS One 2014; 9:e108481. [PMID: 25275464 PMCID: PMC4183538 DOI: 10.1371/journal.pone.0108481] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/12/2014] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE To determine if immune phenotypes associated with immunosenescence predict risk of respiratory viral infection in elderly nursing home residents. METHODS Residents ≥ 65 years from 32 nursing homes in 4 Canadian cities were enrolled in Fall 2009, 2010 and 2011, and followed for one influenza season. Following influenza vaccination, peripheral blood mononuclear cells (PBMCs) were obtained and analysed by flow cytometry for T-regs, CD4+ and CD8+ T-cell subsets (CCR7+CD45RA+, CCR7-CD45RA+ and CD28-CD57+) and CMV-reactive CD4+ and CD8+ T-cells. Nasopharyngeal swabs were obtained and tested for viruses in symptomatic residents. A Cox proportional hazards model adjusted for age, sex and frailty, determined the relationship between immune phenotypes and time to viral infection. RESULTS 1072 residents were enrolled; median age 86 years and 72% female. 269 swabs were obtained, 87 were positive for virus: influenza (24%), RSV (14%), coronavirus (32%), rhinovirus (17%), human metapneumovirus (9%) and parainfluenza (5%). In multivariable analysis, high T-reg% (HR 0.41, 95% CI 0.20-0.81) and high CMV-reactive CD4+ T-cell% (HR 1.69, 95% CI 1.03-2.78) were predictive of respiratory viral infection. CONCLUSIONS In elderly nursing home residents, high CMV-reactive CD4+ T-cells were associated with an increased risk and high T-regs were associated with a reduced risk of respiratory viral infection.
Collapse
Affiliation(s)
- Jennie Johnstone
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | - Robin Parsons
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Fernando Botelho
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Jamie Millar
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Shelly McNeil
- Canadian Center for Vaccinology, IWK Health Centre and Capital Health, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Tamas Fulop
- Department of Medicine, Geriatrics Division, Research Center on Aging, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Janet McElhaney
- Department of Medicine, Northern Ontario School of Medicine, Sudbury, Ontario, Canada
| | - Melissa K. Andrew
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Stephen D. Walter
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | - P. J. Devereaux
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | - Ryan R. Brinkman
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - James Mahony
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan Bramson
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Mark Loeb
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
186
|
Moeller M, Hirose M, Mueller S, Roolf C, Baltrusch S, Ibrahim S, Junghanss C, Wolkenhauer O, Jaster R, Köhling R, Kunz M, Tiedge M, Schofield PN, Fuellen G. Inbred mouse strains reveal biomarkers that are pro-longevity, antilongevity or role switching. Aging Cell 2014; 13:729-38. [PMID: 24862908 PMCID: PMC4326954 DOI: 10.1111/acel.12226] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2014] [Indexed: 01/23/2023] Open
Abstract
Traditionally, biomarkers of aging are classified as either pro-longevity or antilongevity. Using longitudinal data sets from the large-scale inbred mouse strain study at the Jackson Laboratory Nathan Shock Center, we describe a protocol to identify two kinds of biomarkers: those with prognostic implication for lifespan and those with longitudinal evidence. Our protocol also identifies biomarkers for which, at first sight, there is conflicting evidence. Conflict resolution is possible by postulating a role switch. In these cases, high biomarker values are, for example, antilongevity in early life and pro-longevity in later life. Role-switching biomarkers correspond to features that must, for example, be minimized early, but maximized later, for optimal longevity. The clear-cut pro-longevity biomarkers we found reflect anti-inflammatory, anti-immunosenescent or anti-anaemic mechanisms, whereas clear-cut antilongevity biomarkers reflect inflammatory mechanisms. Many highly significant blood biomarkers relate to immune system features, indicating a shift from adaptive to innate processes, whereas most role-switching biomarkers relate to blood serum features and whole-body phenotypes. Our biomarker classification approach is applicable to any combination of longitudinal studies with life expectancy data, and it provides insights beyond a simplified scheme of biomarkers for long or short lifespan.
Collapse
Affiliation(s)
- Mark Moeller
- Institute for Biostatistics and Informatics in Medicine und Ageing Research Rostock University Medical Center Rostock Germany
| | - Misa Hirose
- Department of Dermatology University of Lübeck Lübeck Germany
| | - Sarah Mueller
- Division of Gastroenterology Department of Medicine II Rostock University Medical Center Rostock Germany
| | - Catrin Roolf
- Department of Hematology/Oncology/Palliative Medicine Rostock University Medical Center Rostock Germany
| | - Simone Baltrusch
- Institute of Medical Biochemistry and Molecular Biology Rostock University Medical Center Rostock Germany
| | - Saleh Ibrahim
- Department of Dermatology University of Lübeck Lübeck Germany
| | - Christian Junghanss
- Department of Hematology/Oncology/Palliative Medicine Rostock University Medical Center Rostock Germany
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics University of Rostock Rostock Germany
| | - Robert Jaster
- Division of Gastroenterology Department of Medicine II Rostock University Medical Center Rostock Germany
| | - Rüdiger Köhling
- Institute of Physiology Rostock University Medical Center Rostock Germany
| | - Manfred Kunz
- Department of Dermatology, Venereology and Allergology University of Leipzig Leipzig Germany
| | - Markus Tiedge
- Institute of Medical Biochemistry and Molecular Biology Rostock University Medical Center Rostock Germany
| | - Paul N. Schofield
- Department of Physiology, Development and Neuroscience University of Cambridge Cambridge UK
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine und Ageing Research Rostock University Medical Center Rostock Germany
| |
Collapse
|
187
|
Chen L, Duvvuri B, Grigull J, Jamnik R, Wither JE, Wu GE. Experimental evidence that mutated-self peptides derived from mitochondrial DNA somatic mutations have the potential to trigger autoimmunity. Hum Immunol 2014; 75:873-9. [PMID: 24979674 DOI: 10.1016/j.humimm.2014.06.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 06/19/2014] [Accepted: 06/19/2014] [Indexed: 12/20/2022]
Abstract
Autoimmune disease is a critical health concern, whose etiology remains enigmatic. We hypothesized that immune responses to somatically mutated self proteins could have a role in the development of autoimmune disease. IFN-γ secretion by T cells stimulated with mitochondrial peptides encoded by published mitochondrial DNA was monitored to test the hypothesis. Human peripheral blood mononuclear cells (PBMCs) of healthy controls and autoimmune patients were assessed for their responses to the self peptides and mutated-self peptides differing from self by one amino acid. None of the self peptides but some of the mutated-self peptides elicited an immune response in healthy controls. In some autoimmune patients, PBMCs responded not only to some of the mutated-self peptides, but also to some of the self peptides, suggesting that there is a breach of self-tolerance in these patients. Although PBMCs from healthy controls failed to respond to self peptides when stimulated with self, the mutated-self peptide could elicit a response to the self peptide upon re-stimulation in vitro, suggesting that priming with mutated-self peptides elicits a cross-reactive response with self. The data raise the possibility that DNA somatic mutations are one of the events that trigger and/or sustain T cell responses in autoimmune diseases.
Collapse
Affiliation(s)
- Lina Chen
- Department of Kinesiology and Health Science, York University, Canada.
| | - Bhargavi Duvvuri
- Department of Kinesiology and Health Science, York University, Canada
| | - Jörg Grigull
- Department of Mathematics and Statistics, York University, Canada
| | - Roni Jamnik
- Department of Kinesiology and Health Science, York University, Canada
| | - Joan E Wither
- University Health Network and University of Toronto, Canada
| | - Gillian E Wu
- Department of Kinesiology and Health Science, York University, Canada
| |
Collapse
|
188
|
Hu X, Kim H, Raj T, Brennan PJ, Trynka G, Teslovich N, Slowikowski K, Chen WM, Onengut S, Baecher-Allan C, De Jager PL, Rich SS, Stranger BE, Brenner MB, Raychaudhuri S. Regulation of gene expression in autoimmune disease loci and the genetic basis of proliferation in CD4+ effector memory T cells. PLoS Genet 2014; 10:e1004404. [PMID: 24968232 PMCID: PMC4072514 DOI: 10.1371/journal.pgen.1004404] [Citation(s) in RCA: 36] [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: 02/05/2014] [Accepted: 04/09/2014] [Indexed: 11/18/2022] Open
Abstract
Genome-wide association studies (GWAS) and subsequent dense-genotyping of associated loci identified over a hundred single-nucleotide polymorphism (SNP) variants associated with the risk of rheumatoid arthritis (RA), type 1 diabetes (T1D), and celiac disease (CeD). Immunological and genetic studies suggest a role for CD4-positive effector memory T (CD+ TEM) cells in the pathogenesis of these diseases. To elucidate mechanisms of autoimmune disease alleles, we investigated molecular phenotypes in CD4+ effector memory T cells potentially affected by these variants. In a cohort of genotyped healthy individuals, we isolated high purity CD4+ TEM cells from peripheral blood, then assayed relative abundance, proliferation upon T cell receptor (TCR) stimulation, and the transcription of 215 genes within disease loci before and after stimulation. We identified 46 genes regulated by cis-acting expression quantitative trait loci (eQTL), the majority of which we detected in stimulated cells. Eleven of the 46 genes with eQTLs were previously undetected in peripheral blood mononuclear cells. Of 96 risk alleles of RA, T1D, and/or CeD in densely genotyped loci, eleven overlapped cis-eQTLs, of which five alleles completely explained the respective signals. A non-coding variant, rs389862A, increased proliferative response (p = 4.75×10−8). In addition, baseline expression of seventeen genes in resting cells reliably predicted proliferative response after TCR stimulation. Strikingly, however, there was no evidence that risk alleles modulated CD4+ TEM abundance or proliferation. Our study underscores the power of examining molecular phenotypes in relevant cells and conditions for understanding pathogenic mechanisms of disease variants. Genome-wide association studies have identified hundreds of genetic variants associated to autoimmune diseases. To understand the mechanisms and pathways affected by these variants, follow-up studies of molecular phenotypes and functions are required. Given the diversity of cell types and specialization of functions within the immune system, it is crucial that such studies focus on specific and relevant cell types. Here, we studied genetic and cellular traits of CD4-positive effector memory T (CD4+ TEM) cells, which are particularly important in the onset of rheumatoid arthritis, celiac disease, and type 1 diabetes. In a cohort of healthy individuals, we purified CD4+ TEM cells, assayed genome-wide single nucleotide polymorphisms (SNPs), abundance of CD4+ TEM cells in blood, proliferation upon T cell receptor stimulation, and 215 gene transcripts in resting and stimulated states. We found that expression levels of 46 genes were regulated by nearby SNPs, including disease-associated SNPs. Many of these expression quantitative trait loci were not previously seen in studies of more heterogeneous peripheral blood cells. We demonstrated that relative abundance and proliferative response of CD4+ TEM cells varied in the population, however disease alleles are unlikely to confer risk by modulating these traits in this cell type.
Collapse
MESH Headings
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Autoimmune Diseases/genetics
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/pathology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- Celiac Disease/genetics
- Celiac Disease/metabolism
- Celiac Disease/pathology
- Cell Proliferation/genetics
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Gene Expression Regulation/genetics
- Gene Expression Regulation/immunology
- Genetic Predisposition to Disease
- Genome-Wide Association Study
- Genotype
- Humans
- Phenotype
- Polymorphism, Single Nucleotide/genetics
- Quantitative Trait Loci/genetics
- Receptors, Antigen, T-Cell/biosynthesis
- Receptors, Antigen, T-Cell/genetics
Collapse
Affiliation(s)
- Xinli Hu
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Partners Center for Personalized Genetic Medicine, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Harvard-MIT Division of Health Sciences and Technology, Boston, Massachusetts, United States of America
| | - Hyun Kim
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Partners Center for Personalized Genetic Medicine, Boston, Massachusetts, United States of America
| | - Towfique Raj
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Patrick J. Brennan
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Gosia Trynka
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Partners Center for Personalized Genetic Medicine, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Nikola Teslovich
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Kamil Slowikowski
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Partners Center for Personalized Genetic Medicine, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Wei-Min Chen
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Suna Onengut
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Clare Baecher-Allan
- Department of Dermatology/Harvard Skin Disease Research Center, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Philip L. De Jager
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Barbara E. Stranger
- Section of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, Illinois, United States of America
| | - Michael B. Brenner
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Soumya Raychaudhuri
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Partners Center for Personalized Genetic Medicine, Boston, Massachusetts, United States of America
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
189
|
Johnstone J, Millar J, Lelic A, Verschoor CP, Walter SD, Devereaux PJ, Bramson J, Loeb M. Immunosenescence in the nursing home elderly. BMC Geriatr 2014; 14:50. [PMID: 24742120 PMCID: PMC4013821 DOI: 10.1186/1471-2318-14-50] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 04/07/2014] [Indexed: 12/31/2022] Open
Abstract
Background To describe T-cell and natural killer (NK) cell phenotypes within nursing home elderly. Methods Nursing home elderly were recruited from four nursing homes in Hamilton, Ontario between September 2010 and December 2011. Healthy adults were recruited from McMaster University between September 2011 and December 2011. Nursing home elderly ≥65 years were eligible; those on immunosuppressive medications were excluded. Healthy adults ≥18-64 years were eligible. CD8+ and CD4+ T-cells% and their subsets, T-regs% and NK cell subset% were compared between the nursing home elderly and healthy adults. Results 262 nursing home elderly were enrolled; median age 87 years and 81% were female. 16 healthy adults were enrolled; median age 31 and 50% were female. There was no significant difference between CD8+ T-cell% in nursing home and healthy adults (median 17.1 versus 18.0, p = 0.56), however there were fewer naïve CD8 + T-cell% (median 0.9 versus 5.2, p < 0.001), more terminally differentiated CD8 + T-cell% (median 7.3 versus 4.1, p = 0.004) and more senescent T-cell% (median 5.3 versus 3.1, p = 0.04) in the nursing home elderly. There were more CD4+ T-cell% in the nursing home elderly compared to healthy adults (median 45.5 versus 37.1, p = 0.001). Nursing home elderly had a higher CD4+/CD8+ ratio than healthy adults (2.6 versus 1.9, p = 0.048), higher T-reg% (median 1.8 versus 0.8, p < 0.001) and increased mature NK cell% (median 12.1 versus 5.4, p = 0.001) compared to healthy adults. Conclusion Differences in naïve CD8+ T-cells, terminally differentiated and senescent CD8+ T-cells, T-regs and NK cell subsets were similar to studies involving community dwelling elderly. In contrast, the CD4+/CD8+ ratio was higher in nursing home elderly.
Collapse
Affiliation(s)
- Jennie Johnstone
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON L8S 4 K1, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
190
|
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: 652] [Impact Index Per Article: 59.3] [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
|
191
|
Nutrition, diet and immunosenescence. Mech Ageing Dev 2013; 136-137:116-28. [PMID: 24373813 DOI: 10.1016/j.mad.2013.12.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 12/05/2013] [Accepted: 12/16/2013] [Indexed: 01/10/2023]
Abstract
Ageing is characterized by immunosenescence and the progressive decline in immunity in association with an increased frequency of infections and chronic disease. This complex process affects both the innate and adaptive immune systems with a progressive decline in most immune cell populations and defects in activation resulting in loss of function. Although host genetics and environmental factors, such as stress, exercise and diet can impact on the onset or course of immunosenescence, the mechanisms involved are largely unknown. This review focusses on identifying the most significant aspects of immunosenescence and on the evidence that nutritional intervention might delay this process, and consequently improve the quality of life of the elderly.
Collapse
|
192
|
Reynolds J, Coles M, Lythe G, Molina-París C. Mathematical Model of Naive T Cell Division and Survival IL-7 Thresholds. Front Immunol 2013; 4:434. [PMID: 24391638 PMCID: PMC3870322 DOI: 10.3389/fimmu.2013.00434] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 11/22/2013] [Indexed: 11/18/2022] Open
Abstract
We develop a mathematical model of the peripheral naive T cell population to study the change in human naive T cell numbers from birth to adulthood, incorporating thymic output and the availability of interleukin-7 (IL-7). The model is formulated as three ordinary differential equations: two describe T cell numbers, in a resting state and progressing through the cell cycle. The third is introduced to describe changes in IL-7 availability. Thymic output is a decreasing function of time, representative of the thymic atrophy observed in aging humans. Each T cell is assumed to possess two interleukin-7 receptor (IL-7R) signaling thresholds: a survival threshold and a second, higher, proliferation threshold. If the IL-7R signaling strength is below its survival threshold, a cell may undergo apoptosis. When the signaling strength is above the survival threshold, but below the proliferation threshold, the cell survives but does not divide. Signaling strength above the proliferation threshold enables entry into cell cycle. Assuming that individual cell thresholds are log-normally distributed, we derive population-average rates for apoptosis and entry into cell cycle. We have analyzed the adiabatic change in homeostasis as thymic output decreases. With a parameter set representative of a healthy individual, the model predicts a unique equilibrium number of T cells. In a parameter range representative of persistent viral or bacterial infection, where naive T cell cycle progression is impaired, a decrease in thymic output may result in the collapse of the naive T cell repertoire.
Collapse
Affiliation(s)
- Joseph Reynolds
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds, UK
| | - Mark Coles
- Centre for Immunology and Infection, University of York, York, UK
| | - Grant Lythe
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds, UK
| | - Carmen Molina-París
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds, UK
| |
Collapse
|
193
|
Automatic Classification of Cellular Expression by Nonlinear Stochastic Embedding (ACCENSE). Proc Natl Acad Sci U S A 2013; 111:202-7. [PMID: 24344260 DOI: 10.1073/pnas.1321405111] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Mass cytometry enables an unprecedented number of parameters to be measured in individual cells at a high throughput, but the large dimensionality of the resulting data severely limits approaches relying on manual "gating." Clustering cells based on phenotypic similarity comes at a loss of single-cell resolution and often the number of subpopulations is unknown a priori. Here we describe ACCENSE, a tool that combines nonlinear dimensionality reduction with density-based partitioning, and displays multivariate cellular phenotypes on a 2D plot. We apply ACCENSE to 35-parameter mass cytometry data from CD8(+) T cells derived from specific pathogen-free and germ-free mice, and stratify cells into phenotypic subpopulations. Our results show significant heterogeneity within the known CD8(+) T-cell subpopulations, and of particular note is that we find a large novel subpopulation in both specific pathogen-free and germ-free mice that has not been described previously. This subpopulation possesses a phenotypic signature that is distinct from conventional naive and memory subpopulations when analyzed by ACCENSE, but is not distinguishable on a biaxial plot of standard markers. We are able to automatically identify cellular subpopulations based on all proteins analyzed, thus aiding the full utilization of powerful new single-cell technologies such as mass cytometry.
Collapse
|
194
|
Hsu DC, Kerr SJ, Iampornsin T, Pett SL, Avihingsanon A, Thongpaeng P, Zaunders JJ, Ubolyam S, Ananworanich J, Kelleher AD, Cooper DA. Restoration of CMV-specific-CD4 T cells with ART occurs early and is greater in those with more advanced immunodeficiency. PLoS One 2013; 8:e77479. [PMID: 24130889 PMCID: PMC3795037 DOI: 10.1371/journal.pone.0077479] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 09/02/2013] [Indexed: 12/21/2022] Open
Abstract
Objectives Restoration of Cytomegalovirus-specific-CD4 T cell (CMV-Sp-CD4) responses partly accounts for the reduction of CMV-disease with antiretroviral-therapy (ART), but CMV-Sp-CD4 may also drive immune activation and immunosenescence. This study characterized the dynamics of CMV-Sp-CD4 after ART initiation and explored associations with CD4 T cell recovery as well as frequency of naïve CD4 T cells at week 96. Methods Fifty HIV-infected, ART-naïve Thai adults with CD4 T cell count ≤350cells/µL and starting ART were evaluated over 96 weeks (ClinicalTrials.gov identifier NCT01296373). CMV-Sp-CD4 was detected by co-expression of CD25/CD134 by flow cytometry after CMV-antigen stimulation. Results All subjects were CMV sero-positive, 4 had quantifiable CMV-DNA (range 2.3-3.9 log10 copies/mL) at baseline but none had clinically apparent CMV-disease. Baseline CMV-Sp-CD4 response was positive in 40 subjects. Those with CD4 T cell count <100cells/µL were less likely to have positive baseline CMV-Sp-CD4 response (P=0.003). Positive baseline CMV-Sp-CD4 response was associated with reduced odds of quantifiable CMV-DNA (P=0.022). Mean CD4 T cell increase at week 96 was 213 cells/µL. This was associated positively with baseline HIV-VL (P=0.001) and negatively with age (P=0.003). The frequency of CMV-Sp-CD4 increased at week 4 (P=0.008), then declined. Those with lower baseline CMV-Sp-CD4 (P=0.009) or CDC category C (P<0.001) had greater increases in CMV-Sp-CD4 at week 4. At week 96, CD4 T cell count was positively (P<0.001) and the frequency of CMV-Sp-CD4 was negatively (P=0.001) associated with the percentage of naïve CD4 T cells. Conclusions Increases in CMV-Sp-CD4 with ART occurred early and were greater in those with more advanced immunodeficiency. The frequency of CMV-Sp-CD4 was associated with reduced naïve CD4 T cells, a marker associated with immunosenescence.
Collapse
Affiliation(s)
- Denise C. Hsu
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- * E-mail:
| | - Stephen J. Kerr
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Thatri Iampornsin
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Sarah L. Pett
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
- St Vincent’s Centre for Applied Medical Research, Sydney, Australia
| | - Anchalee Avihingsanon
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Parawee Thongpaeng
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - John J. Zaunders
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
- St Vincent’s Centre for Applied Medical Research, Sydney, Australia
| | - Sasiwimol Ubolyam
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Jintanat Ananworanich
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Anthony D. Kelleher
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
- St Vincent’s Centre for Applied Medical Research, Sydney, Australia
| | - David A. Cooper
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
- St Vincent’s Centre for Applied Medical Research, Sydney, Australia
| |
Collapse
|
195
|
Larbi A, Fulop T. From "truly naïve" to "exhausted senescent" T cells: when markers predict functionality. Cytometry A 2013; 85:25-35. [PMID: 24124072 DOI: 10.1002/cyto.a.22351] [Citation(s) in RCA: 233] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 05/27/2013] [Accepted: 07/30/2013] [Indexed: 02/06/2023]
Abstract
The study of T cell biology has been accelerated by substantial progress at the technological level, particularly through the continuing advancement of flow cytometry. The conventional approach of observing T cells as either T helper or T cytotoxic is overly simplistic and does not allow investigators to clearly identify immune mechanisms or alterations in physiological processes that impact on clinical outcomes. The complexity of T cell sub-populations, as we understand them today, combined with the immunological and functional diversity of these subsets represent significant complications for the study of T cell biology. In this article, we review the use of classical markers in delineating T cell sub-populations, from "truly naïve" T cells (recent thymic emigrants with no proliferative history) to "exhausted senescent" T cells (poorly proliferative cells that display severe functional abnormalities) wherein the different phenotypes of these populations reflect their disparate functionalities. In addition, since persistent infections and chronological aging have been shown to be associated with significant alterations in human T cell distribution and function, we also discuss age-associated and cytomegalovirus-driven alterations in the expression of key subset markers.
Collapse
Affiliation(s)
- Anis Larbi
- Singapore Immunology Network (SIgN), Biopolis, Agency for Science Technology and Research (A*STAR), Singapore
| | | |
Collapse
|
196
|
Fülöp T, Larbi A, Pawelec G. Human T cell aging and the impact of persistent viral infections. Front Immunol 2013; 4:271. [PMID: 24062739 PMCID: PMC3772506 DOI: 10.3389/fimmu.2013.00271] [Citation(s) in RCA: 230] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 08/27/2013] [Indexed: 12/30/2022] Open
Abstract
Aging is associated with a dysregulation of the immune response, loosely termed “immunosenescence.” Each part of the immune system is influenced to some extent by the aging process. However, adaptive immunity seems more extensively affected and among all participating cells it is the T cells that are most altered. There is a large body of experimental work devoted to the investigation of age-associated differences in T cell phenotypes and functions in young and old individuals, but few longitudinal studies in humans actually delineating changes at the level of the individual. In most studies, the number and proportion of late-differentiated T cells, especially CD8+ T cells, is reported to be higher in the elderly than in the young. Limited longitudinal studies suggest that accumulation of these cells is a dynamic process and does indeed represent an age-associated change. Accumulations of such late-stage cells may contribute to the enhanced systemic pro-inflammatory milieu commonly seen in older people. We do not know exactly what causes these observed changes, but an understanding of the possible causes is now beginning to emerge. A favored hypothesis is that these events are at least partly due to the effects of the maintenance of essential immune surveillance against persistent viral infections, notably Cytomegalovirus (CMV), which may exhaust the immune system over time. It is still a matter of debate as to whether these changes are compensatory and beneficial or pathological and detrimental to the proper functioning of the immune system and whether they impact longevity. Here, we will review present knowledge of T cell changes with aging and their relation to chronic viral and possibly other persistent infections.
Collapse
Affiliation(s)
- T Fülöp
- Geriatrics Division, Department of Medicine, Research Center on Aging, University of Sherbrooke , Sherbrooke, QC , Canada
| | | | | |
Collapse
|
197
|
Holly AC, Melzer D, Pilling LC, Fellows AC, Tanaka T, Ferrucci L, Harries LW. Changes in splicing factor expression are associated with advancing age in man. Mech Ageing Dev 2013; 134:356-66. [PMID: 23747814 PMCID: PMC5863542 DOI: 10.1016/j.mad.2013.05.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 05/16/2013] [Accepted: 05/25/2013] [Indexed: 11/22/2022]
Abstract
Human ageing is associated with decreased cellular plasticity and adaptability. Changes in alternative splicing with advancing age have been reported in man, which may arise from age-related alterations in splicing factor expression. We determined whether the mRNA expression of key splicing factors differed with age, by microarray analysis in blood from two human populations and by qRT-PCR in senescent primary fibroblasts and endothelial cells. Potential regulators of splicing factor expression were investigated by siRNA analysis. Approximately one third of splicing factors demonstrated age-related transcript expression changes in two human populations. Ataxia Telangiectasia Mutated (ATM) transcript expression correlated with splicing factor expression in human microarray data. Senescent primary fibroblasts and endothelial cells also demonstrated alterations in splicing factor expression, and changes in alternative splicing. Targeted knockdown of the ATM gene in primary fibroblasts resulted in up-regulation of some age-responsive splicing factor transcripts. We conclude that isoform ratios and splicing factor expression alters with age in vivo and in vitro, and that ATM may have an inhibitory role on the expression of some splicing factors. These findings suggest for the first time that ATM, a core element in the DNA damage response, is a key regulator of the splicing machinery in man.
Collapse
Affiliation(s)
- Alice C. Holly
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - David Melzer
- Epidemiology and Public Health, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Luke C. Pilling
- Epidemiology and Public Health, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Alexander C. Fellows
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | | | | | - Lorna W. Harries
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| |
Collapse
|
198
|
Human lymphocyte repertoires in ageing. Curr Opin Immunol 2013; 25:511-5. [PMID: 23992996 DOI: 10.1016/j.coi.2013.07.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 07/19/2013] [Accepted: 07/22/2013] [Indexed: 12/22/2022]
Abstract
Deterioration of adaptive immunity with ageing may reflect changes in the repertoire of T cells and B cells available to respond to antigenic challenges, due to altered proportions and absolute numbers of lymphocyte subpopulations as well as changes in the repertoire of antigen receptor genes expressed by these cells. High-throughput DNA sequencing (HTS) now facilitates examination of immunoglobulin and T cell receptor gene rearrangements, and initial studies using these methods to study immune system ageing in humans have demonstrated age-related alterations in the receptor populations within lymphocyte subsets, as well as in repertoires responding to vaccination. Accurate measurement of repertoire diversity remains an experimental challenge. Studies of larger numbers of human subjects, analysis of defined lymphocyte subpopulations including antigen-specific populations, and controlling for factors such as chronic viral infections, will be important for gaining additional understanding of the impact of ageing on human lymphocyte populations.
Collapse
|
199
|
Abstract
BACKGROUND Allogeneic bone marrow transplantation is under investigation for a range of nonmalignant indications, including tolerance induction through mixed chimerism. This strategy has so far been tested experimentally only in young recipients. Due to immunosenescence, older patients have an increase in memory T cells (TMEM) as well as other alterations to their immune system, which may influence the potential to induce tolerance. We therefore investigated the impact of immunosenescence on chimerism-based tolerance induction. METHODS Groups of young (2 months) and old (12 months) C57BL/6 recipients received BALB/c bone marrow under nonmyeloablative (3 Gy) and minimal (1 Gy) total body irradiation and treatment with costimulation blockade, T-cell depletion, or rapamycin. Multilineage chimerism, clonal deletion, and lymphocyte subsets were analyzed by flow cytometry. Tolerance was assessed by skin and heart grafts and enzyme-linked immunospot, intracellular cytokine, and mixed lymphocyte reaction assays. RESULTS Unexpectedly, chimerism and tolerance were established in old recipients with comparable-and in some cases increased-efficacy as in young recipients employing costimulation blockade-based or T-cell depletion-based conditioning with 1 or 3 Gy total body irradiation. TMEM reactivity in (naïve) old mice was augmented in response to polyclonal but not to allogeneic stimulation, providing a mechanistic underpinning for the susceptibility to chimerism induction despite increased TMEM frequencies. Tolerance in old recipients was associated with peripheral and central clonal deletion and a higher frequency of regulatory T cells. CONCLUSION Advanced age does not impair bone marrow engraftment, thereby widening the clinical potential of experimental protocols inducing transplantation tolerance through mixed chimerism.
Collapse
|
200
|
Scherpereel A, Grigoriu BD, Noppen M, Gey T, Chahine B, Baldacci S, Trauet J, Copin MC, Dessaint JP, Porte H, Labalette M. Defect in recruiting effector memory CD8+ T-cells in malignant pleural effusions compared to normal pleural fluid. BMC Cancer 2013; 13:324. [PMID: 23816056 PMCID: PMC3718618 DOI: 10.1186/1471-2407-13-324] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 06/28/2013] [Indexed: 02/06/2023] Open
Abstract
Background Malignant pleural effusions (MPE) are a common and fatal complication in cancers including lung or breast cancers, or malignant pleural mesothelioma (MPM). MPE animal models and immunotherapy trials in MPM patients previously suggested defects of the cellular immunity in MPE. However only few observational studies of the immune response were done in MPM patients, using questionable control groups (transudate…). Methods We compared T cell populations evaluated by flow cytometry from blood and pleural effusion of untreated patients with MPM (n = 58), pleural metastasis of adenocarcinoma (n = 30) or with benign pleural lesions associated with asbestos exposure (n = 23). Blood and pleural fluid were also obtained from healthy subjects, providing normal values for T cell populations. Results Blood CD4+ or CD8+ T cells percentages were similar in all groups of patients or healthy subjects. Whereas pleural fluid from healthy controls contained mainly CD8+ T cells, benign or malignant pleural effusions included mainly CD4+ T cells. Effector memory T cells were the main T cell subpopulation in pleural fluid from healthy subjects. In contrast, there was a striking and selective recruitment of central memory CD4+ T cells in MPE, but not of effector cells CD8+ T cells or NK cells in the pleural fluid as one would expect in order to obtain an efficient immune response. Conclusions Comparing for the first time MPE to pleural fluid from healthy subjects, we found a local defect in recruiting effector CD8+ T cells, which may be involved in the escape of tumor cells from immune response. Further studies are needed to characterize which subtypes of effector CD8+ T cells are involved, opening prospects for cell therapy in MPE and MPM.
Collapse
|