1
|
Shirafkan F, Hensel L, Rattay K. Immune tolerance and the prevention of autoimmune diseases essentially depend on thymic tissue homeostasis. Front Immunol 2024; 15:1339714. [PMID: 38571951 PMCID: PMC10987875 DOI: 10.3389/fimmu.2024.1339714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Abstract
The intricate balance of immune reactions towards invading pathogens and immune tolerance towards self is pivotal in preventing autoimmune diseases, with the thymus playing a central role in establishing and maintaining this equilibrium. The induction of central immune tolerance in the thymus involves the elimination of self-reactive T cells, a mechanism essential for averting autoimmunity. Disruption of the thymic T cell selection mechanisms can lead to the development of autoimmune diseases. In the dynamic microenvironment of the thymus, T cell migration and interactions with thymic stromal cells are critical for the selection processes that ensure self-tolerance. Thymic epithelial cells are particularly significant in this context, presenting self-antigens and inducing the negative selection of autoreactive T cells. Further, the synergistic roles of thymic fibroblasts, B cells, and dendritic cells in antigen presentation, selection and the development of regulatory T cells are pivotal in maintaining immune responses tightly regulated. This review article collates these insights, offering a comprehensive examination of the multifaceted role of thymic tissue homeostasis in the establishment of immune tolerance and its implications in the prevention of autoimmune diseases. Additionally, the developmental pathways of the thymus are explored, highlighting how genetic aberrations can disrupt thymic architecture and function, leading to autoimmune conditions. The impact of infections on immune tolerance is another critical area, with pathogens potentially triggering autoimmunity by altering thymic homeostasis. Overall, this review underscores the integral role of thymic tissue homeostasis in the prevention of autoimmune diseases, discussing insights into potential therapeutic strategies and examining putative avenues for future research on developing thymic-based therapies in treating and preventing autoimmune conditions.
Collapse
|
2
|
Dinges SS, Amini K, Notarangelo LD, Delmonte OM. Primary and secondary defects of the thymus. Immunol Rev 2024; 322:178-211. [PMID: 38228406 PMCID: PMC10950553 DOI: 10.1111/imr.13306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
The thymus is the primary site of T-cell development, enabling generation, and selection of a diverse repertoire of T cells that recognize non-self, whilst remaining tolerant to self- antigens. Severe congenital disorders of thymic development (athymia) can be fatal if left untreated due to infections, and thymic tissue implantation is the only cure. While newborn screening for severe combined immune deficiency has allowed improved detection at birth of congenital athymia, thymic disorders acquired later in life are still underrecognized and assessing the quality of thymic function in such conditions remains a challenge. The thymus is sensitive to injury elicited from a variety of endogenous and exogenous factors, and its self-renewal capacity decreases with age. Secondary and age-related forms of thymic dysfunction may lead to an increased risk of infections, malignancy, and autoimmunity. Promising results have been obtained in preclinical models and clinical trials upon administration of soluble factors promoting thymic regeneration, but to date no therapy is approved for clinical use. In this review we provide a background on thymus development, function, and age-related involution. We discuss disease mechanisms, diagnostic, and therapeutic approaches for primary and secondary thymic defects.
Collapse
Affiliation(s)
- Sarah S. Dinges
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kayla Amini
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ottavia M. Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
3
|
Sonar SA, Watanabe M, Nikolich JŽ. Disorganization of secondary lymphoid organs and dyscoordination of chemokine secretion as key contributors to immune aging. Semin Immunol 2023; 70:101835. [PMID: 37651849 PMCID: PMC10840697 DOI: 10.1016/j.smim.2023.101835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023]
Abstract
Aging is characterized by progressive loss of organ and tissue function, and the immune system is no exception to that inevitable principle. Of all the age-related changes in the body, reduction of the size of, and naïve T (Tn) cell output from, the thymus occurs earliest, being prominent already before or by the time of puberty. Therefore, to preserve immunity against new infections, over much of their lives, vertebrates dominantly rely on peripheral maintenance of the Tn cell pool in the secondary lymphoid organs (SLO). However, SLO structure and function subsequently also deteriorate with aging. Several recent studies have made a convincing case that this deterioration is of major importance to the erosion of protective immunity in the last third of life. Specifically, the SLO were found to accumulate multiple degenerative changes with aging. Importantly, the results from adoptive transfer and parabiosis studies teach us that the old microenvironment is the limiting factor for protective immunity in old mice. In this review, we discuss the extent, mechanisms, and potential role of stromal cell aging in the age-related alteration of T cell homeostatic maintenance and immune function decline. We use that discussion to frame the potential strategies to correct the SLO stromal aging defects - in the context of other immune rejuvenation approaches, - to improve functional immune responses and protective immunity in older adults.
Collapse
Affiliation(s)
- Sandip Ashok Sonar
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Makiko Watanabe
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Janko Ž Nikolich
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; the Aegis Consortium for Pandemic-free Future, University of Arizona Health Sciences, USA; BIO5 Institute, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
4
|
Cardinale A, De Luca CD, Locatelli F, Velardi E. Thymic Function and T-Cell Receptor Repertoire Diversity: Implications for Patient Response to Checkpoint Blockade Immunotherapy. Front Immunol 2021; 12:752042. [PMID: 34899700 PMCID: PMC8652142 DOI: 10.3389/fimmu.2021.752042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/05/2021] [Indexed: 01/05/2023] Open
Abstract
The capacity of T cells to recognize and mount an immune response against tumor antigens depends on the large diversity of the T-cell receptor (TCR) repertoire generated in the thymus during the process of T-cell development. However, this process is dramatically impaired by immunological insults, such as that caused by cytoreductive cancer therapies and infections, and by the physiological decline of thymic function with age. Defective thymic function and a skewed TCR repertoire can have significant clinical consequences. The presence of an adequate pool of T cells capable of recognizing specific tumor antigens is a prerequisite for the success of cancer immunotherapy using checkpoint blockade therapy. However, while this approach has improved the chances of survival of patients with different types of cancer, a large proportion of them do not respond. The limited response rate to checkpoint blockade therapy may be linked to a suboptimal TCR repertoire in cancer patients prior to therapy. Here, we focus on the role of the thymus in shaping the T-cell pool in health and disease, discuss how the TCR repertoire influences patients’ response to checkpoint blockade therapy and highlight approaches able to manipulate thymic function to enhance anti-tumor immunity.
Collapse
Affiliation(s)
- Antonella Cardinale
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | | | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy.,Department of Maternal and Child Health, Sapienza University of Rome, Rome, Italy
| | - Enrico Velardi
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
5
|
Ducloux D, Legendre M, Bamoulid J, Saas P, Courivaud C, Crepin T. End-Stage Renal Disease-Related Accelerated Immune Senescence: Is Rejuvenation of the Immune System a Therapeutic Goal? Front Med (Lausanne) 2021; 8:720402. [PMID: 34540869 PMCID: PMC8446427 DOI: 10.3389/fmed.2021.720402] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/17/2021] [Indexed: 02/05/2023] Open
Abstract
End-stage renal disease (ESRD) patients exhibit clinical features of premature ageing, including frailty, cardiovascular disease, and muscle wasting. Accelerated ageing also concerns the immune system. Patients with ESRD have both immune senescence and chronic inflammation that are resumed in the so-called inflammaging syndrome. Immune senescence is particularly characterised by premature loss of thymic function that is associated with hyporesponsiveness to vaccines, susceptibility to infections, and death. ESRD-related chronic inflammation has multiple causes and participates to accelerated cardiovascular disease. Although, both characterisation of immune senescence and its consequences are relatively well-known, mechanisms are more uncertain. However, prevention of immune senescence/inflammation or/and rejuvenation of the immune system are major goal to ameliorate clinical outcomes of ESRD patients.
Collapse
Affiliation(s)
- Didier Ducloux
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,University Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Mathieu Legendre
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,University Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France
| | - Jamal Bamoulid
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,University Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Philippe Saas
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,University Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,EFS Bourgogne Franche-Comté, Plateforme de Biomonitoring, CIC 1431/UMR1098, Besançon, France
| | - Cécile Courivaud
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,University Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Thomas Crepin
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| |
Collapse
|
6
|
Granadier D, Iovino L, Kinsella S, Dudakov JA. Dynamics of thymus function and T cell receptor repertoire breadth in health and disease. Semin Immunopathol 2021; 43:119-134. [PMID: 33608819 PMCID: PMC7894242 DOI: 10.1007/s00281-021-00840-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/12/2021] [Indexed: 12/26/2022]
Abstract
T cell recognition of unknown antigens relies on the tremendous diversity of the T cell receptor (TCR) repertoire; generation of which can only occur in the thymus. TCR repertoire breadth is thus critical for not only coordinating the adaptive response against pathogens but also for mounting a response against malignancies. However, thymic function is exquisitely sensitive to negative stimuli, which can come in the form of acute insult, such as that caused by stress, infection, or common cancer therapies; or chronic damage such as the progressive decline in thymic function with age. Whether it be prolonged T cell deficiency after hematopoietic cell transplantation (HCT) or constriction in the breadth of the peripheral TCR repertoire with age; these insults result in poor adaptive immune responses. In this review, we will discuss the importance of thymic function for generation of the TCR repertoire and how acute and chronic thymic damage influences immune health. We will also discuss methods that are used to measure thymic function in patients and strategies that have been developed to boost thymic function.
Collapse
Affiliation(s)
- David Granadier
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
- Department of Molecular and Cellular Biology, University of Washington, Seattle, WA, USA
| | - Lorenzo Iovino
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sinéad Kinsella
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jarrod A Dudakov
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Department of Immunology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
7
|
Abstract
Following periods of haematopoietic cell stress, such as after chemotherapy, radiotherapy, infection and transplantation, patient outcomes are linked to the degree of immune reconstitution, specifically of T cells. Delayed or defective recovery of the T cell pool has significant clinical consequences, including prolonged immunosuppression, poor vaccine responses and increased risks of infections and malignancies. Thus, strategies that restore thymic function and enhance T cell reconstitution can provide considerable benefit to individuals whose immune system has been decimated in various settings. In this Review, we focus on the causes and consequences of impaired adaptive immunity and discuss therapeutic strategies that can recover immune function, with a particular emphasis on approaches that can promote a diverse repertoire of T cells through de novo T cell formation.
Collapse
|
8
|
Coles AJ, Azzopardi L, Kousin-Ezewu O, Mullay HK, Thompson SA, Jarvis L, Davies J, Howlett S, Rainbow D, Babar J, Sadler TJ, Brown JWL, Needham E, May K, Georgieva ZG, Handel AE, Maio S, Deadman M, Rota I, Holländer G, Dawson S, Jayne D, Seggewiss-Bernhardt R, Douek DC, Isaacs JD, Jones JL. Keratinocyte growth factor impairs human thymic recovery from lymphopenia. JCI Insight 2019; 5:125377. [PMID: 31063156 PMCID: PMC6629095 DOI: 10.1172/jci.insight.125377] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND The lymphocyte-depleting antibody alemtuzumab is a highly effective treatment of relapsing-remitting multiple sclerosis (RRMS); however 50% of patients develop novel autoimmunity post-treatment. Most at risk are individuals who reconstitute their T-cell pool by proliferating residual cells, rather than producing new T-cells in the thymus; raising the possibility that autoimmunity might be prevented by increasing thymopoiesis. Keratinocyte growth factor (palifermin) promotes thymopoiesis in non-human primates. METHODS Following a dose-tolerability sub-study, individuals with RRMS (duration ≤10 years; expanded disability status scale ≤5·0; with ≥2 relapses in the previous 2 years) were randomised to placebo or 180mcg/kg/day palifermin, given for 3 days immediately prior to and after each cycle of alemtuzumab, with repeat doses at M1 and M3. The interim primary endpoint was naïve CD4+ T-cell count at M6. Exploratory endpoints included: number of recent thymic-emigrants (RTEs) and signal-joint T-cell receptor excision circles (sjTRECs)/mL of blood. The trial primary endpoint was incidence of autoimmunity at M30. FINDINGS At M6, individuals receiving palifermin had fewer naïve CD4+T-cells (2.229x107/L vs. 7.733x107/L; p=0.007), RTEs (16% vs. 34%) and sjTRECs/mL (1100 vs. 3396), leading to protocol-defined termination of recruitment. No difference was observed in the rate of autoimmunity between the two groupsConclusion: In contrast to animal studies, palifermin reduced thymopoiesis in our patients. These results offer a note of caution to those using palifermin to promote thymopoiesis in other settings, particularly in the oncology/haematology setting where alemtuzumab is often used as part of the conditioning regime. TRIAL REGISTRATION ClinicalTrials.gov NCT01712945Funding: MRC and Moulton Charitable Foundation.
Collapse
Affiliation(s)
- Alasdair J Coles
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Laura Azzopardi
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Onajite Kousin-Ezewu
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Harpreet Kaur Mullay
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Sara Aj Thompson
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Lorna Jarvis
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Jessica Davies
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Sarah Howlett
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Daniel Rainbow
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Judith Babar
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Timothy J Sadler
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - J William L Brown
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Edward Needham
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Karen May
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Zoya G Georgieva
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | | | - Stefano Maio
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Mary Deadman
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Ioanna Rota
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Georg Holländer
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Sarah Dawson
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom.,Medical Research Council (MRC) Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, United Kingdom
| | - David Jayne
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Ruth Seggewiss-Bernhardt
- University Hospital of Würzburg, Würzburg, Germany.,Department of Hematology/Oncology, Soziastiftung Bamberg, Bamberg, Germany
| | - Daniel C Douek
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - John D Isaacs
- Institute of Cellular Medicine, Newcastle University, and Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Joanne L Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
9
|
Thompson HL, Smithey MJ, Uhrlaub JL, Jeftić I, Jergović M, White SE, Currier N, Lang AM, Okoye A, Park B, Picker LJ, Surh CD, Nikolich-Žugich J. Lymph nodes as barriers to T-cell rejuvenation in aging mice and nonhuman primates. Aging Cell 2019; 18:e12865. [PMID: 30430748 PMCID: PMC6351843 DOI: 10.1111/acel.12865] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/04/2018] [Accepted: 09/27/2018] [Indexed: 01/16/2023] Open
Abstract
In youth, thymic involution curtails production of new naïve T cells, placing the onus of T-cell maintenance upon secondary lymphoid organs (SLO). This peripheral maintenance preserves the size of the T-cell pool for much of the lifespan, but wanes in the last third of life, leading to a dearth of naïve T cells in blood and SLO, and contributing to suboptimal immune defense. Both keratinocyte growth factor (KGF) and sex steroid ablation (SSA) have been shown to transiently increase the size and cellularity of the old thymus. It is less clear whether this increase can improve protection of old animals from infectious challenge. Here, we directly measured the extent to which thymic rejuvenation benefits the peripheral T-cell compartment of old mice and nonhuman primates. Following treatment of old animals with either KGF or SSA, we observed robust rejuvenation of thymic size and cellularity, and, in a reporter mouse model, an increase in recent thymic emigrants (RTE) in the blood. However, few RTE were found in the spleen and even fewer in the lymph nodes, and SSA-treated mice showed no improvement in immune defense against West Nile virus. In parallel, we found increased disorganization and fibrosis in old LN of both mice and nonhuman primates. These results suggest that SLO defects with aging can negate the effects of successful thymic rejuvenation in immune defense.
Collapse
Affiliation(s)
- Heather L. Thompson
- Department of Immunobiology; University of Arizona College of Medicine-Tucson; Tucson Arizona
- Arizona Center on Aging; University of Arizona College of Medicine-Tucson; Tucson Arizona
| | - Megan J. Smithey
- Department of Immunobiology; University of Arizona College of Medicine-Tucson; Tucson Arizona
- Arizona Center on Aging; University of Arizona College of Medicine-Tucson; Tucson Arizona
| | - Jennifer L. Uhrlaub
- Department of Immunobiology; University of Arizona College of Medicine-Tucson; Tucson Arizona
- Arizona Center on Aging; University of Arizona College of Medicine-Tucson; Tucson Arizona
| | - Ilija Jeftić
- Department of Immunobiology; University of Arizona College of Medicine-Tucson; Tucson Arizona
- Arizona Center on Aging; University of Arizona College of Medicine-Tucson; Tucson Arizona
| | - Mladen Jergović
- Department of Immunobiology; University of Arizona College of Medicine-Tucson; Tucson Arizona
- Arizona Center on Aging; University of Arizona College of Medicine-Tucson; Tucson Arizona
| | - Sarah E. White
- Department of Immunobiology; University of Arizona College of Medicine-Tucson; Tucson Arizona
- Arizona Center on Aging; University of Arizona College of Medicine-Tucson; Tucson Arizona
- Honors College; University of Arizona; Tucson Arizona
| | - Noreen Currier
- Vaccine and Gene Therapy Institute; Oregon Health and Science University; Beaverton Oregon
- Oregon National Primate Research Center; Beaverton Oregon
| | - Anna M. Lang
- Vaccine and Gene Therapy Institute; Oregon Health and Science University; Beaverton Oregon
- Oregon National Primate Research Center; Beaverton Oregon
| | - Afam Okoye
- Vaccine and Gene Therapy Institute; Oregon Health and Science University; Beaverton Oregon
- Oregon National Primate Research Center; Beaverton Oregon
| | - Byung Park
- Knight Cancer Center; Oregon Health and Science University; Portland Oregon
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute; Oregon Health and Science University; Beaverton Oregon
- Oregon National Primate Research Center; Beaverton Oregon
| | - Charles D. Surh
- Academy of Immunology and Microbiology; Institute for Basic Science; Pohang South Korea
- Department of Integrative Biosciences and Biotechnology; Pohang University of Science and Technology; Pohang South Korea
- Division of Developmental Immunology; La Jolla Institute for Allergy and Immunology; California
| | - Janko Nikolich-Žugich
- Department of Immunobiology; University of Arizona College of Medicine-Tucson; Tucson Arizona
- Arizona Center on Aging; University of Arizona College of Medicine-Tucson; Tucson Arizona
- Oregon National Primate Research Center; Beaverton Oregon
| |
Collapse
|
10
|
Fulop T, Franceschi C, Hirokawa K, Pawelec G. Nonhuman Primate Models of Immunosenescence. HANDBOOK OF IMMUNOSENESCENCE 2019. [PMCID: PMC7121907 DOI: 10.1007/978-3-319-99375-1_80] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Due to a dramatic increase in life expectancy, the number of individuals aged 65 and older is rapidly rising. This presents considerable challenges to our health care system since advanced age is associated with a higher susceptibility to infectious diseases due to immune senescence. However, the mechanisms underlying age-associated dysregulated immunity are still incompletely understood. Advancement in our comprehension of mechanisms of immune senescence and development of interventions to improve health span requires animal models that closely recapitulate the physiological changes that occur with aging in humans. Nonhuman primates (NHPs) are invaluable preclinical models to study the underlying causal mechanism of pathogenesis due to their outbred nature, high degree of genetic and physiological similarity to humans, and their susceptibility to human pathogens. In this chapter, we review NHP models available for biogerontology research, advantages and challenges they present, and advances they facilitated. Furthermore, we emphasize the utility of NHPs in characterizing immune senescence, evaluating interventions to reverse aging of the immune system, and development of vaccine strategies that are better suited for this vulnerable population.
Collapse
Affiliation(s)
- Tamas Fulop
- Division of Geriatrics Research Center on Aging, University of Sherbrooke Department of Medicine, Sherbrooke, QC Canada
| | - Claudio Franceschi
- Department of Experimental Pathology, University of Bologna, Bologna, Italy
| | | | - Graham Pawelec
- Center for Medical Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
11
|
Duggal NA. Reversing the immune ageing clock: lifestyle modifications and pharmacological interventions. Biogerontology 2018; 19:481-496. [PMID: 30269199 PMCID: PMC6223743 DOI: 10.1007/s10522-018-9771-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/16/2018] [Indexed: 12/20/2022]
Abstract
It is widely accepted that ageing is accompanied by remodelling of the immune system, including reduced numbers of naïve T cells, increased senescent or exhausted T cells, compromise to monocyte, neutrophil and natural killer cell function and an increase in systemic inflammation. In combination these changes result in increased risk of infection, reduced immune memory, reduced immune tolerance and immune surveillance, with significant impacts upon health in old age. More recently it has become clear that the rate of decline in the immune system is malleable and can be influenced by environmental factors such as physical activity as well as pharmacological interventions. This review discusses briefly our current understanding of immunesenescence and then focuses on lifestyle interventions and therapeutic strategies that have been shown to restore immune functioning in aged individuals.
Collapse
Affiliation(s)
- Niharika A Duggal
- MRC-Arthritis Research UK Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, Birmingham University, Birmingham, UK.
| |
Collapse
|
12
|
Aspinall R, Lang PO. Interventions to restore appropriate immune function in the elderly. IMMUNITY & AGEING 2018; 15:5. [PMID: 29416551 PMCID: PMC5785902 DOI: 10.1186/s12979-017-0111-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/28/2017] [Indexed: 01/03/2023]
Abstract
Advanced age is one indicator of likely immune dysfunction. As worldwide, the global population contains progressively more and more older individuals there is likelihood of an increased prevalence and incidence of infectious diseases due to common and emergent pathogens. The resultant increase in mortality and morbidity would be matched by the risk of functional decline and disability. Maintaining immune function at a plateau throughout life may therefore be associated with considerable cost savings. The aim of improving immune function in older individuals may be achieved through considering a therapeutic approach to rejuvenate, stimulate or support the indigenous immune system to perform in a more optimal manner. In terms of cost effectiveness a therapeutic approach may prove difficult because of issues associated with; identifying those who would benefit the most from this treatment, identifying the type of treatment which would suit them and identifying whether the treatment was successful. The alternative of supporting or providing a stronger stimulus through vaccination, whilst more cost effective, may be a more valuable option in the short term. Both approaches will be addressed in this review.
Collapse
Affiliation(s)
- Richard Aspinall
- Rivock Ltd, Bury St Edmunds, UK.,3Anglia Ruskin University, Cambridge, UK
| | - Pierre Olivier Lang
- 2Geriatric and Geriatric Rehabilitation Division, Department of Medicine, University Hospital of Lausanne, Lausanne, Switzerland.,3Anglia Ruskin University, Cambridge, UK
| |
Collapse
|
13
|
Wu C, Espinoza DA, Koelle SJ, Potter EL, Lu R, Li B, Yang D, Fan X, Donahue RE, Roederer M, Dunbar CE. Geographic clonal tracking in macaques provides insights into HSPC migration and differentiation. J Exp Med 2017; 215:217-232. [PMID: 29141868 PMCID: PMC5748860 DOI: 10.1084/jem.20171341] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/21/2017] [Accepted: 10/12/2017] [Indexed: 01/13/2023] Open
Abstract
Wu et al. use barcode tracking to uncover prolonged geographic bone marrow segregation of regenerating hematopoietic stem and progenitor cell clones after transplantation and provide evidence for local bone marrow production of T cells. The geographic distribution of hematopoiesis at a clonal level is of interest in understanding how hematopoietic stem and progenitor cells (HSPCs) and their progeny interact with bone marrow (BM) niches during regeneration. We tagged rhesus macaque autologous HSPCs with genetic barcodes, allowing clonal tracking over time and space after transplantation. We found marked geographic segregation of CD34+ HSPCs for at least 6 mo posttransplantation, followed by very gradual clonal mixing at different BM sites over subsequent months to years. Clonal mapping was used to document local production of granulocytes, monocytes, B cells, and CD56+ natural killer (NK) cells. In contrast, CD16+CD56− NK cells were not produced in the BM, and in fact were clonally distinct from multipotent progenitors producing all other lineages. Most surprisingly, we documented local BM production of CD3+ T cells early after transplantation, using both clonal mapping and intravascular versus tissue-resident T cell staining, suggesting a thymus-independent T cell developmental pathway operating during BM regeneration, perhaps before thymic recovery.
Collapse
Affiliation(s)
- Chuanfeng Wu
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Diego A Espinoza
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Samson J Koelle
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - E Lake Potter
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Rong Lu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Brian Li
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Di Yang
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD.,Institute of hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Fan
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Robert E Donahue
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Cynthia E Dunbar
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
14
|
Majumdar S, Nandi D. Thymic Atrophy: Experimental Studies and Therapeutic Interventions. Scand J Immunol 2017; 87:4-14. [PMID: 28960415 DOI: 10.1111/sji.12618] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 09/01/2017] [Accepted: 09/25/2017] [Indexed: 12/13/2022]
Abstract
The thymus is essential for T cell development and maturation. It is extremely sensitive to atrophy, wherein loss in cellularity of the thymus and/or disruption of the thymic architecture occur. This may lead to lower naïve T cell output and limited TCR diversity. Thymic atrophy is often associated with ageing. What is less appreciated is that proper functioning of the thymus is critical for reduction in morbidity and mortality associated with various clinical conditions including infections and transplantation. Therefore, therapeutic interventions which possess thymopoietic potential and lower thymic atrophy are required. These treatments enhance thymic output, which is a vital factor in generating favourable outcomes in clinical conditions. In this review, experimental studies on thymic atrophy in rodents and clinical cases where the thymus atrophies are discussed. In addition, mechanisms leading to thymic atrophy during ageing as well as during various stress conditions are reviewed. Therapies such as zinc supplementation, IL7 administration, leptin treatment, keratinocyte growth factor administration and sex steroid ablation during thymic atrophy involving experiments in animals and various clinical scenarios are reviewed. Interventions that have been used across different scenarios to reduce the extent of thymic atrophy and enhance its output are discussed. This review aims to speculate on the roles of combination therapies, which by acting additively or synergistically may further alleviate thymic atrophy and boost its function, thereby strengthening cellular T cell responses.
Collapse
Affiliation(s)
- S Majumdar
- Department of Biochemistry & Centre for Infectious Diseases Research, Indian Institute of Science, Bangalore, India
| | - D Nandi
- Department of Biochemistry & Centre for Infectious Diseases Research, Indian Institute of Science, Bangalore, India
| |
Collapse
|
15
|
Li Y, Xu L. Evaluation of TCR repertoire diversity in patients after hematopoietic stem cell transplantation. Stem Cell Investig 2015; 2:17. [PMID: 27358885 DOI: 10.3978/j.issn.2306-9759.2015.09.01] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 08/19/2015] [Indexed: 12/16/2022]
Abstract
T-cell receptor (TCR) repertoire analyses have been widely used to identify T cell populations of interest in cancer and autoimmunity and for characterizing immune repertoire reconstitution after hematopoietic stem cell transplantation (HSCT). Several decades of development and progress have led to the use of techniques for evaluating TCR repertoires in a more comprehensive, unbiased and fast manner, and the mechanisms of T cell immune reconstitution after HSCT and the new approaches used for recovering T cell repertoire diversity post HSCT have been more exhaustively documented to some degree. To better understand and characterize this progress, here we review recent studies on TCR repertoire diversity recovery in patients with leukemia and autoimmune disease who have received HSCT, impact factors and improvements in approaches for TCR repertoire recovery after HSCT.
Collapse
Affiliation(s)
- Yangqiu Li
- 1 Institute of Hematology, Medical College, 2 Department of Hematology, First Affiliated Hospital, 3 Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Ling Xu
- 1 Institute of Hematology, Medical College, 2 Department of Hematology, First Affiliated Hospital, 3 Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou 510632, China
| |
Collapse
|
16
|
A prospective study of incidence, clinical and quality of life consequences of oral mucositis post palifermin prophylaxis in patients undergoing high-dose chemotherapy and autologous hematopoietic cell transplantation. Ann Hematol 2015; 94:1733-40. [PMID: 26141369 DOI: 10.1007/s00277-015-2437-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 06/21/2015] [Indexed: 10/23/2022]
Abstract
Autologous hematopoietic cell transplantation (AHCT) has presented a revolutionary advance in the management of hematologic malignancies with low toxicity. However, oral mucositis (OM) remains a distressing toxic effect of AHCT and one of the major side effects of the conditioning. This prospective, observational study aimed to evaluate the severity of oral cavity pain and quality of life (QOL) and explore incidence, duration, and potential risk factors of moderate/severe OM. Thirty-nine patients receiving prophylactic palifermin post-AHCT were enrolled. QOL and severity of pain were assessed using validated questionnaires (Functional Assessment of Cancer Therapy-General (FACT-G) and mouth and throat soreness (MTS), respectively). The incidence of moderate/severe OM was 28.2 % with a median duration of 5 days and was associated with younger age and female gender. Severity of pain related to OM was generally low or moderate with only 25 % of patients reporting a score >6 on the MTS scale of 0-10 on day +7. Health-related QOL was worse on day +7 in the transplant unit compared to day 1, while on discharge day, all scores recovered and the total FACT-G score was not different from that on day 1. In our population, the incidence and duration of OM and the severity of pain related to OM appeared to be lower compared to that reported in previous studies. The impact of OM on QOL assessments seemed to be reversible with optimal supportive care despite the major transient disabilities mainly attributable to OM.
Collapse
|
17
|
Wu C, Li B, Lu R, Koelle SJ, Yang Y, Jares A, Krouse AE, Metzger M, Liang F, Loré K, Wu CO, Donahue RE, Chen ISY, Weissman I, Dunbar CE. Clonal tracking of rhesus macaque hematopoiesis highlights a distinct lineage origin for natural killer cells. Cell Stem Cell 2014; 14:486-499. [PMID: 24702997 DOI: 10.1016/j.stem.2014.01.020] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 12/09/2013] [Accepted: 01/30/2014] [Indexed: 01/15/2023]
Abstract
Analysis of hematopoietic stem cell function in nonhuman primates provides insights that are relevant for human biology and therapeutic strategies. In this study, we applied quantitative genetic barcoding to track the clonal output of transplanted autologous rhesus macaque hematopoietic stem and progenitor cells over a time period of up to 9.5 months. We found that unilineage short-term progenitors reconstituted myeloid and lymphoid lineages at 1 month but were supplanted over time by multilineage clones, initially myeloid restricted, then myeloid-B clones, and then stable myeloid-B-T multilineage, long-term repopulating clones. Surprisingly, reconstitution of the natural killer (NK) cell lineage, and particularly the major CD16(+)/CD56(-) peripheral blood NK compartment, showed limited clonal overlap with T, B, or myeloid lineages, and therefore appears to be ontologically distinct. Thus, in addition to providing insights into clonal behavior over time, our analysis suggests an unexpected paradigm for the relationship between NK cells and other hematopoietic lineages in primates.
Collapse
Affiliation(s)
- Chuanfeng Wu
- Hematology Branch; National Heart, Lung and Blood Institute; National Institutes of Health, Bethesda, MD 20892, USA
| | - Brian Li
- Hematology Branch; National Heart, Lung and Blood Institute; National Institutes of Health, Bethesda, MD 20892, USA
| | - Rong Lu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Samson J Koelle
- Hematology Branch; National Heart, Lung and Blood Institute; National Institutes of Health, Bethesda, MD 20892, USA
| | - Yanqin Yang
- DNA Sequencing and Genomics Core; National Heart, Lung and Blood Institute; National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander Jares
- Hematology Branch; National Heart, Lung and Blood Institute; National Institutes of Health, Bethesda, MD 20892, USA
| | - Alan E Krouse
- Hematology Branch; National Heart, Lung and Blood Institute; National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark Metzger
- Hematology Branch; National Heart, Lung and Blood Institute; National Institutes of Health, Bethesda, MD 20892, USA
| | - Frank Liang
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Karin Loré
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Colin O Wu
- Office of Biostatistics Research, National Heart, Lung and Blood Institute; National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert E Donahue
- Hematology Branch; National Heart, Lung and Blood Institute; National Institutes of Health, Bethesda, MD 20892, USA
| | - Irvin S Y Chen
- UCLA AIDS Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Irving Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Cynthia E Dunbar
- Hematology Branch; National Heart, Lung and Blood Institute; National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
A randomized controlled trial of palifermin (recombinant human keratinocyte growth factor) for the treatment of inadequate CD4+ T-lymphocyte recovery in patients with HIV-1 infection on antiretroviral therapy. J Acquir Immune Defic Syndr 2014; 66:399-406. [PMID: 24815851 DOI: 10.1097/qai.0000000000000195] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Poor CD4 lymphocyte recovery on antiretroviral therapy (ART) is associated with reduced function of the thymus. Palifermin (keratinocyte growth factor), by providing support to the thymic epithelium, promotes lymphopoiesis in animal models of bone marrow transplantation and graft-versus-host disease. METHODS In AIDS Clinical Trials Group A5212, a randomized, double-blind, placebo-controlled study, 99 HIV-infected patients on ART with plasma HIV-1 RNA levels ≤200 copies per milliliter for ≥6 months and CD4 lymphocyte counts <200 cells per cubic milliliter were randomized 1:1:1:1 to receive once daily intravenous administration of placebo or 20, 40, or 60 μg/kg of palifermin on 3 consecutive days. RESULTS The median change in the CD4 T-cell count from baseline to week 12 was not significantly different between the placebo arm [15 (-16, 23) cells/mm] and the 20-μg/kg dose [11 (2, 32) cells/mm], the 40-μg/kg dose [12 (-2, 25) cells/mm], or the 60-μg/kg dose arm [8 (-13, 35) cells/mm] of palifermin. No significant changes were observed in thymus size or in the number of naive T cells or recent thymic emigrants. CONCLUSIONS Palifermin in the doses studied was not effective in improving thymic function and did not raise CD4 lymphocyte counts in HIV-infected patients with low CD4 cell counts despite virologically effective ART.
Collapse
|
19
|
Finch PW, Mark Cross LJ, McAuley DF, Farrell CL. Palifermin for the protection and regeneration of epithelial tissues following injury: new findings in basic research and pre-clinical models. J Cell Mol Med 2014; 17:1065-87. [PMID: 24151975 PMCID: PMC4118166 DOI: 10.1111/jcmm.12091] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 05/06/2013] [Accepted: 05/15/2013] [Indexed: 02/06/2023] Open
Abstract
Keratinocyte growth factor (KGF) is a paracrine-acting epithelial mitogen produced by cells of mesenchymal origin, that plays an important role in protecting and repairing epithelial tissues. Pre-clinical data initially demonstrated that a recombinant truncated KGF (palifermin) could reduce gastrointestinal injury and mortality resulting from a variety of toxic exposures. Furthermore, the use of palifermin in patients with hematological malignancies reduced the incidence and duration of severe oral mucositis experienced after intensive chemoradiotherapy. Based upon these findings, as well as the observation that KGF receptors are expressed in many, if not all, epithelial tissues, pre-clinical studies have been conducted to determine the efficacy of palifermin in protecting different epithelial tissues from toxic injury in an attempt to model various clinical situations in which it might prove to be of benefit in limiting tissue damage. In this article, we review these studies to provide the pre-clinical background for clinical trials that are described in the accompanying article and the rationale for additional clinical applications of palifermin.
Collapse
|
20
|
Chidgey A. Effects of growth hormone in enhancing thymic regrowth and T-cell reconstitution. Expert Rev Clin Immunol 2014; 4:433-9. [DOI: 10.1586/1744666x.4.4.433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
21
|
Vadhan-Raj S, Goldberg JD, Perales MA, Berger DP, Brink MRM. Clinical applications of palifermin: amelioration of oral mucositis and other potential indications. J Cell Mol Med 2013; 17:1371-84. [PMID: 24251854 PMCID: PMC4117550 DOI: 10.1111/jcmm.12169] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 09/16/2013] [Indexed: 12/25/2022] Open
Abstract
Mucositis is one of the most significant toxicities in cancer patients undergoing cytotoxic treatment. It can have a negative impact on both quality of life and health economics. Severe oral mucositis can contribute to hospitalization, need for narcotic analgesics, total parentral nutrition, suboptimal delivery of anti-neoplastic treatment, and morbidity and mortality. Palifermin, a recombinant derivative of human keratinocyte growth factor, is the first active agent approved by the FDA for the prevention of severe oral mucositis in patients undergoing haematopoietic stem cell transplantation (HSCT). Several studies have also shown significant reduction in the incidence, severity and/or duration of oral mucositis in other high-risk settings such as concurrent chemoradiotherapy (CT/RT) for patients with head and neck cancer, and use of mucotoxic chemotherapeutic agents such as doxorubicin in sarcoma and fluorouracil for the treatment of colorectal cancer. The reduction in mucositis has translated into amelioration of symptoms and improvement in daily functioning as measured by patient-reported outcome in multiple studies. The clinical response to palifermin appears to be related in part to epithelial proliferation and mucosal thickening. Palifermin also has other potential clinical applications including the acceleration of immune reconstitution and inhibition of graft-versus-host disease in patients undergoing HSCT, and mitigation of dysphagia in lung cancer patients treated with concurrent CT/RT. Palifermin is generally well tolerated with mild-to-moderate skin and oral adverse events. Future studies may expand the use of palifermin into other areas that would benefit from its cytoprotective and regenerative effects.
Collapse
Affiliation(s)
- Saroj Vadhan-Raj
- Department of Sarcoma Medical Oncology, Section of Cytokines and Supportive Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer CenterHouston, TX, USA
| | - Jenna D Goldberg
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
- Weill Cornell Medical CollegeNew York, NY, USA
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
- Weill Cornell Medical CollegeNew York, NY, USA
| | - Dietmar P Berger
- Amgen Inc.Thousand Oaks, CA, USA
- Department of Oncology, Genentech Inc.South San Francisco, CA, USA
| | - Marcel RM Brink
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
- Weill Cornell Medical CollegeNew York, NY, USA
| |
Collapse
|
22
|
Boehm T, Swann JB. Thymus involution and regeneration: two sides of the same coin? Nat Rev Immunol 2013; 13:831-8. [DOI: 10.1038/nri3534] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
23
|
Velardi E, Dudakov JA, van den Brink MR. Clinical strategies to enhance thymic recovery after allogeneic hematopoietic stem cell transplantation. Immunol Lett 2013; 155:31-5. [PMID: 24120996 PMCID: PMC3871183 DOI: 10.1016/j.imlet.2013.09.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The thymus is particularly sensitive to injury caused by cytoreductive chemo- or radiation therapy, shock, infection and graft versus host disease. Insufficient thymic recovery has been directly correlated with increased risk of opportunistic infections and poor clinical outcomes in recipients of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Prolonged immune deficiency is particularly pronounced in older patients whose thymi are already significantly impaired due to age-related thymic involution. Preclinical and clinical studies have revealed several strategies that can enhance thymic function and immune reconstitution after transplant, including sex steroid ablation, growth factors (growth hormone, keratinocyte growth factor, insulin-like growth factor 1, interleukin-7) and ex vivo generated precursor T cells. In addition, recent studies have shown that other approaches, such as interleukein-22 and nutritional changes, may represent additional candidates to enhance thymic regeneration. In this review we provide updates on these strategies and comment on their potential to be translated into clinical therapies.
Collapse
Affiliation(s)
- Enrico Velardi
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
- Department of Clinical and Experimental Medicine, University of Perugia, 06122 Perugia, Italy
| | - Jarrod A. Dudakov
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
- Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Melbourne, Australia 3800
| | - Marcel R.M. van den Brink
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| |
Collapse
|
24
|
Yao YF, Zhao JJ, Dai QX, Li JY, Zhou L, Wang YT, Ni QY, Zhang MW, Xu HL. Identification and characterization of the major histocompatibility complex class II DQB (MhcMath-DQB1) alleles in Tibetan macaques (Macaca thibetana). ACTA ACUST UNITED AC 2013; 82:113-21. [PMID: 23745600 DOI: 10.1111/tan.12145] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 04/25/2013] [Accepted: 05/19/2013] [Indexed: 11/26/2022]
Abstract
Tibetan macaque (Macaca thibetana), an endangered primate species endemic to China, have been used as experimental animal model for various human diseases. Major histocompatibility complex (MHC) genes play a crucial role in the susceptibility and/or resistance to many human diseases, but little is known about Tibetan macaques. To gain an insight into the MHC background and to facilitate the experimental use of Tibetan macaques, the second exon of Mhc-DQB1 gene was sequenced in a cohort of wild Tibetan macaques living in the Sichuan province of China. A total of 23 MhcMath-DQB1 alleles were identified for the first time, illustrating a marked allelic polymorphism at the DQB1 locus for these macaques. Most of the sequences (74%) observed in this study belong to DQB1*06 (9 alleles) and DQB1*18 (8 alleles) lineages, and the rest (26%) belong to DQB1*15 (3 alleles) and DQB1*17 (3 alleles) lineages. The most frequent alleles detected among these macaques were MhcMath-DQB1*15:02:02 (17.9%), followed by Math-DQB1*06:06, 17:03 and 18:01, which were detected in 9 (16.1%) of the monkeys, respectively. Non-synonymous substitutions occurred at a significantly higher frequency than synonymous substitutions in the peptide-binding region, suggesting balancing selection for maintaining polymorphisms at the MHC class II DQB1 locus. Phylogenetic analyses confirms the trans-species model of evolution of the Mhc-DQB1 genes in non-human primates, and in particular, the extensive allele sharing is observed between Tibetan and other macaque species.
Collapse
Affiliation(s)
- Y-F Yao
- College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, China
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Liu W, Yao Y, Zhou L, Ni Q, Xu H. Evolutionary analysis of the short-type peptidoglycan-recognition protein gene (PGLYRP1) in primates. GENETICS AND MOLECULAR RESEARCH 2013; 12:453-62. [DOI: 10.4238/2013.february.8.10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
26
|
Meyer C, Kerns A, Haberthur K, Messaoudi I. Improving immunity in the elderly: current and future lessons from nonhuman primate models. AGE (DORDRECHT, NETHERLANDS) 2012; 34:1157-1168. [PMID: 22180097 PMCID: PMC3448983 DOI: 10.1007/s11357-011-9353-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 12/01/2011] [Indexed: 05/31/2023]
Abstract
The immune system must overcome daily challenges from pathogens to protect the body from infection. The success of the immune response to infection relies on the ability to sense and evaluate microbial threats and organize their elimination, while limiting damage to host tissues. This delicate balance is achieved through coordinated action of the innate and adaptive arms of the immune system. Aging results in several structural and functional changes in the immune system, often described under the umbrella term "immune senescence". Age-related changes affect both the innate and adaptive arms of the immune system and are believed to result in increased susceptibility and severity of infectious diseases, which is further exacerbated by reduced vaccine efficacy in the elderly. Therefore, multiple strategies to improve immune function in the aged are being investigated. Traditionally, studies on immune senescence are conducted using inbred specific pathogen free (SPF) rodents. This animal model has provided invaluable insight into the mechanisms of aging. However, the limited genetic heterogeneity and the SPF status of this model restrict the successful transfer of immunological discoveries between murine models and the clinical setting. More recently, nonhuman primates (NHPs) have emerged as a leading translational model to investigate immune senescence and to test interventions aimed at delaying/reversing age-related changes in immune function. In this article, we review and summarize advances in immuno-restorative approaches investigated in the NHP model system and discuss where the NHP model can support the development of novel therapeutics.
Collapse
Affiliation(s)
- Christine Meyer
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR USA
| | - Amelia Kerns
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR USA
| | - Kristen Haberthur
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR USA
- Graduate Program in Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR USA
| | - Ilhem Messaoudi
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR USA
- Graduate Program in Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR USA
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR USA
- Vaccine and Gene Therapy Institute, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006 USA
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Successful immune reconstitution is important for decreasing posthematopoietic cell transplant (post-HCT) infections, relapse, and secondary malignancy, without increasing graft-versus-host disease (GVHD). Here we review how different parts of the immune system recover, and the relationship between recovery and clinical outcomes. RECENT FINDINGS Innate immunity (e.g., neutrophils, natural killer cells) recovers within weeks, whereas adaptive immunity (B and T cells) recovers within months to years. This has been known for years; however, more recently, the pattern of recovery of additional immune cell subsets has been described. The role of these subsets in transplant complications like infections, GVHD and relapse is becoming increasingly recognized, as gleaned from studies of the association between subset counts or function and complications/outcomes, and from studies depleting or adoptively transferring various subsets. SUMMARY Lessons learned from observational studies on immune reconstitution are leading to new strategies to prevent or treat posttransplant infections. Additional knowledge is needed to develop effective strategies to prevent or treat relapse, second malignancies and GVHD.
Collapse
|
28
|
Raber-Durlacher JE, von Bültzingslöwen I, Logan RM, Bowen J, Al-Azri AR, Everaus H, Gerber E, Gomez JG, Pettersson BG, Soga Y, Spijkervet FKL, Tissing WJE, Epstein JB, Elad S, Lalla RV. Systematic review of cytokines and growth factors for the management of oral mucositis in cancer patients. Support Care Cancer 2012; 21:343-55. [PMID: 22987094 DOI: 10.1007/s00520-012-1594-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 08/30/2012] [Indexed: 11/30/2022]
Abstract
PURPOSE The aim of this project was to review the literature and define clinical practice guidelines for the use of cytokines and growth factor agents for the prevention or treatment of oral mucositis induced by cancer chemotherapy or radiotherapy. METHODS A systematic review was conducted by the Mucositis Study Group of the Multinational Association of Supportive Care in Cancer/International Society for Oral Oncology (MASCC/ISOO). The body of evidence for each intervention, in each cancer treatment setting, was assigned an evidence level. Based on the evidence level, one of the following three guideline determinations was possible: Recommendation, Suggestion, No guideline possible. RESULTS Sixty-four clinical studies across 11 interventions were evaluated. A recommendation was made for the use of recombinant human KGF-1 (palifermin) at a dose of 60 μg/kg per day for 3 days prior to conditioning treatment and for 3 days post-transplant for prevention of oral mucositis in patients receiving high-dose chemotherapy and total body irradiation followed by autologous stem cell transplantation for hematological malignancies. A suggestion was made against using granulocyte macrophage colony-stimulating factor mouthwash for the prevention of oral mucositis in the setting of high-dose chemotherapy followed by autologous or allogeneic stem cell transplantation. No guideline was possible for any other cytokine or growth factor agents due to inconclusive evidence. CONCLUSIONS Of the cytokine and growth factor agents studied for oral mucositis, the evidence only supports use of palifermin in the specific population listed above. Additional well-designed research is needed on other cytokine and growth factor interventions and in other cancer treatment settings.
Collapse
Affiliation(s)
- Judith E Raber-Durlacher
- Department of Periodontology ACTA (Academic Centre for Dentistry Amsterdam), University of Amsterdam and VU University Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Zeng M, Haase AT, Schacker TW. Lymphoid tissue structure and HIV-1 infection: life or death for T cells. Trends Immunol 2012; 33:306-14. [PMID: 22613276 DOI: 10.1016/j.it.2012.04.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 03/22/2012] [Accepted: 04/04/2012] [Indexed: 01/07/2023]
Abstract
Secondary lymphoid tissue (LT) structure facilitates immune responses and maintains homeostasis of T cells through production of survival factors, such as interleukin (IL)-7 that is 'posted' on the stromal fibroblastic reticular cell (FRC) network on which T cells traffic. Here, we examine the pathological changes that occur in LTs during HIV and simian immunodeficiency virus (SIV) infection. Immune activation leads to collagen deposition and loss of the FRC network itself. This decreases access to IL-7 and reduces the major source of IL-7, both of which deplete naïve T cells to limit immune reconstitution with antiretroviral treatment. We discuss the implications of LT structure damage for the timing of antiretroviral therapy and consider the development of adjunctive antifibrotic agents to improve immune reconstitution in HIV infection.
Collapse
Affiliation(s)
- Ming Zeng
- Department of Microbiology, Medical School, University of Minnesota, MMC 196, 420 Delaware Street S.E., Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
30
|
Srinivasan A, Kasow KA, Cross S, Parrish M, Wang C, Srivastava DK, Cai X, Panetta JC, Leung W. Phase I study of the tolerability and pharmacokinetics of palifermin in children undergoing allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2012; 18:1309-14. [PMID: 22542710 DOI: 10.1016/j.bbmt.2012.04.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 04/19/2012] [Indexed: 11/19/2022]
Abstract
The maximum tolerated dose of palifermin, a keratinocyte growth factor, in children is not known, and its pharmacokinetics in this population has not been well studied. This is a phase I study of palifermin was designed to evaluate its tolerability at doses of 40, 60, and 90 μg/kg/day in children age 2-18 years of age, receiving a myeloablative preparative regimen for allogeneic hematopoietic stem cell transplantation (HSCT). In each cohort, palifermin was given for 3 consecutive days before the preparative regimen and for 3 days after the stem cell infusion. Twelve patients were enrolled. Palifermin 90 μg/kg/day was tolerated in 6 patients without dose-limiting toxicity. All patients had at least 1 adverse event, mostly National Cancer Institute grade 1 or 2 severity. Skin rash, grade 2 or lower, was the most common adverse event, seen in 67% of patients. Only 3 patients (25%) had mucositis. The area under the concentration-time curve increased proportionally to the dose, and approximately 97% of palifermin exposure occurred in the first 24 hours after administration. Palifermin clearance increased linearly with body weight, supporting dosing by body weight. The mean clearance was 1893 mL/hour/kg, and it did not change significantly between administration of the first and last doses (P = .80). The mean elimination half-life was 4.6 hours. Our data show that palifermin was tolerated at a dose of 90 μg/kg/day, and exhibits linear pharmacokinetics in children undergoing allogeneic HSCT.
Collapse
Affiliation(s)
- Ashok Srinivasan
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Incomplete immune recovery in HIV infection: mechanisms, relevance for clinical care, and possible solutions. Clin Dev Immunol 2012; 2012:670957. [PMID: 22474480 PMCID: PMC3312328 DOI: 10.1155/2012/670957] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 12/22/2011] [Indexed: 12/31/2022]
Abstract
Treatment of HIV-infected patients with highly active antiretroviral therapy (HAART) usually results in diminished viral replication, increasing CD4+ cell counts, a reversal of most immunological disturbances, and a reduction in risk of morbidity and mortality. However, approximately 20% of all HIV-infected patients do not achieve optimal immune reconstitution despite suppression of viral replication. These patients are referred to as immunological nonresponders (INRs). INRs present with severely altered immunological functions, including malfunction and diminished production of cells within lymphopoetic tissue, perturbed frequencies of immune regulators such as regulatory T cells and Th17 cells, and increased immune activation, immunosenescence, and apoptosis. Importantly, INRs have an increased risk of morbidity and mortality compared to HIV-infected patients with an optimal immune reconstitution. Additional treatment to HAART that may improve immune reconstitution has been investigated, but results thus far have proved disappointing. The reason for immunological nonresponse is incompletely understood. This paper summarizes the known and unknown factors regarding the incomplete immune reconstitution in HIV infection, including mechanisms, relevance for clinical care, and possible solutions.
Collapse
|
32
|
Slatter MA, Cant AJ. Hematopoietic stem cell transplantation for primary immunodeficiency diseases. Ann N Y Acad Sci 2012; 1238:122-31. [PMID: 22129059 DOI: 10.1111/j.1749-6632.2011.06243.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) is now highly successfully curing a widening range of primary immunodeficiencies (PIDs). Better tissue typing, matching of donors, less toxic chemotherapy, better virus detection and treatment, improved supportive care, and graft-versus-host disease prophylaxis mean up to a 90% cure for severe combined immunodeficiency patients and a 70-80% cure for other PIDs given a matched unrelated donor, and rising to 95% for young patients with specific PIDs, such as Wiskott-Aldrich syndrome. Precise molecular diagnosis, detailed data on prognosis, and careful pre-HSCT assessment of infective lung and liver damage will ensure an informed benefit analysis of HSCT and the best outcome. It is now recognized that the best treatment option for chronic granulomatous disease is HSCT, which can also be curative for CD40 ligand deficiency and complex immune dysregulation disorders.
Collapse
Affiliation(s)
- Mary A Slatter
- Department of Paediatric Immunology, Newcastle upon Tyne Hospital NHS Foundation Trust, United Kingdom.
| | | |
Collapse
|
33
|
Keratinocyte Growth Factor and Stem Cell Factor to Improve Thymopoiesis after Autologous CD34+ Cell Transplantation in Rhesus Macaques. Biol Blood Marrow Transplant 2012; 18:55-65. [DOI: 10.1016/j.bbmt.2011.09.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 09/23/2011] [Indexed: 01/07/2023]
|
34
|
Rizwan R, Levine JE, Defor T, Ferarra JLM, Weisdorf DJ, Blazar BR, Verneris MR. Peritransplant palifermin use and lymphocyte recovery after T-cell replete, matched related allogeneic hematopoietic cell transplantation. Am J Hematol 2011; 86:879-82. [PMID: 21922528 DOI: 10.1002/ajh.22136] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Romee Rizwan
- The Blood and Marrow Transplantation Program, University of Minnesota, Minneapolis, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Khoury G, Rajasuriar R, Cameron PU, Lewin SR. The role of naïve T-cells in HIV-1 pathogenesis: an emerging key player. Clin Immunol 2011; 141:253-67. [PMID: 21996455 DOI: 10.1016/j.clim.2011.09.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 09/07/2011] [Accepted: 09/08/2011] [Indexed: 10/17/2022]
Abstract
Functional naïve T-cells are critical for an effective immune response to multiple pathogens. HIV leads to a significant reduction in CD4+ naïve T-cell number and impaired function and there is incomplete recovery following combination antiretroviral therapy (cART). Here we review the basic homeostatic mechanisms that maintain naïve CD4+ T-cells and discuss recent developments in understanding the impact of HIV infection on naïve CD4+ T-cells. Finally we review therapeutic interventions in HIV-infected individuals aimed at specifically enhancing recovery of naïve CD4+ T-cells.
Collapse
Affiliation(s)
- Gabriela Khoury
- Department of Medicine, Monash University, Melbourne Victoria, 3004, Australia
| | | | | | | |
Collapse
|
36
|
Liu JH, Liu JH, Dou LP, Dou LP, Wang LX, Wang LX, Wang LL, Wang LL, Zhou F, Yu L. α-GalCer administration after allogeneic bone marrow transplantation improves immune reconstitution in mice. ACTA ACUST UNITED AC 2011; 26:91-7. [PMID: 21703116 DOI: 10.1016/s1001-9294(11)60026-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To explore the effect of α- galactosyleramide( α-GalCer ) on immune reconstitution under acute graft-versus-host disease (aGVHD). METHODS BALB/c mice were transplanted wit hallogeneic C57BL/6 bone marrow cells and splenocytes (both 1×10(7))after receiving lethal total-body irradiation. α-GalCer (100 ug/kg) or vehicle (dimethylsulfoxide) was administered intraperitoneally immediately after transplantation. The effects of α-GalCer on immune reconstitution,proliferation of T cells and B cells, hematopoiesis,and thymic microenvironment were assessed. RESULTS The α-GalCer group exhibited higher percentages of CD3(+),CD4(+), CD8(+), B220(+), CD40(+), and CD86(+)cells compared with the vehicle group . The number of colony forming unit per 1000 CD34(+) cells in the α-GalCer group was higher than in the vehicle group ( P=0.0012).In vitro proliferation assays showed that the α-GalCer group had higher percentages of CD3(+), CD4(+), CD8(+),and B220(+) cells compared with the vehicle group. As for the results of in vivo proliferation assays, the numbers of CD3(+), CD4(+), CD8(+), and B220(+)cells were higher in the α-GalCer group than in the normal group ,especially the number of B220(+) cells ( P=0.007).Significant difference was not found in thymocyte count between the α-GalCer group and the vehicle group, nor in the percentages of CD3(+), CD4(+), and CD8(+) cells. CONCLUSION Administration of α-GalCer after allogeneic bone marrow transplantation may promote immune reconstitution in the presence of aGVHD.
Collapse
Affiliation(s)
- Jing-hua Liu
- Department of Hematology, General Hospital of the Chinese People's Liberation Army, Beijing 100853, [corrected] China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Mackall CL, Fry TJ, Gress RE. Harnessing the biology of IL-7 for therapeutic application. Nat Rev Immunol 2011; 11:330-42. [PMID: 21508983 DOI: 10.1038/nri2970] [Citation(s) in RCA: 426] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Interleukin-7 (IL-7) is required for T cell development and for maintaining and restoring homeostasis of mature T cells. IL-7 is a limiting resource under normal conditions, but it accumulates during lymphopaenia, leading to increased T cell proliferation. The administration of recombinant human IL-7 to normal or lymphopenic mice, non-human primates and humans results in widespread T cell proliferation, increased T cell numbers, modulation of peripheral T cell subsets and increased T cell receptor repertoire diversity. These effects raise the prospect that IL-7 could mediate therapeutic benefits in several clinical settings. This Review summarizes the biology of IL-7 and the results of its clinical use that are available so far to provide a perspective on the opportunities for clinical application of this cytokine.
Collapse
Affiliation(s)
- Crystal L Mackall
- Immunology Section, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
38
|
Wang Y, Chen G, Qiao S, Ma X, Tang X, Sun A, Wu D. Keratinocyte growth factor enhanced immune reconstitution in murine allogeneic umbilical cord blood cell transplant. Leuk Lymphoma 2011; 52:1556-66. [PMID: 21649545 DOI: 10.3109/10428194.2011.573037] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Umbilical cord blood (UCB) is used increasingly as a source of hematopoietic cells because of a lower risk of graft-versus-host disease (GVHD). Myeloablative conditioning before allogeneic umbilical cord blood transplant (allo-UCBT) results in thymic epithelial cell injury and T-cell immune deficiency. Full-term fetal blood cells were used as hematopoietic cells in a previous murine allo-UCBT model with a limited number of mice surviving the myeloablative conditioning. We designed a viable murine allo-UCBT protocol with platelet concentrate support. Keratinocyte growth factor (KGF) is a mitogen of thymic epithelial cells that promotes recovery of thymic epithelium when given before total body irradiation (TBI)-containing conditioning in experimental murine models. We hypothesized that KGF pre-administration would improve post-allo-UCBT thymopoiesis. To test this hypothesis, allo-UCBT recipient mice were given KGF or control saline prior to UCBT. Platelet concentrate support significantly improved the survival rate of murine allo-UCBT recipients. KGF administration significantly increased donor-derived T and natural killer T (NKT) cells at day +35 in spleens of allo-UCBT recipients. KGF administration also improved thymic function after allo-UCBT, resulting in higher copies of signal joint T-cell receptor rearrangement excision circles (sjTRECs) in splenocytes. Finally, we found that KGF pre-administration could enhance the graft-versus-leukemia effect. In conclusion, KGF can be administered safely to recipients of allo-UCBT to enhance T-cell immune reconstitution.
Collapse
Affiliation(s)
- Yi Wang
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, the First Affiliated Hospital of Soochow University, Suzhou, China
| | | | | | | | | | | | | |
Collapse
|
39
|
Lehrnbecher T, Tramsen L, Koehl U, Schmidt S, Bochennek K, Klingebiel T. Immunotherapy against invasive fungal diseases in stem cell transplant recipients. Immunol Invest 2011; 40:839-52. [PMID: 21627412 DOI: 10.3109/08820139.2011.581732] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Despite the availability of new antifungal compounds, morbidity and mortality of invasive fungal disease in allogeneic hematopoietic stem cell recipients are still unacceptably high. Over the past decade, one could witness an exciting improvement of the understanding of the molecular pathogenesis and of the complexity of host antifungal immune responses. This, in turn, provides critical information to augment host immunity against fungal pathogens. Strategies for enhancing the immune system include the administration of effector and regulatory cells (e.g., granulocytes, antigen-specific T cells, dendritic cells) as well as the administration of recombinant cytokines, interferons and growth factors (e.g., interferon-γ, keratinocyte growth factor, granulocyte- and granulocyte-macrophage colony stimulating factor). One has to recognize at the same time, however, that data of in vitro assays and animal models cannot necessarily be transferred into the clinical setting. In addition, meaningful clinical trials in allogeneic stem cell recipients suffering from invasive fungal disease require sufficiently large and homogenous cohorts of patients and can only be performed in international collaboration, but may ultimately improve the outcome of allogeneic transplant recipients with invasive fungal disease.
Collapse
Affiliation(s)
- Thomas Lehrnbecher
- Pediatric Hematology and Oncology, Children's Hospital III, Johann Wolfgang Goethe University, Theodor-Stern-Kai 7, Frankfurt, Germany.
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
Abstract The immune system of an organism is an essential component of the defense mechanism aimed at combating pathogenic stress. Age-associated immune dysfunction, also dubbed "immune senescence," manifests as increased susceptibility to infections, increased onset and progression of autoimmune diseases, and onset of neoplasia. Over the years, extensive research has generated consensus in terms of the phenotypic and functional defects within the immune system in various organisms, including humans. Indeed, age-associated alterations such as thymic involution, T cell repertoire skewing, decreased ability to activate naïve T cells and to generate robust memory responses, have been shown to have a causative role in immune decline. Further, understanding the molecular mechanisms underlying the generation of proteotoxic stress, DNA damage response, modulation of ubiquitin proteasome pathway, and regulation of transcription factor NFκB activation, in immune decline, have paved the way to delineating signaling pathways that cross-talk and impact immune senescence. Given the role of the immune system in combating infections, its effectiveness with age may well be a marker of health and a predictor of longevity. It is therefore believed that a better understanding of the mechanisms underlying immune senescence will lead to an effective interventional strategy aimed at improving the health span of individuals. Antioxid. Redox Signal. 14, 1551-1585.
Collapse
Affiliation(s)
- Subramaniam Ponnappan
- Department of Geriatrics, University of Arkansas for Medical Sciences, 4301 W. Markham, Little Rock, AR 72205, USA
| | | |
Collapse
|
41
|
Lewis DB, Haines C, Ross D. Protein tyrosine kinase 7: a novel surface marker for human recent thymic emigrants with potential clinical utility. J Perinatol 2011; 31 Suppl 1:S72-81. [PMID: 21448210 DOI: 10.1038/jp.2010.187] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent thymic emigrants (RTEs) are antigenically naive T cells that have recently completed intrathymic maturation and have emigrated from the thymus to the periphery. RTEs are clinically and immunologically important as they are essential for maintaining peripheral T cells in sufficient numbers in order to recognize, by their αβT-cell receptors (TCRs), a diverse array of foreign peptide antigens. However, RTE frequency and function has been poorly understood because of a lack of surface markers to distinguish them from older non-RTE naive T cells. This review summarizes the biology of the intrathymic generation and function of RTEs, including the recent identification of protein tyrosine kinase 7 (PTK7) as a novel marker for human RTEs of the CD4 (helper) T-cell lineage. PTK7+ RTEs in adults have a reduced capacity for activation-induced proliferation and cytokine production (interleukin-2 and interferon-γ) than older PTK7- naive CD4 T cells. Importantly, this immaturity in CD4 RTE effector function may contribute to the reduced adaptive immune responses observed in situations in which CD4 RTEs predominate, including the fetus, neonate and young infant, and following immune reconstitution, such as post-hematopoietic stem cell transplant. The ability to identify viable CD4+ RTEs based on PTK7 surface staining may be particularly useful in the infant for better defining the impact of nutritional and environmental factors on thymic output, peripheral T-cell function and adaptive immune responses to vaccination and infection.
Collapse
Affiliation(s)
- D B Lewis
- Division of Immunology and Allergy, Department of Pediatrics, The Institute for Immunology, Transplantation, and Infectious Diseases, The Interdepartmental Program Immunology, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
42
|
Abstract
Nonhuman primates have been used for biomedical research for several decades. The high level of genetic homology to humans coupled with their outbred nature has made nonhuman primates invaluable preclinical models. In this review, we summarize recent advances in our understanding of the nonhuman primate immune system, with special emphasis on studies carried out in rhesus macaque (Macaca mulatta). We highlight the utility of nonhuman primates in the characterization of immune senescence and the evaluation of new interventions to slow down the aging of the immune system.
Collapse
Affiliation(s)
- Ilhem Messaoudi
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon 97006, USA.
| | | | | | | |
Collapse
|
43
|
Calder AE, Hince MN, Dudakov JA, Chidgey AP, Boyd RL. Thymic involution: where endocrinology meets immunology. Neuroimmunomodulation 2011; 18:281-9. [PMID: 21952680 DOI: 10.1159/000329496] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The decline in immune function with aging represents a major clinical challenge in many disease conditions. It is manifest in many parameters but is essentially linked to the adaptive immune responses. The prediction would be that abnormalities in both T and B lymphocytes underlie the loss of cellular and humoral capacity, respectively. Somewhat surprisingly, this is not reflected in numerical losses but more in alterations at the population and single cell levels. There is a major reduction in naïve T cells with a proportional increase in memory cells, and also a generally reduced function of these cells. While bone marrow function reduces with age, the most obvious reason for the T cell defects is the severe atrophy of the thymus. This is closely aligned with puberty, thereby implicating a major aetiological role for sex steroids in both thymus and immune system deterioration with age. Accordingly surgical or chemical castration (utilizing luteinizing hormone-releasing hormone) blocks sex steroids resulting in profound rejuvenation of the immune system.
Collapse
Affiliation(s)
- Adrienne E Calder
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, Vic., Australia
| | | | | | | | | |
Collapse
|
44
|
Reimann C, Dal Cortivo L, Hacein-Bey-Abina S, Fischer A, André-Schmutz I, Cavazzana-Calvo M. Advances in adoptive immunotherapy to accelerate T-cellular immune reconstitution after HLA-incompatible hematopoietic stem cell transplantation. Immunotherapy 2010; 2:481-96. [PMID: 20636003 DOI: 10.2217/imt.10.36] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Although partially HLA-mismatched hematopoietic stem cell transplantation (HSCT) has become an important therapeutic option for children with primary immunodeficiencies, delayed reconstitution of the T-cell compartment remains a major clinical concern. Adoptive immunotherapies to provide recipients with a protective and diverse T-cell repertoire in the months following HSCT are warranted. In order to improve T-cell reconstitution after T-cell-depleted HSCT, different strategies are currently being studied. Some are based on administration of modified mature T cells (e.g., allodepleted T cells or pathogen-specific T cells). Others aim at accelerating de novo thymopoiesis from donor-derived hematopoietic stem cells in vivo via the administration of thymopoietic agents or the transfer of large numbers of T-cell precursors generated ex vivo. The present article will provide a brief summary of recent advances in the field of allodepletion and adoptive transfer of pathogen-specific T cells and a detailed discussion of strategies for enhancing thymopoiesis in vivo.
Collapse
Affiliation(s)
- Christian Reimann
- Institut National de la Santé et de la Recherche Médicale (INSERM), Département de Biothérapie, Hopital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, and Université Paris Descartes 75015 Paris, France
| | | | | | | | | | | |
Collapse
|
45
|
Xu HL, Wang YT, Cheng AC, Yao YF, Ni QY, Zeng W, Bi FJ, Yang ZX, Chen XY. [Polymorphism of MHC-DPB1 gene exon 2 in rhesus macaques (Macaca mulatta)]. YI CHUAN = HEREDITAS 2010; 32:588-98. [PMID: 20566463 DOI: 10.3724/sp.j.1005.2010.00588] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Rhesus macaque (Macaca mulatta) has long been used as an experimental model animal for biomedical research and was under the key state protection (class II) from Chinese government. In order to facilitate the use of Chinese rhesus macaques in biomedical research and their protection based on better understanding of the major mistocompability complex (MHC) genes in these macaques, the exon 2 of Mamu-DPB1 genes were determined in 106 wild rhesus macaques using DGGE, cloning and sequencing. A total of 21 Mamu-DPB1 alleles were obtained, of which 15 alleles were novel sequences that had not been documented previously. Mamu-DPB1 30 was the most frequent allele in the whole large population comprising all 106 rhesus macaque individuals (0.1120) and in Xiaojin population (0.1120), Mamu-DPB1 04 in Heishui (0.1702), -DPB1 32 in Bazhong (0.1613), -DPB1 30 in Hanyuan (0.1120), and -DPB1 04 in Jiulong (0.1139). The alignment of the amino acids sequences showed that 12 variable sites were species-specific, of which 9 sites occurred in the putative amino acids sequences of the 15 novel Mamu-DPB1 alleles. Trans-species polymorphism was observed on the phylogenetic tree based on the DPB1 alleles of rhesus macaques and cynomolgus (Macaca fascicularis). In addition, these results also demonstrated that significant genetic differentiation has occurred between Chinese and Indian rhesus macaque population.
Collapse
Affiliation(s)
- Huai-Liang Xu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW The thymus provides a unique and essential microenvironment for T-cell precursors to develop into mature functionally competent T lymphocytes. Ageing causes architectural changes in the thymus resulting in a loss of thymic epithelial space required for thymopoiesis - a process known as thymic involution. Additionally, cytoablative regimens used to treat malignancies also destroy thymic architecture. The net result of both processes is diminished thymic output and function that may lead to impaired immunity. Thus, immunocompromised individuals would benefit from strategies aimed at enhancing T-cell reconstitution. RECENT FINDINGS Here we discuss strategies such as the use of sex steroid ablation, keratinocyte growth factor, interleukin-7, and in-vitro-generated progenitor T cells as candidates for restoring T-cell immunity. Using various animal models of ageing or hematopoietic stem cell transplantation, these strategies have been shown to restore thymic architecture and cellularity, resulting in increased output and T-cell function in the periphery. SUMMARY These candidate approaches are currently being tested in clinical trials, with preliminary evidence showing encouraging effects on T-cell reconstitution. Nevertheless, although these strategies show clear promise in animal models, and in early human trials, further data are needed to determine their efficacy in patients.
Collapse
|
47
|
Ponchel F, Cuthbert RJ, Goëb V. IL-7 and lymphopenia. Clin Chim Acta 2010; 412:7-16. [PMID: 20850425 DOI: 10.1016/j.cca.2010.09.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 09/01/2010] [Accepted: 09/01/2010] [Indexed: 01/10/2023]
Abstract
Interleukin-7 (IL-7) is a growth and anti-apoptotic factor for T-lymphocytes, with potential for clinical use in the treatment of immunodeficiencies due to loss of T-cells. Lymphopenia induced by disease (HIV infection, hemodialysis or Idiopathic CD4+ lymphopenia) or by treatment (high dose chemotherapy or depleting antibodies) for cancer or auto-immune diseases results in increased circulating levels of IL-7 which decline with T-cell recovery, however, the mechanism of such response remains to be elucidated. Furthermore, IL-7 is a major player in the regulation of peripheral T-cell homeostasis and as such is an important candidate cytokine for therapy aimed at improving T-cell reconstitution following lymphopenia. Anti- IL-7 is on the other hand proposed to treat conditions where IL-7 may play a more direct role in pathogenesis such as autoimmune disease like Rheumatoid Arthritis, Multiple Sclerosis or Inflammatory Bowel disease.
Collapse
Affiliation(s)
- Frederique Ponchel
- Leeds Institute of Molecular Medicine, Section of Musculoskeletal disease, the University of Leeds, Leeds, UK.
| | | | | |
Collapse
|
48
|
Drouet M, Hérodin F. Radiation victim management and the haematologist in the future: time to revisit therapeutic guidelines? Int J Radiat Biol 2010; 86:636-48. [PMID: 20597842 DOI: 10.3109/09553001003789604] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE The use of nuclear/radiation devices against the civilian population is now a realistic scenario. Haematopoietic syndrome is the primary therapeutic challenge in the case of whole body acute exposure over 2 Grays (Gy) whereas burns and combined injuries would be frequently observed in myelo-suppressed patients. Optimisation of scoring and treatments are important goals to achieve. CONCLUSION The European Response Category (RC) concept represents an attempt to integratively assess haematological/extrahematological radiation-induced lesions. Based on the frequently observed heterogeneity of bone marrow damage in accidental/intentional irradiations, the stimulation of residual stem cells using granulocyte Colony-stimulating factor remains the therapeutic standard after exposure to less than the lethal dose 50 % (Haematopoietic[H] score 3-H3). Allogeneic stem cell transplantation is indicated in case of medullary eradication (Haematopoietic score 4-H4) whereas extramedullary toxicity may determine the outcome. Especially in case of numerous casualties exhibiting acute radiation syndrome, the administration of survival factor combinations remains questionable, at least as a palliative treatment. In addition pleiotropic cytokines injection such as erythropoietin and keratinocyte growth factor and grafting multipotent mesenchymal stem cells - from underexposed bone marrow areas or fat tissues - could be proposed to prevent multiple organ failure syndrome development. Multi-disciplinary teams should be prepared to manage such patients.
Collapse
|
49
|
Haberthur K, Engelman F, Barron A, Messaoudi I. Immune senescence in aged nonhuman primates. Exp Gerontol 2010; 45:655-61. [PMID: 20558288 DOI: 10.1016/j.exger.2010.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 05/27/2010] [Accepted: 06/03/2010] [Indexed: 12/22/2022]
Abstract
Aging is accompanied by a general dysregulation in immune system function, commonly referred to as 'immune senescence'. This progressive deterioration affects both innate and adaptive immunity, although accumulating evidence indicates that the adaptive arm of the immune system may exhibit more profound changes. Most of our current understanding of immune senescence stems from clinical and rodent studies. More recently, the use of nonhuman primates (NHPs) to investigate immune senescence and test interventions aimed at delaying/reversing age-related changes in immune function has dramatically increased. These studies have been greatly facilitated by several key advances in our understanding of the immune system of old world monkeys, specifically the rhesus macaques. In this review we describe the hallmarks of immune senescence in this species and compare them to those described in humans. We also discuss the impact of immune senescence on the response to vaccination and the efficacy of immuno-restorative interventions investigated in this model system.
Collapse
Affiliation(s)
- Kristen Haberthur
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, United States
| | | | | | | |
Collapse
|
50
|
Holländer GA, Krenger W, Blazar BR. Emerging strategies to boost thymic function. Curr Opin Pharmacol 2010; 10:443-53. [PMID: 20447867 DOI: 10.1016/j.coph.2010.04.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 04/06/2010] [Accepted: 04/06/2010] [Indexed: 11/28/2022]
Abstract
The thymus constitutes the primary lymphoid organ for the generation of T cells. Its function is particularly susceptible to various negative influences ranging from age-related involution to atrophy as a consequence of malnutrition, infection or harmful iatrogenic influences such as chemotherapy and radiation. The loss of regular thymus function significantly increases the risk for infections and cancer because of a restricted capacity for immune surveillance. In recent years, thymus-stimulatory, thymus-regenerative, and thymus-protective strategies have been developed to enhance and repair thymus function in the elderly and in individuals undergoing hematopoietic stem cell transplantation. These strategies include the use of sex steroid ablation, the administration of growth and differentiation factors, the inhibition of p53, and the transfer of T cell progenitors to alleviate the effects of thymus dysfunction and consequent T cell deficiency.
Collapse
Affiliation(s)
- Georg A Holländer
- Laboratory of Pediatric Immunology, Department of Biomedicine, University of Basel, The University Children's Hospital (UKBB), Mattenstrasse 28, 4058 Basel, Switzerland.
| | | | | |
Collapse
|