1
|
Mapuskar KA, London B, Zacharias ZR, Houtman JC, Allen BG. Immunometabolism in the Aging Heart. J Am Heart Assoc 2025; 14:e039216. [PMID: 39719411 PMCID: PMC12054428 DOI: 10.1161/jaha.124.039216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/19/2024] [Indexed: 12/26/2024]
Abstract
Structural, functional, and molecular-level changes in the aging heart are influenced by a dynamic interplay between immune signaling and cellular metabolism that is referred to as immunometabolism. This review explores the crosstalk between cellular metabolic pathways including glycolysis, oxidative phosphorylation, fatty acid metabolism, and the immune processes that govern cardiac aging. With a rapidly aging population that coincides with increased cardiovascular risk and cancer incidence rates, understanding the immunometabolic underpinnings of cardiac aging provides a foundation for identifying therapeutic targets to mitigate cardiac dysfunction. Aging alters the immune environment of the heart by concomitantly driving the changes in immune cell metabolism, mitochondrial dysfunction, and redox signaling. Shifts in these metabolic pathways exacerbate inflammation and impair tissue repair, creating a vicious cycle that accelerates cardiac functional decline. Treatment with cancer therapy further complicates this landscape, as aging-associated immunometabolic disruptions augment the susceptibility to cardiotoxicity. The current review highlights therapeutic strategies that target the immunometabolic axis to alleviate cardiac aging pathologies. Interventions include modulating metabolic intermediates, improving mitochondrial function, and leveraging immune signaling pathways to restore cardiac health. Advances in immunometabolism thus hold significant potential for translating preclinical findings into therapies that improve the quality of life for the aging population and underscore the need for approaches that address the immunometabolic mechanisms of cardiac aging, providing a framework for future research.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Department of Radiation OncologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Barry London
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Department of Internal MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Zeb R. Zacharias
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Human Immunology CoreUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Jon C.D. Houtman
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Human Immunology CoreUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Department of Microbiology and ImmunologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Bryan G. Allen
- Department of Radiation OncologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| |
Collapse
|
2
|
Skariah N, James OJ, Swamy M. Signalling mechanisms driving homeostatic and inflammatory effects of interleukin-15 on tissue lymphocytes. DISCOVERY IMMUNOLOGY 2024; 3:kyae002. [PMID: 38405398 PMCID: PMC10883678 DOI: 10.1093/discim/kyae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/19/2023] [Accepted: 01/26/2024] [Indexed: 02/27/2024]
Abstract
There is an intriguing dichotomy in the function of cytokine interleukin-15-at low levels, it is required for the homeostasis of the immune system, yet when it is upregulated in response to pathogenic infections or in autoimmunity, IL-15 drives inflammation. IL-15 associates with the IL-15Rα within both myeloid and non-haematopoietic cells, where IL-15Rα trans-presents IL-15 in a membrane-bound form to neighboring cells. Alongside homeostatic maintenance of select lymphocyte populations such as NK cells and tissue-resident T cells, when upregulated, IL-15 also promotes inflammatory outcomes by driving effector function and cytotoxicity in NK cells and T cells. As chronic over-expression of IL-15 can lead to autoimmunity, IL-15 expression is tightly regulated. Thus, blocking dysregulated IL-15 and its downstream signalling pathways are avenues for immunotherapy. In this review we discuss the molecular pathways involved in IL-15 signalling and how these pathways contribute to both homeostatic and inflammatory functions in IL-15-dependent mature lymphoid populations, focusing on innate, and innate-like lymphocytes in tissues.
Collapse
Affiliation(s)
- Neema Skariah
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Olivia J James
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Mahima Swamy
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| |
Collapse
|
3
|
Sottini A, Quaresima V, Barbaro M, Moiola L, Filippi M, Malentacchi M, Capobianco M, Puthenparampil M, Gallo P, Cocco E, Frau J, Zaffaroni M, Guaschino C, Stampatori C, Mancinelli C, Brambilla L, Clerici VT, Vianello M, Vitetta F, Ferraro D, Rosettani P, Danni MC, Conti M, Grimoldi M, Capra R, Imberti L. Clinical relevance of thymic and bone marrow outputs in multiple sclerosis patients treated with alemtuzumab. J Neuroimmunol 2023; 382:578170. [PMID: 37579546 DOI: 10.1016/j.jneuroim.2023.578170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/16/2023]
Abstract
Thymic and bone marrow outputs were evaluated in 13 sequential samples of 68 multiple sclerosis patients who initiated alemtuzumab and were clinically followed for 48 months. Three months after alemtuzumab infusions, the levels of new T lymphocytes were significantly reduced, but progressively increased reaching the highest values at 36 months, indicating the remarkable capacity of thymic function recovery. Newly produced B cells exceeded baseline levels as early as 3 months after alemtuzumab initiation. Heterogeneous patterns of new T- and B-cell recovery were identified, but without associations with age, sex, previous therapies, development of secondary autoimmunity or infections, and disease re-emergence. Trial registration version 2.0-27/01/2016.
Collapse
Affiliation(s)
- Alessandra Sottini
- Diagnostic Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy.
| | - Virginia Quaresima
- Diagnostic Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Mosè Barbaro
- Diagnostic Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy; Laboratorio analisi, Ospedale Civile di Sondrio, ASST Valtellina e Alto Lario, Sondrio, Italy
| | - Lucia Moiola
- Neurology Department-Multiple Sclerosis Center, IRCCS San Raffaele Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Filippi
- Neurology and Neurorehabilitation Units, MS Center, Headache Center, Epilepsy Center, and Stroke Unit, Neurophysiology Service, and Neuroimaging Research Unit, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Maria Malentacchi
- SCDO Neurologia e Centro di Riferimento Regionale Sclerosi Multipla, AOU San Luigi Gonzaga, Orbassano, Italy
| | - Marco Capobianco
- SCDO Neurologia e Centro di Riferimento Regionale Sclerosi Multipla, AOU San Luigi Gonzaga, Orbassano, Italy
| | - Marco Puthenparampil
- Department of Neuroscience (DNS), School of Medicine - University of Padua, Padua, Italy
| | - Paolo Gallo
- Department of Neuroscience (DNS), School of Medicine - University of Padua, Padua, Italy
| | - Eleonora Cocco
- Centro Sclerosi Multipla AOU Cagliari - University of Cagliari, Italy
| | | | - Mauro Zaffaroni
- Centro Sclerosi Multipla, Ospedale di Gallarate, ASST della Valle Olona, Gallarate, Italy
| | - Clara Guaschino
- Centro Sclerosi Multipla, Ospedale di Gallarate, ASST della Valle Olona, Gallarate, Italy
| | - Chiara Stampatori
- Centro Regionale per la Sclerosi Multipla, ASST Spedali Civili di Brescia, Montichiari, Brescia, Italy
| | - Chiara Mancinelli
- Centro Regionale per la Sclerosi Multipla, ASST Spedali Civili di Brescia, Montichiari, Brescia, Italy; U.O. Neuroimmunologia e Malattie Neuromuscolari, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Laura Brambilla
- U.O. Neuroimmunologia e Malattie Neuromuscolari, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Valentina Torri Clerici
- U.O. Neuroimmunologia e Malattie Neuromuscolari, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Francesca Vitetta
- Centro Malattie Demielinizzanti, Ospedale Civile Baggiovara, AOU Modena, Italy
| | - Diana Ferraro
- Centro Malattie Demielinizzanti, Ospedale Civile Baggiovara, AOU Modena, Italy
| | - Pamela Rosettani
- Clinica Neurologica, Azienda Ospedaliero Universitaria delle Marche, Torrette, Ancona, Italy
| | - Maura Chiara Danni
- Clinica Neurologica, Azienda Ospedaliero Universitaria delle Marche, Torrette, Ancona, Italy
| | - Marta Conti
- Department of Neurology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Maria Grimoldi
- Department of Neurology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Ruggero Capra
- Centro Regionale per la Sclerosi Multipla, ASST Spedali Civili di Brescia, Montichiari, Brescia, Italy
| | - Luisa Imberti
- Diagnostic Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy; Section of Microbiology, University of Brescia, P. le Spedali Civili, 1, Brescia, Italy
| |
Collapse
|
4
|
Liu Q, Zheng Y, Goronzy JJ, Weyand CM. T cell aging as a risk factor for autoimmunity. J Autoimmun 2023; 137:102947. [PMID: 36357240 PMCID: PMC10164202 DOI: 10.1016/j.jaut.2022.102947] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 10/23/2022] [Indexed: 11/09/2022]
Abstract
Immune aging is a complex process rendering the host susceptible to cancer, infection, and insufficient tissue repair. Many autoimmune diseases preferentially occur during the second half of life, counterintuitive to the concept of excess adaptive immunity driving immune-mediated tissue damage. T cells are particularly susceptible to aging-imposed changes, as they are under extreme proliferative pressure to fulfill the demands of clonal expansion and of homeostatic T cell repopulation. T cells in older adults have a footprint of genetic and epigenetic changes, lack mitochondrial fitness, and fail to maintain proteostasis, diverging them from host protection to host injury. Here, we review recent progress in understanding how the human T-cell system ages and the evidence detailing how T cell aging contributes to autoimmune conditions. T cell aging is now recognized as a risk determinant in two prototypic autoimmune syndromes; rheumatoid arthritis and giant cell arteritis. The emerging concept adds susceptibility to autoimmune and autoinflammatory disease to the spectrum of aging-imposed adaptations and opens new opportunities for immunomodulatory therapy by restoring the functional intactness of aging T cells.
Collapse
Affiliation(s)
- Qingxiang Liu
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Yanyan Zheng
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Cardiovascular Medicine, Mayo Alix School of Medicine, Rochester, MN, USA
| | - Jorg J Goronzy
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94306, USA
| | - Cornelia M Weyand
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Cardiovascular Medicine, Mayo Alix School of Medicine, Rochester, MN, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94306, USA.
| |
Collapse
|
5
|
Silaeva YY, Kalinina AA, Khromykh LM, Deykin AV, Kazansky DB. Formation of a Unique Population of CD8+ T Lymphocytes after Adoptive Transfer of Syngeneic Splenocytes to Mice with Lymphopenia. DOKL BIOCHEM BIOPHYS 2021; 497:71-74. [PMID: 33666806 PMCID: PMC8068684 DOI: 10.1134/s1607672921020137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 11/23/2022]
Abstract
Under conditions of lymphopenia, T lymphocytes proliferate and acquire a surface activation phenotype, which in many respects is similar to the phenotype of true memory T cells. We investigated the phenotypic features of the CD8+ T-cell population formed from donor lymphocytes after adoptive transfer of syngeneic splenocytes to sublethally irradiated mice. This population expresses markers CD44, CD122, CD5, CD49d and the chemokine receptor CXCR3. Thus, for the first time, the phenomenon of the formation of a population of T cells with signs of suppressive CD8+ T lymphocytes and true memory cells was demonstrated.
Collapse
Affiliation(s)
- Yu Yu Silaeva
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.
| | - A A Kalinina
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - L M Khromykh
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A V Deykin
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - D B Kazansky
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
6
|
Kalinina AA, Khromykh LM, Kazansky DB, Deykin AV, Silaeva YY. Suppression of the Immune Response by Syngeneic Splenocytes Adoptively Transferred to Sublethally Irradiated Mice. Acta Naturae 2021; 13:116-126. [PMID: 33959391 PMCID: PMC8084293 DOI: 10.32607/actanaturae.11252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The peripheral T-cell pool consists of several, functionally distinct
populations of CD8+ T cells. CD44 and CD62L are among the major
surface markers that allow us to define T-cell populations. The expression of
these molecules depends on the functional status of a T lymphocyte. Under
lymphopenic conditions, peripheral T cells undergo homeostatic proliferation
and acquire the memory-like surface phenotype CD44hiCD62Lhi. However, the data
on the functional activity of these cells remains controversial. In this paper,
we analyzed the effects of the adoptive transfer of syngeneic splenocytes on
the recovery of CD8+ T cells in sublethally irradiated mice. Our
data demonstrate that under lymphopenia, donor lymphocytes form a population of
memory-like CD8+ T cells with the phenotype CD122+CD5+CD49dhiCXCR3+
that shares the phenotypic characteristics of true memory cells and suppressive
CD8+ T cells. Ex vivo experiments showed that after
adoptive transfer in irradiated mice, T cells lacked the functions of true
effector or memory cells; the allogeneic immune response and immune response to
pathogens were greatly suppressed in these mice.
Collapse
Affiliation(s)
- A. A. Kalinina
- Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow, 115478 Russia
| | - L. M. Khromykh
- Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow, 115478 Russia
| | - D. B. Kazansky
- Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow, 115478 Russia
| | - A. V. Deykin
- Core Facility Centre, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
| | - Yu. Yu. Silaeva
- Core Facility Centre, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
| |
Collapse
|
7
|
Abdalla AME, Xiao L, Miao Y, Huang L, Fadlallah GM, Gauthier M, Ouyang C, Yang G. Nanotechnology Promotes Genetic and Functional Modifications of Therapeutic T Cells Against Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903164. [PMID: 32440473 PMCID: PMC7237845 DOI: 10.1002/advs.201903164] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/23/2020] [Indexed: 05/24/2023]
Abstract
Growing experience with engineered chimeric antigen receptor (CAR)-T cells has revealed some of the challenges associated with developing patient-specific therapy. The promising clinical results obtained with CAR-T therapy nevertheless demonstrate the urgency of advancements to promote and expand its uses. There is indeed a need to devise novel methods to generate potent CARs, and to confer them and track their anti-tumor efficacy in CAR-T therapy. A potentially effective approach to improve the efficacy of CAR-T cell therapy would be to exploit the benefits of nanotechnology. This report highlights the current limitations of CAR-T immunotherapy and pinpoints potential opportunities and tremendous advantages of using nanotechnology to 1) introduce CAR transgene cassettes into primary T cells, 2) stimulate T cell expansion and persistence, 3) improve T cell trafficking, 4) stimulate the intrinsic T cell activity, 5) reprogram the immunosuppressive cellular and vascular microenvironments, and 6) monitor the therapeutic efficacy of CAR-T cell therapy. Therefore, genetic and functional modifications promoted by nanotechnology enable the generation of robust CAR-T cell therapy and offer precision treatments against cancer.
Collapse
Affiliation(s)
- Ahmed M. E. Abdalla
- Department of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074China
- Department of BiochemistryCollege of Applied ScienceUniversity of BahriKhartoum1660/11111Sudan
| | - Lin Xiao
- Department of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074China
| | - Yu Miao
- Department of Vascular SurgeryGeneral Hospital of Ningxia Medical UniversityYinchuan750004China
| | - Lixia Huang
- Hubei Key Laboratory of Purification and Application of Plant Anti‐Cancer Active IngredientsSchool of Chemistry and Life SciencesHubei University of EducationWuhan430205China
| | - Gendeal M. Fadlallah
- Department of Chemistry and BiologyFaculty of EducationUniversity of GeziraWad‐Medani2667Sudan
| | - Mario Gauthier
- Department of ChemistryUniversity of WaterlooWaterlooN2L 3G1Canada
| | - Chenxi Ouyang
- Department of Vascular SurgeryFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Guang Yang
- Department of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074China
| |
Collapse
|
8
|
Unregulated antigen-presenting cell activation by T cells breaks self tolerance. Proc Natl Acad Sci U S A 2018; 116:1007-1016. [PMID: 30598454 DOI: 10.1073/pnas.1818624116] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
T cells proliferate vigorously following acute depletion of CD4+ Foxp3+ T regulatory cells [natural Tregs (nTregs)] and also when naive T cells are transferred to syngeneic, nTreg-deficient Rag1 -/- hosts. Here, using mice raised in an antigen-free (AF) environment, we show that proliferation in these two situations is directed to self ligands rather than food or commensal antigens. In both situations, the absence of nTregs elevates B7 expression on host dendritic cells (DCs) and enables a small subset of naive CD4 T cells with high self affinity to respond overtly to host DCs: bidirectional T/DC interaction ensues, leading to progressive DC activation and reciprocal strong proliferation of T cells accompanied by peripheral Treg (pTreg) formation. Likewise, high-affinity CD4 T cells proliferate vigorously and form pTregs when cultured with autologous DCs in vitro in the absence of nTregs: this anti-self response is MHCII/peptide dependent and elicited by the raised level of B7 on cultured DCs. The data support a model in which self tolerance is imposed via modulation of CD28 signaling and explains the pathological effects of superagonistic CD28 antibodies.
Collapse
|
9
|
Evaluation of quantitative changes in regulatory T cells in peripheral blood of kidney transplant recipients with skin cancer after conversion to mTOR inhibitors. Postepy Dermatol Alergol 2018; 35:474-480. [PMID: 30429704 PMCID: PMC6232553 DOI: 10.5114/ada.2018.77237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/23/2017] [Indexed: 11/17/2022] Open
Abstract
Introduction Immunosuppressive therapy, necessary for graft survival, has its clinical consequences with an increased risk of developing malignancies being one of them. It seems that the maintenance of a proper balance between cytotoxic and regulatory activity of the immune system may prevent graft rejection, and with a lower risk of cancer. Aim To assess the quantitative changes in regulatory T cells (Tregs) in peripheral blood of kidney transplant recipients with post-transplantation skin neoplasm after conversion to mTOR inhibitors (mTORi) and to assess the incidence of secondary skin cancer in that group of patients. Material and methods Fourteen patients with post-transplant cutaneous malignancies converted to mTORi were included into the study. The control group consisted of eighteen patients maintained on immunosuppressive regimens without mTORi. The level of Tregs with a phenotype defined as CD4lowCD25high was measured before, and 6 months after, mTORi introduction. Results In all cases, 6 months after conversion, a significant decrease in the ratio of CD4+CD25+ to CD4lowCD25high from 6.52 to 4.29 was detected (p = 0.035). One patient converted to mTORi developed subsequent skin cancer, while in the control group, subsequent skin cancer was recognized in eight patients. Moreover, introducing mTORi significantly improved progression-free survival in this group of patients (p = 0.016). Conclusions Introducing mTORi to the immunosuppressive regimen resulted in an increase in the number of regulatory cells without increasing the incidence of secondary skin cancer in the investigated group of patients.
Collapse
|
10
|
Massey JC, Sutton IJ, Ma DDF, Moore JJ. Regenerating Immunotolerance in Multiple Sclerosis with Autologous Hematopoietic Stem Cell Transplant. Front Immunol 2018; 9:410. [PMID: 29593711 PMCID: PMC5857574 DOI: 10.3389/fimmu.2018.00410] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/14/2018] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disorder of the central nervous system where evidence implicates an aberrant adaptive immune response in the accrual of neurological disability. The inflammatory phase of the disease responds to immunomodulation to varying degrees of efficacy; however, no therapy has been proven to arrest progression of disability. Recently, more intensive therapies, including immunoablation with autologous hematopoietic stem cell transplantation (AHSCT), have been offered as a treatment option to retard inflammatory disease, prior to patients becoming irreversibly disabled. Empirical clinical observations support the notion that the immune reconstitution (IR) that occurs following AHSCT is associated with a sustained therapeutic benefit; however, neither the pathogenesis of MS nor the mechanism by which AHSCT results in a therapeutic benefit has been clearly delineated. Although the antigenic target of the aberrant immune response in MS is not defined, accumulated data suggest that IR following AHSCT results in an immunotolerant state through deletion of pathogenic clones by a combination of direct ablation and induction of a lymphopenic state driving replicative senescence and clonal attrition. Restoration of immunoregulation is evidenced by changes in regulatory T cell populations following AHSCT and normalization of genetic signatures of immune homeostasis. Furthermore, some evidence exists that AHSCT may induce a rebooting of thymic function and regeneration of a diversified naïve T cell repertoire equipped to appropriately modulate the immune system in response to future antigenic challenge. In this review, we discuss the immunological mechanisms of IR therapies, focusing on AHSCT, as a means of recalibrating the dysfunctional immune response observed in MS.
Collapse
Affiliation(s)
- Jennifer C Massey
- Haematology and Bone Marrow Transplantation, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,Neurology, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Ian J Sutton
- Neurology, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - David D F Ma
- Haematology and Bone Marrow Transplantation, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - John J Moore
- Haematology and Bone Marrow Transplantation, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
11
|
Przybylski S, Gasch M, Marschner A, Ebert M, Ewe A, Helmig G, Hilger N, Fricke S, Rudzok S, Aigner A, Burkhardt J. Influence of nanoparticle-mediated transfection on proliferation of primary immune cells in vitro and in vivo. PLoS One 2017; 12:e0176517. [PMID: 28463994 PMCID: PMC5412997 DOI: 10.1371/journal.pone.0176517] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 03/19/2017] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION One of the main obstacles in the widespread application of gene therapeutic approaches is the necessity for efficient and safe transfection methods. For the introduction of small oligonucleotide gene therapeutics into a target cell, nanoparticle-based methods have been shown to be highly effective and safe. While immune cells are a most interesting target for gene therapy, transfection might influence basic immune functions such as cytokine expression and proliferation, and thus positively or negatively affect therapeutic intervention. Therefore, we investigated the effects of nanoparticle-mediated transfection such as polyethylenimine (PEI) or magnetic beads on immune cell proliferation. METHODS Human adherent and non-adherent PBMCs were transfected by various methods (e.g. PEI, Lipofectamine® 2000, magnetofection) and stimulated. Proliferation was measured by lymphocyte transformation test (LTT). Cell cycle stages as well as expression of proliferation relevant genes were analyzed. Additionally, the impact of nanoparticles was investigated in vivo in a murine model of the severe systemic immune disease GvHD (graft versus host disease). RESULTS The proliferation of primary immune cells was influenced by nanoparticle-mediated transfection. In particular in the case of magnetic beads, proliferation inhibition coincided with short-term cell cycle arrest and reduced expression of genes relevant for immune cell proliferation. Notably, proliferation inhibition translated into beneficial effects in a murine GvHD model with animals treated with PEI-nanoparticles showing increased survival (pPEI = 0.002) most likely due to reduced inflammation. CONCLUSION This study shows for the first time that nanoparticles utilized for gene therapeutic transfection are able to alter proliferation of immune cells and that this effect depends on the type of nanoparticle. For magnetic beads, this was accompanied by temporary cell cycle arrest. Notably, in GvHD this nonspecific anti-proliferative effect might contribute to reduced inflammation and increased survival.
Collapse
Affiliation(s)
- Susanne Przybylski
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Michaela Gasch
- Translationszentrum für Regenerative Medizin (TRM), University of Leipzig, Leipzig, Germany
| | - Anne Marschner
- Institute for Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Marcus Ebert
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Alexander Ewe
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Leipzig, Germany
| | - Gisa Helmig
- Institute for Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Nadja Hilger
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Stephan Fricke
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Susanne Rudzok
- Translationszentrum für Regenerative Medizin (TRM), University of Leipzig, Leipzig, Germany
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Leipzig, Germany
| | - Jana Burkhardt
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- * E-mail:
| |
Collapse
|
12
|
Imko-Walczuk B, Piesiaków M, Trzonkowski P, Pikuła M, Dębska-Ślizień A, Rutkowski B. Associations of Selected Cytokines Levels in Organ Transplant Recipients Without and With Malignant Skin Neoplasms. Transplant Proc 2016; 48:1654-9. [DOI: 10.1016/j.transproceed.2016.03.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/26/2016] [Accepted: 03/30/2016] [Indexed: 01/08/2023]
|
13
|
Alemtuzumab as Antirejection Therapy: T Cell Repopulation and Cytokine Responsiveness. Transplant Direct 2016; 2:e83. [PMID: 27500273 PMCID: PMC4946521 DOI: 10.1097/txd.0000000000000595] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/10/2016] [Indexed: 01/05/2023] Open
Abstract
Supplemental digital content is available in the text. Background Alemtuzumab induction therapy in kidney transplant patients results in T cell depletion followed by slow immune reconstitution of memory T cells with reduced immune functions. The kinetics and functional characteristics of T cell reconstitution when alemtuzumab is given during immune activation, ie, as antirejection therapy, are unknown. Methods Patients (n = 12) with glucocorticoid-resistant or severe vascular kidney transplant rejection were treated with alemtuzumab. Flow cytometric analysis was performed on whole blood to measure cell division by the marker Ki-67, and cytokine responsiveness by IL-2–mediated and IL-7–mediated phosphorylation of signal transducer and activator of transcription 5 of T cells before and during the first year after rejection therapy. Results At 1 year after alemtuzumab antirejection therapy, the total T cell population recovered to baseline level. Repopulation of CD4+ and CD8+ T cells was associated with increased percentages of Ki-67+ proliferating T cells (P < 0.05). In addition, both populations showed a phenotypic shift toward relatively more memory T cells (P < 0.01). At the functional level, IL-7 reactivity of CD4+ memory T cells was diminished, reflected by a decreased capacity to phosphorylate signal transducer and activator of transcription 5 during the first 6 months after alemtuzumab treatment (P < 0.05), whereas reactivity to IL-2 was preserved. CD8+ T cells were affected in terms of both IL-2 and IL-7 responses (both P < 0.05). After reconstitution, relatively more regulatory T cells were present, and a relatively high proportion of Ki-67+ T cells was observed. Conclusions Preliminary data from this small series suggest that alemtuzumab antirejection therapy induces homeostatic proliferation of memory and regulatory T cells with diminished responsiveness to the homeostatic cytokine IL-7. IL-2 responsiveness was affected in repopulated CD8+ T cells.
Collapse
|
14
|
Reading JL, Vaes B, Hull C, Sabbah S, Hayday T, Wang NS, DiPiero A, Lehman NA, Taggart JM, Carty F, English K, Pinxteren J, Deans R, Ting AE, Tree TIM. Suppression of IL-7-dependent Effector T-cell Expansion by Multipotent Adult Progenitor Cells and PGE2. Mol Ther 2015. [PMID: 26216515 DOI: 10.1038/mt.2015.131] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
T-cell depletion therapy is used to prevent acute allograft rejection, treat autoimmunity and create space for bone marrow or hematopoietic cell transplantation. The evolved response to T-cell loss is a transient increase in IL-7 that drives compensatory homeostatic proliferation (HP) of mature T cells. Paradoxically, the exaggerated form of this process that occurs following lymphodepletion expands effector T-cells, often causing loss of immunological tolerance that results in rapid graft rejection, autoimmunity, and exacerbated graft-versus-host disease (GVHD). While standard immune suppression is unable to treat these pathologies, growing evidence suggests that manipulating the incipient process of HP increases allograft survival, prevents autoimmunity, and markedly reduces GVHD. Multipotent adult progenitor cells (MAPC) are a clinical grade immunomodulatory cell therapy known to alter γ-chain cytokine responses in T-cells. Herein, we demonstrate that MAPC regulate HP of human T-cells, prevent the expansion of Th1, Th17, and Th22 effectors, and block the development of pathogenic allograft responses. This occurs via IL-1β-primed secretion of PGE2 and activates T-cell intrinsic regulatory mechanisms (SOCS2, GADD45A). These data provide proof-of-principle that HP of human T-cells can be targeted by cellular and molecular therapies and lays a basis for the development of novel strategies to prevent immunopathology in lymphodepleted patients.
Collapse
Affiliation(s)
- James L Reading
- Department of Immunobiology, King's College London, London, UK.
| | | | - Caroline Hull
- Department of Immunobiology, King's College London, London, UK
| | - Shereen Sabbah
- Department of Immunobiology, King's College London, London, UK
| | - Thomas Hayday
- Department of Immunobiology, King's College London, London, UK
| | | | | | | | | | - Fiona Carty
- Department of Biology, Institute of Immunology, National University of Ireland, Maynooth, Ireland
| | - Karen English
- Department of Biology, Institute of Immunology, National University of Ireland, Maynooth, Ireland
| | | | | | | | - Timothy I M Tree
- Department of Immunobiology, King's College London, London, UK; NIHR Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
15
|
Parzych EM, Li H, Yin X, Liu Q, Wu TL, Podsakoff GM, High KA, Levine MH, Ertl HCJ. Effects of immunosuppression on circulating adeno-associated virus capsid-specific T cells in humans. Hum Gene Ther 2014; 24:431-42. [PMID: 23461589 DOI: 10.1089/hum.2012.246] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In humans adeno-associated virus (AAV)-mediated gene transfer is followed by expansion of AAV capsid-specific T cells, evidence of cell damage, and loss of transgene product expression, implicating immunological rejection of vector-transduced cells, which may be prevented by immunosuppressive drugs. We undertook this study to assess the effect of immunosuppression (IS) used for organ transplantation on immune responses to AAV capsid antigens. Recipients of liver or kidney transplants were tested before and 4 weeks after induction of IS in comparison with matched samples from healthy human adults and an additional cohort with comorbid conditions similar to those of the transplant patients. Our data show that transplant patients and comorbid control subjects have markedly higher frequencies of circulating AAV capsid-specific T cells compared with healthy adults. On average, IS resulted in a reduction of AAV-specific CD4⁺ T cells, whereas numbers of circulating CD8⁺ effector and central memory T cells tended to increase. Independent of the type of transplant or the IS regimens, the trend of AAV capsid-specific T cell responses after drug treatment varied; in some patients responses were unaffected whereas others showed decreases or even pronounced increases, casting doubt on the usefulness of prophylactic IS for AAV vector recipients.
Collapse
|
16
|
Goronzy JJ, Li G, Yang Z, Weyand CM. The janus head of T cell aging - autoimmunity and immunodeficiency. Front Immunol 2013; 4:131. [PMID: 23761790 PMCID: PMC3671290 DOI: 10.3389/fimmu.2013.00131] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 05/21/2013] [Indexed: 01/09/2023] Open
Abstract
Immune aging is best known for its immune defects that increase susceptibility to infections and reduce adaptive immune responses to vaccination. In parallel, the aged immune system is prone to autoimmune responses and many autoimmune diseases increase in incidence with age or are even preferentially encountered in the elderly. Why an immune system that suboptimally responds to exogenous antigen fails to maintain tolerance to self-antigens appears to be perplexing. In this review, we will discuss age-associated deviations in the immune repertoire and the regulation of signaling pathways that may shed light on this conundrum.
Collapse
Affiliation(s)
- Jörg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine , Stanford, CA , USA ; Department of Medicine, Palo Alto Veteran Administration Health Care System , Palo Alto, CA , USA
| | | | | | | |
Collapse
|
17
|
Mihalj M, Kellermayer Z, Balogh P. Follicles in gut-associated lymphoid tissues create preferential survival niches for follicular Th cells escaping Thy-1-specific depletion in mice. Int Immunol 2013; 25:423-35. [PMID: 23449667 DOI: 10.1093/intimm/dxt001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Although a substantial number of T cells may escape depletion following in vivo mAb treatment in patients undergoing immunosuppression, their specific tissue location and phenotypic characteristics in different peripheral lymphoid tissues have not been analyzed in detail. Here we investigated the survival of CD4(+) T cells immediately following anti-Thy-1 mAb treatment in mice. We found a preferential survival of CD4(+) T cells expressing Thy-1 antigen in the Peyer's patches (PP) and also in mesenteric lymph nodes (MLN), where the relative majority of the surviving CD4(+) T cells displayed CD44(high)/CD62L(-) phenotype corresponding to effector memory T-cell features. These CD4(+) T cells also expressed CXCR5 and PD-1 (programmed cell death-1) markers characteristic for follicular Th cells (TFH). We also demonstrate that the immediate survival of these cells does not involve proliferation and is independent of IL-7. Induction of germinal center formation in spleen enhanced while the dissolution of follicular architecture by lymphotoxin-β receptor antagonist treatment slightly reduced TFH survival. Our results thus raise the possibility that the follicles within PP and MLN may create natural support niches for the preferential survival of TFH cells of the memory phenotype, thus allowing their escape during T-cell depletion.
Collapse
Affiliation(s)
- Martina Mihalj
- Department of Immunology & Biotechnology, Faculty of Medicine, University of Pécs, Szigeti ut 12, Pécs H-7624, Hungary
| | | | | |
Collapse
|
18
|
del Rio ML, Jones ND, Buhler L, Norris P, Shintani Y, Ware CF, Rodriguez-Barbosa JI. Selective blockade of herpesvirus entry mediator-B and T lymphocyte attenuator pathway ameliorates acute graft-versus-host reaction. THE JOURNAL OF IMMUNOLOGY 2012; 188:4885-96. [PMID: 22490863 DOI: 10.4049/jimmunol.1103698] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The cosignaling network mediated by the herpesvirus entry mediator (HVEM; TNFRSF14) functions as a dual directional system that involves proinflammatory ligand, lymphotoxin that exhibits inducible expression and competes with HSV glycoprotein D for HVEM, a receptor expressed by T lymphocytes (LIGHT; TNFSF14), and the inhibitory Ig family member B and T lymphocyte attenuator (BTLA). To dissect the differential contributions of HVEM/BTLA and HVEM/LIGHT interactions, topographically-specific, competitive, and nonblocking anti-HVEM Abs that inhibit BTLA binding, but not LIGHT, were developed. We demonstrate that a BTLA-specific competitor attenuated the course of acute graft-versus-host reaction in a murine F(1) transfer semiallogeneic model. Selective HVEM/BTLA blockade did not inhibit donor T cell infiltration into graft-versus-host reaction target organs, but decreased the functional activity of the alloreactive T cells. These results highlight the critical role of HVEM/BTLA pathway in the control of the allogeneic immune response and identify a new therapeutic target for transplantation and autoimmune diseases.
Collapse
Affiliation(s)
- Maria-Luisa del Rio
- Immunobiology Section, Institute of Biomedicine, University of Leon, 24007 Leon, Spain
| | | | | | | | | | | | | |
Collapse
|
19
|
Immune aging and autoimmunity. Cell Mol Life Sci 2012; 69:1615-23. [PMID: 22466672 DOI: 10.1007/s00018-012-0970-0] [Citation(s) in RCA: 192] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Revised: 03/13/2012] [Accepted: 03/13/2012] [Indexed: 01/09/2023]
Abstract
Age is an important risk for autoimmunity, and many autoimmune diseases preferentially occur in the second half of adulthood when immune competence has declined and thymic T cell generation has ceased. Many tolerance checkpoints have to fail for an autoimmune disease to develop, and several of those are susceptible to the immune aging process. Homeostatic T cell proliferation which is mainly responsible for T cell replenishment during adulthood can lead to the selection of T cells with increased affinity to self- or neoantigens and enhanced growth and survival properties. These cells can acquire a memory-like phenotype, in particular under lymphopenic conditions. Accumulation of end-differentiated effector T cells, either specific for self-antigen or for latent viruses, have a low activation threshold due to the expression of signaling and regulatory molecules and generate an inflammatory environment with their ability to be cytotoxic and to produce excessive amounts of cytokines and thereby inducing or amplifying autoimmune responses.
Collapse
|
20
|
Marek N, Bieniaszewska M, Krzystyniak A, Juścińska J, Myśliwska J, Witkowski P, Hellmann A, Trzonkowski P. The time is crucial for ex vivo expansion of T regulatory cells for therapy. Cell Transplant 2011; 20:1747-58. [PMID: 21457615 DOI: 10.3727/096368911x566217] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Ex vivo expanded CD4(+)CD25(high)CD127(-) T regulatory cells (Tregs) are recognized as a promising candidate for immunosuppressive therapy in humans. However, due to the plasticity of Tregs lineage and artificial environment present during ex vivo expansion, Tregs easily lose suppressive activity. Here, we followed expanding CD4(+)CD25(high)CD127(-) Tregs and their naive (CD45RA(+)) and memory-like (CD45RA(-)) subsets in order to establish the best conditions of the expansion. We found that, regardless of the phenotype sorted, expanding Tregs were undergoing changes resembling homeostatic proliferation and transformed into effector memory-like cells which produced not only suppressive interleukin-10 (IL-10) but also IL-6, IL-17, and interferon-γ (IFN-γ). With the time ex vivo, Tregs were losing the expression of FoxP3 and suppressive activity both when stimulated and when at rest. The only variable that helped preserve suppressive abilities of Tregs was the limitation of the time of ex vivo cultures to 2 weeks only. According to our study, the highest number of highly suppressive Tregs could be yielded with CD4(+)CD25(high)CD127(-) Tregs cultured no longer than 2 weeks. Thorough quality check, preferentially with the assessment of FoxP3 expression and IFN-γ suppression assay, should be applied to assess suppressive activity of the cells.
Collapse
Affiliation(s)
- Natalia Marek
- Department of Clinical Immunology and Transplantology, Medical University of Gdańsk, Gdańsk Poland
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Pothoven KL, Kheradmand T, Yang Q, Houlihan JL, Zhang H, DeGutes M, Miller SD, Luo X. Rapamycin-conditioned donor dendritic cells differentiate CD4CD25Foxp3 T cells in vitro with TGF-beta1 for islet transplantation. Am J Transplant 2010; 10:1774-84. [PMID: 20626386 PMCID: PMC3995630 DOI: 10.1111/j.1600-6143.2010.03199.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Dendritic cells (DCs) conditioned with the mammalian target of rapamycin (mTOR) inhibitor rapamycin have been previously shown to expand naturally existing regulatory T cells (nTregs). This work addresses whether rapamycin-conditioned donor DCs could effectively induce CD4(+)CD25(+)Foxp3(+) Tregs (iTregs) in cell cultures with alloantigen specificities, and whether such in vitro-differentiated CD4(+)CD25(+)Foxp3(+) iTregs could effectively control acute rejection in allogeneic islet transplantation. We found that donor BALB/c bone marrow-derived DCs (BMDCs) pharmacologically modified by the mTOR inhibitor rapamycin had significantly enhanced ability to induce CD4(+)CD25(+)Foxp3(+) iTregs of recipient origin (C57BL/6 (B6)) in vitro under Treg driving conditions compared to unmodified BMDCs. These in vitro-induced CD4(+)CD25(+)Foxp3(+) iTregs exerted donor-specific suppression in vitro, and prolonged allogeneic islet graft survival in vivo in RAG(-/-) hosts upon coadoptive transfer with T-effector cells. The CD4(+)CD25(+)Foxp3(+) iTregs expanded and preferentially maintained Foxp3 expression in the graft draining lymph nodes. Finally, the CD4(+)CD25(+)Foxp3(+) iTregs were further able to induce endogenous naïve T cells to convert to CD4(+)CD25(+)Foxp3(+) T cells. We conclude that rapamycin-conditioned donor BMDCs can be exploited for efficient in vitro differentiation of donor antigen-specific CD4(+)CD25(+)Foxp3(+) iTregs. Such in vitro-generated donor-specific CD4(+)CD25(+)Foxp3(+) iTregs are able to effectively control allogeneic islet graft rejection.
Collapse
Affiliation(s)
- K. L. Pothoven
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - T. Kheradmand
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Q. Yang
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - J. L. Houlihan
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - H. Zhang
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - M. DeGutes
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - S. D. Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - X. Luo
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL,Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL,Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL,Corresponding author: Xunrong Luo,
| |
Collapse
|
22
|
Boyd AS, Fairchild PJ. Approaches for immunological tolerance induction to stem cell-derived cell replacement therapies. Expert Rev Clin Immunol 2010; 6:435-48. [PMID: 20441429 DOI: 10.1586/eci.10.20] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The shortage of donors for organ transplantation and also to treat degenerative diseases has led to the development of the new field of regenerative medicine. One aim of this field, in addition to in vivo induction of endogenous tissue regeneration, is to utilize stem cells as a supplementary source of cells to repair or replace tissues or organs that have ceased to function owing to ageing or autoimmunity. Embryonic stem cells hold promise in this respect because of their developmental capacity to generate all tissues within the body. More recently, the discovery of induced pluripotent stem cells, somatic cells reprogrammed to a primitive embryonic-like state by the introduction of pluripotency factors, may also act as an important cell source for cell replacement therapy. However, before cell replacement therapy can become a reality, one must consider how to overcome the potential transplant rejection of stem cell-derived products. There are several potential ways to circumvent the hurdles presented by the immune system in this setting, not least the induction of immunological tolerance in the host. In this review, we consider this and other approaches for engendering acceptance of stem cell-derived tissues.
Collapse
Affiliation(s)
- Ashleigh S Boyd
- Stem Cell Sciences Lab, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX13RE, UK.
| | | |
Collapse
|
23
|
Abstract
Accumulating evidence from murine and human studies supports a key role for interleukin-17 (IL-17) and IL-21 in the pathogenesis of inflammatory arthritis. The pathways and molecular mechanisms that underlie the production of IL-17 and IL-21 are being rapidly elucidated. This review focuses on interferon regulatory factor 4 (IRF4), a member of the IRF family of transcription factors, which has emerged as a crucial controller of both IL-17 and IL-21 production. We first outline the complex role of IRF4 in the function of CD4(+) T cells and then discuss recent studies from our laboratory that have revealed a surprising role for components of Rho GTPase-mediated pathways in controlling the activity of IRF4. A better understanding of these novel pathways will hopefully provide new insights into mechanisms responsible for the development of inflammatory arthritis and potentially guide the design of novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Govind Bhagat
- Department of Pathology, Columbia University, New York, NY, USA
| | | |
Collapse
|
24
|
|
25
|
Cravedi P, Mannon RB. Noninvasive methods to assess the risk of kidney transplant rejection. Expert Rev Clin Immunol 2009; 5:535-546. [PMID: 20161000 PMCID: PMC2756773 DOI: 10.1586/eci.09.36] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In current clinical practice, immune reactivity of kidney transplant recipients is estimated by monitoring the levels of immunosuppressive drugs, and by functional and/or histological evaluation of the allograft. The availability of assays that could directly quantify the extent of the recipient's immune response towards the allograft would help clinicians to customize the prescription of immunosuppressive drugs to individual patients. Importantly, these assays might provide a more in-depth understanding of the complex mechanisms of acute rejection, chronic injury, and tolerance in organ transplantation, allowing the design of new and potentially more effective strategies for the minimization of immunosuppression, or even for the induction of immunological tolerance. The purpose of this review is to summarize results from recent studies in this field.
Collapse
Affiliation(s)
- Paolo Cravedi
- Mario Negri Institute for Pharmacological Research, Bergamo, Italy, Tel.: +39 035 453 5405, Fax: +39 035 453 5370,
| | - Roslyn B Mannon
- Division of Nephrology, Department of Medicine, 1900 University Boulevard, THT 611G, Birmingham, AL 35294, USA, Tel.: +1 205 996 6383, Fax: +1 205 996 6659,
| |
Collapse
|
26
|
Interleukin 7 receptor α as a potential therapeutic target in transplantation. Arch Immunol Ther Exp (Warsz) 2009; 57:253-61. [DOI: 10.1007/s00005-009-0036-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2008] [Accepted: 04/30/2009] [Indexed: 10/20/2022]
|
27
|
Abstract
T-cell depletion strategies are an efficient therapy for the treatment of acute rejection after organ transplantation and have been successfully used as induction regimens. Although eliminating whole T cells blocks alloreactivity, this therapy challenges the development of regulatory mechanisms because it depletes regulatory cells and modifies the profile of T cells after homeostatic repopulation. Targeting T-cell subpopulations or selectively activated T cells, without modifying Treg cells, could constitute a pro-tolerogenic approach. However, the perfect molecular target that would be totally specific probably still needs to be identified. In this study, we have reviewed the biological activities of broad or specific T-cell depletion strategies as these contribute to the induction of regulatory cells and tolerance in organ transplantation.
Collapse
Affiliation(s)
- Thomas Haudebourg
- INSERM, U643, CHU Nantes, Institut de Transplantation et de Recherche en Transplantation, ITERT, Université de Nantes, Faculté de Médecine, Nantes, France
| | | | | |
Collapse
|
28
|
From bench to bedside and back: the SENIEUR Protocol and the efficacy of influenza vaccination in the elderly. Biogerontology 2008; 10:83-94. [DOI: 10.1007/s10522-008-9155-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Accepted: 06/02/2008] [Indexed: 01/08/2023]
|
29
|
Riordan NH, Chan K, Marleau AM, Ichim TE. Cord blood in regenerative medicine: do we need immune suppression? J Transl Med 2007; 5:8. [PMID: 17261200 PMCID: PMC1796850 DOI: 10.1186/1479-5876-5-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Accepted: 01/30/2007] [Indexed: 12/31/2022] Open
Abstract
Cord blood is currently used as an alternative to bone marrow as a source of stem cells for hematopoietic reconstitution after ablation. It is also under intense preclinical investigation for a variety of indications ranging from stroke, to limb ischemia, to myocardial regeneration. A major drawback in the current use of cord blood is that substantial morbidity and mortality are associated with pre-transplant ablation of the recipient hematopoietic system. Here we raise the possibility that due to unique immunological properties of both the stem cell and non-stem cell components of cord blood, it may be possible to utilize allogeneic cells for regenerative applications without needing to fully compromise the recipient immune system. Issues raised will include: graft versus host potential, the immunogenicity of the cord blood graft, and the parallels between cord blood transplantation and fetal to maternal trafficking. The previous use of unmatched cord blood in absence of any immune ablation, as well as potential steps for widespread clinical implementation of allogeneic cord blood grafts will also be discussed.
Collapse
Affiliation(s)
| | - Kyle Chan
- Institute for Molecular Medicine, Huntington Beach, California, USA
| | | | | |
Collapse
|
30
|
Matei IR, Guidos CJ, Danska JS. ATM-dependent DNA damage surveillance in T-cell development and leukemogenesis: the DSB connection. Immunol Rev 2006; 209:142-58. [PMID: 16448540 DOI: 10.1111/j.0105-2896.2006.00361.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The immune system is capable of recognizing and eliminating an enormous array of pathogens due to the extremely diverse antigen receptor repertoire of T and B lymphocytes. However, the development of lymphocytes bearing receptors with unique specificities requires the generation of programmed double strand breaks (DSBs) coupled with bursts of proliferation, rendering lymphocytes susceptible to mutations contributing to oncogenic transformation. Consequently, mechanisms responsible for monitoring global genomic integrity must be activated during lymphocyte development to limit the oncogenic potential of antigen receptor locus recombination. Mutations in ATM (ataxia-telangiectasia mutated), a kinase that coordinates DSB monitoring and the response to DNA damage, result in impaired T-cell development and predispose to T-cell leukemia. Here, we review recent evidence providing insight into the mechanisms by which ATM promotes normal lymphocyte development and protects from neoplastic transformation.
Collapse
Affiliation(s)
- Irina R Matei
- Program in Developmental Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | | | | |
Collapse
|
31
|
Metcalfe SM, Calne RY, Cobbold SP. Introduction: immunoregulation: harnessing T cell biology for therapeutic benefit. Philos Trans R Soc Lond B Biol Sci 2005; 360:1641-3. [PMID: 16147527 PMCID: PMC1569538 DOI: 10.1098/rstb.2005.1709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Su M Metcalfe
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, UK.
| | | | | |
Collapse
|