101
|
Melamed A, Laydon DJ, Al Khatib H, Rowan AG, Taylor GP, Bangham CRM. HTLV-1 drives vigorous clonal expansion of infected CD8(+) T cells in natural infection. Retrovirology 2015; 12:91. [PMID: 26552867 PMCID: PMC4640420 DOI: 10.1186/s12977-015-0221-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 11/02/2015] [Indexed: 12/20/2022] Open
Abstract
Background Human T-lymphotropic Virus Type I (HTLV-1) is a retrovirus that persistently infects 5–10 million individuals worldwide and causes disabling or fatal inflammatory and malignant diseases. The majority of the HTLV-1 proviral load is found in CD4+ T cells, and the phenotype of adult T cell leukemia (ATL) is typically CD4+. HTLV-1 also infects CD8+ cells in vivo, but the relative abundance and clonal composition of the two infected subpopulations have not been studied. We used a high-throughput DNA sequencing protocol to map and quantify HTLV-1 proviral integration sites in separated populations of CD4+ cells, CD8+ cells and unsorted peripheral blood mononuclear cells from 12 HTLV-1-infected individuals. Results We show that the infected CD8+ cells constitute a median of 5 % of the HTLV-1 proviral load. However, HTLV-1-infected CD8+ clones undergo much greater oligoclonal proliferation than the infected CD4+ clones in infected individuals, regardless of disease manifestation. The CD8+ clones are over-represented among the most abundant clones in the blood and are redetected even after several years. Conclusions We conclude that although they make up only 5 % of the proviral load, the HTLV-1-infected CD8+ T-cells make a major impact on the clonal composition of HTLV-1-infected cells in the blood. The greater degree of oligoclonal expansion observed in the infected CD8+ T cells, contrasts with the CD4+ phenotype of ATL; cases of CD8+ adult T-cell leukaemia/lymphoma are rare. This work is consistent with growing evidence that oligoclonal expansion of HTLV-1-infected cells is not sufficient for malignant transformation. Electronic supplementary material The online version of this article (doi:10.1186/s12977-015-0221-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anat Melamed
- Section of Virology, Imperial College London, Wright-Fleming Institute, Norfolk Place, London, W2 1PG, UK.
| | - Daniel J Laydon
- Section of Virology, Imperial College London, Wright-Fleming Institute, Norfolk Place, London, W2 1PG, UK.
| | - Hebah Al Khatib
- Section of Virology, Imperial College London, Wright-Fleming Institute, Norfolk Place, London, W2 1PG, UK.
| | - Aileen G Rowan
- Section of Virology, Imperial College London, Wright-Fleming Institute, Norfolk Place, London, W2 1PG, UK.
| | - Graham P Taylor
- Section of Virology, Imperial College London, Wright-Fleming Institute, Norfolk Place, London, W2 1PG, UK.
| | - Charles R M Bangham
- Section of Virology, Imperial College London, Wright-Fleming Institute, Norfolk Place, London, W2 1PG, UK.
| |
Collapse
|
102
|
Tomala J, Kovar M. IL-2/anti-IL-2 mAb immunocomplexes: A renascence of IL-2 in cancer immunotherapy? Oncoimmunology 2015; 5:e1102829. [PMID: 27141363 DOI: 10.1080/2162402x.2015.1102829] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 09/28/2015] [Indexed: 10/22/2022] Open
Abstract
The in vivo biological activity of IL-2 can be dramatically increased by complexing with anti-IL-2 mAb. Moreover, IL-2/anti-IL-2 mAb immunocomplexes selectively stimulate different subsets of immune cells, depending on the clone of anti-IL-2 mAb that is used. Thus, IL-2/S4B6 mAb complexes strongly stimulate CD122high populations, namely NK and memory CD8+ T cells. They also intermediately stimulate Treg cells. Conversely, IL-2/JES6.1 mAb immunocomplexes have no stimulatory activity for CD122high populations. However, they potently and highly selectively stimulate CD25+ cells (i.e., Treg and activated T cells). IL-2/S4B6 mAb immunocomplexes have also been shown to possess antitumor activity in various mouse tumor models.
Collapse
Affiliation(s)
- Jakub Tomala
- Laboratory of Tumor Immunology, Institute of Microbiology, Academy of Sciences of the Czech Republic , Prague, Czech Republic
| | - Marek Kovar
- Laboratory of Tumor Immunology, Institute of Microbiology, Academy of Sciences of the Czech Republic , Prague, Czech Republic
| |
Collapse
|
103
|
Liu J, Chen D, Nie GD, Dai Z. CD8(+)CD122(+) T-Cells: A Newly Emerging Regulator with Central Memory Cell Phenotypes. Front Immunol 2015; 6:494. [PMID: 26539191 PMCID: PMC4610204 DOI: 10.3389/fimmu.2015.00494] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/10/2015] [Indexed: 11/16/2022] Open
Abstract
CD8(+)CD122(+) T-cells have been traditionally described as antigen-specific memory T-cells that respond to previously encountered antigens more quickly and vigorously than their naïve counterparts. However, mounting evidence has demonstrated that murine CD8(+)CD122(+) T-cells exhibit a central memory phenotype (CD44(high)CD62L(high)), regulate T cell homeostasis, and act as regulatory T-cells (Treg) by suppressing both autoimmune and alloimmune responses. Importantly, naturally occurring murine CD8(+)CD122(+) Tregs are more potent in immunosuppression than their CD4(+)CD25(+) counterparts. They appear to be acting in an antigen-non-specific manner. Human CD8(+)CXCR3(+) T-cells are the equivalent of murine CD8(+)CD122(+) Tregs and also exhibit central memory phenotypes. In this mini-review article, we will summarize recent progresses in their phenotypes, homeostatic expansion, antigen-specificity, roles in the suppression of alloimmune and autoimmune responses, and the mechanisms underlying their inhibitory function.
Collapse
Affiliation(s)
- Junfeng Liu
- Section of Immunology, Division of Dermatology, Second Affiliated Hospital, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dacan Chen
- Section of Immunology, Division of Dermatology, Second Affiliated Hospital, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Golay D. Nie
- School of Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Zhenhua Dai
- Section of Immunology, Division of Dermatology, Second Affiliated Hospital, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
104
|
Synchronizing transcriptional control of T cell metabolism and function. Nat Rev Immunol 2015; 15:574-84. [DOI: 10.1038/nri3874] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
105
|
Richer MJ, Pewe LL, Hancox LS, Hartwig SM, Varga SM, Harty JT. Inflammatory IL-15 is required for optimal memory T cell responses. J Clin Invest 2015; 125:3477-90. [PMID: 26241055 DOI: 10.1172/jci81261] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 06/18/2015] [Indexed: 12/22/2022] Open
Abstract
Due to their ability to rapidly proliferate and produce effector cytokines, memory CD8+ T cells increase protection following reexposure to a pathogen. However, low inflammatory immunizations do not provide memory CD8+ T cells with a proliferation advantage over naive CD8+ T cells, suggesting that cell-extrinsic factors enhance memory CD8+ T cell proliferation in vivo. Herein, we demonstrate that inflammatory signals are critical for the rapid proliferation of memory CD8+ T cells following infection. Using murine models of viral infection and antigen exposure, we found that type I IFN-driven expression of IL-15 in response to viral infection prepares memory CD8+ T cells for rapid division independently of antigen reexposure by transiently inducing cell-cycle progression via a pathway dependent on mTOR complex-1 (mTORC1). Moreover, exposure to IL-15 allowed more rapid division of memory CD8+ T cells following antigen encounter and enhanced their protective capacity against viral infection. Together, these data reveal that inflammatory IL-15 promotes optimal responses by memory CD8+ T cells.
Collapse
|
106
|
Echeverría A, Moro-García MA, Asensi V, Cartón JA, López-Larrea C, Alonso-Arias R. CD4⁺CD28null T lymphocytes resemble CD8⁺CD28null T lymphocytes in their responses to IL-15 and IL-21 in HIV-infected patients. J Leukoc Biol 2015; 98:373-84. [PMID: 26034206 DOI: 10.1189/jlb.1a0514-276rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 05/01/2015] [Indexed: 01/09/2023] Open
Abstract
HIV-infected individuals suffer from accelerated immunologic aging. One of the most prominent changes during T lymphocyte aging is the accumulation of CD28(null) T lymphocytes, mainly CD8(+) but also CD4(+) T lymphocytes. Enhancing the functional properties of these cells may be important because they provide antigen-specific defense against chronic infections. The objective of this study was to compare the responses of CD4(+)CD28(null) and CD8(+)CD28(null) T lymphocytes from HIV-infected patients to the immunomodulatory effects of cytokines IL-15 and IL-21. We quantified the frequencies of CD4(+)CD28(null) and CD8(+)CD28(null) T lymphocytes in peripheral blood from 110 consecutive, HIV-infected patients and 25 healthy controls. Patients showed increased frequencies of CD4(+)CD28(null) and CD8(+)CD28(null). Both subsets were positively correlated to each other and showed an inverse correlation with the absolute counts of CD4(+) T lymphocytes. Higher frequencies of HIV-specific and CMV-specific cells were found in CD28(null) than in CD28(+) T lymphocytes. Activation of STAT5 by IL-15 and STAT3 by IL-21 was higher in CD28(null) compared with CD28(+) T lymphocytes. Proliferation, expression of CD69, and IFN-γ production in CD28(null) T lymphocytes were increased after treatment with IL-15, and IL-21 potentiated most of those effects. Nevertheless, IL-21 alone reduced IFN-γ production in response to anti-CD3 stimulation but increased CD28 expression, even counteracting the inhibitory effect of IL-15. Intracytoplasmic stores of granzyme B and perforin were increased by IL-15, whereas IL-21 and simultaneous treatment with the 2 cytokines also significantly enhanced degranulation in CD4(+)CD28(null) and CD8(+)CD28(null) T lymphocytes. IL-15 and IL-21 could have a role in enhancing the effector response of CD28(null) T lymphocytes against their specific chronic antigens in HIV-infected patients.
Collapse
Affiliation(s)
- Ainara Echeverría
- *Immunology Department and Infectious Diseases Unit, Hospital Universitario Central de Asturias, Oviedo, Spain; and Fundación Renal "Iñigo Alvarez de Toledo," Madrid, Spain
| | - Marco A Moro-García
- *Immunology Department and Infectious Diseases Unit, Hospital Universitario Central de Asturias, Oviedo, Spain; and Fundación Renal "Iñigo Alvarez de Toledo," Madrid, Spain
| | - Víctor Asensi
- *Immunology Department and Infectious Diseases Unit, Hospital Universitario Central de Asturias, Oviedo, Spain; and Fundación Renal "Iñigo Alvarez de Toledo," Madrid, Spain
| | - José A Cartón
- *Immunology Department and Infectious Diseases Unit, Hospital Universitario Central de Asturias, Oviedo, Spain; and Fundación Renal "Iñigo Alvarez de Toledo," Madrid, Spain
| | - Carlos López-Larrea
- *Immunology Department and Infectious Diseases Unit, Hospital Universitario Central de Asturias, Oviedo, Spain; and Fundación Renal "Iñigo Alvarez de Toledo," Madrid, Spain
| | - Rebeca Alonso-Arias
- *Immunology Department and Infectious Diseases Unit, Hospital Universitario Central de Asturias, Oviedo, Spain; and Fundación Renal "Iñigo Alvarez de Toledo," Madrid, Spain
| |
Collapse
|
107
|
Villanueva JE, Malle EK, Gardam S, Silveira PA, Zammit NW, Walters SN, Brink R, Grey ST. TRAF2 regulates peripheral CD8(+) T-cell and NKT-cell homeostasis by modulating sensitivity to IL-15. Eur J Immunol 2015; 45:1820-31. [PMID: 25931426 DOI: 10.1002/eji.201445416] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/18/2015] [Accepted: 04/28/2015] [Indexed: 11/07/2022]
Abstract
In this study, a critical and novel role for TNF receptor (TNFR) associated factor 2 (TRAF2) is elucidated for peripheral CD8(+) T-cell and NKT-cell homeostasis. Mice deficient in TRAF2 only in their T cells (TRAF2TKO) show ∼40% reduction in effector memory and ∼50% reduction in naïve CD8(+) T-cell subsets. IL-15-dependent populations were reduced further, as TRAF2TKO mice displayed a marked ∼70% reduction in central memory CD8(+) CD44(hi) CD122(+) T cells and ∼80% decrease in NKT cells. TRAF2TKO CD8(+) CD44(hi) T cells exhibited impaired dose-dependent proliferation to exogenous IL-15. In contrast, TRAF2TKO CD8(+) T cells proliferated normally to anti-CD3 and TRAF2TKO CD8(+) CD44(hi) T cells exhibited normal proliferation to exogenous IL-2. TRAF2TKO CD8(+) T cells expressed normal levels of IL-15-associated receptors and possessed functional IL-15-mediated STAT5 phosphorylation, however TRAF2 deletion caused increased AKT activation. Loss of CD8(+) CD44(hi) CD122(+) and NKT cells was mechanistically linked to an inability to respond to IL-15. The reduced CD8(+) CD44(hi) CD122(+) T-cell and NKT-cell populations in TRAF2TKO mice were rescued in the presence of high dose IL-15 by IL-15/IL-15Rα complex administration. These studies demonstrate a critical role for TRAF2 in the maintenance of peripheral CD8(+) CD44(hi) CD122(+) T-cell and NKT-cell homeostasis by modulating sensitivity to T-cell intrinsic growth factors such as IL-15.
Collapse
Affiliation(s)
| | | | - Sandra Gardam
- B cell Biology Group, Immunology Division, Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | | | | | - Robert Brink
- B cell Biology Group, Immunology Division, Garvan Institute of Medical Research, Darlinghurst, Australia
| | | |
Collapse
|
108
|
Wojta-Stremayr D, Neunkirchner A, Srinivasan B, Trapin D, Schmetterer KG, Pickl WF. CD8+ T Cell Fate and Function Influenced by Antigen-Specific Virus-Like Nanoparticles Co-Expressing Membrane Tethered IL-2. PLoS One 2015; 10:e0126034. [PMID: 25946103 PMCID: PMC4422701 DOI: 10.1371/journal.pone.0126034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 03/27/2015] [Indexed: 01/21/2023] Open
Abstract
A variety of adjuvants fostering humoral immunity are known as of today. However, there is a lack of adjuvants or adjuvant strategies, which directly target T cellular effector functions and memory. We here determined whether systemically toxic cytokines such as IL-2 can be restricted to the site of antigen presentation and used as ‘natural adjuvants’. Therefore, we devised antigen-presenting virus-like nanoparticles (VNP) co-expressing IL-2 attached to different membrane-anchors and assessed their potency to modulate CD8+ T cell responses in vitro and in vivo. Efficient targeting of IL-2 to lipid rafts and ultimately VNP was achieved by fusing IL-2 at its C-terminus to a minimal glycosylphosphatidylinositol (GPI)-anchor acceptor sequence. To identify optimal membrane-anchor dimensions we inserted one (1Ig), two (2Ig) or four (4Ig) immunoglobulin(Ig)-like domains of CD16b between IL-2 and the minimal GPI-anchor acceptor sequence of CD16b (GPI). We found that the 2IgGPI version was superior to all other evaluated IL-2 variants (IL-2v) in terms of its i) degree of targeting to lipid rafts and to the VNP surface, ii) biological activity, iii) co-stimulation of cognate T cells in the absence of bystander activation and iv) potency to induce differentiation and acquisition of CD8+ T cell effector functions in vitro and in vivo. In contrast, the GPI version rather favored memory precursor cell formation. These results exemplify novel beneficial features of membrane-bound IL-2, which in addition to its mere T cell stimulatory capacity include the induction of differential effector and memory functions in CD8+ T lymphocytes.
Collapse
Affiliation(s)
- Daniela Wojta-Stremayr
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Immunomodulation, Vienna, Austria
| | - Alina Neunkirchner
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Immunomodulation, Vienna, Austria
| | - Bharani Srinivasan
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Doris Trapin
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Klaus G Schmetterer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Winfried F Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Immunomodulation, Vienna, Austria
| |
Collapse
|
109
|
Wang Y, Lavender P, Watson J, Arno M, Lehner T. Stress-activated Dendritic Cells (DC) Induce Dual Interleukin (IL)-15- and IL1β-mediated Pathways, Which May Elicit CD4+ Memory T Cells and Interferon (IFN)-stimulated Genes. J Biol Chem 2015; 290:15595-15609. [PMID: 25907558 DOI: 10.1074/jbc.m115.645754] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Indexed: 01/03/2023] Open
Abstract
The prevailing evidence suggests that immunological memory does not require antigenic re-stimulation but is maintained by low level tonic stimulation. We examined the hypothesis that stress agents contribute to tonic cellular activation and maintain immunological memory. Stimulation of monocyte-derived dendritic cells (DC) with stress agents elicits reactive oxygen species and HSP70. NFκB is activated, which up-regulates membrane-associated (ma) IL-15, caspase-1 and IL-1β. Co-culture of stress-treated DC with mononuclear cells activates IL-15 and IL-1β receptors on CD4(+) T cells, eliciting CD40L, proliferation, and up-regulation of CD45RO(+) memory T cells. The transcription factors Tbet(high) and RORγt are up-regulated, whereas FoxP3 is down-regulated, resulting in enhanced Th1 and Th17 expression and the corresponding cytokines. The interaction between maIL-15 expressed by DC and IL-15R on CD4(+) T cells results in one pathway and the corresponding cells expressing IL-1β and IL1βR as a second pathway. Importantly, inhibition studies with IL-15 antibodies and IL-1βR inhibitor suggest that both pathways may be required for optimum CD4(+) CD45RO(+) memory T cell expression. Type 1 IFN expression in splenic CD11c DC of stress-treated mice demonstrated a significant increase of IFN-α in CD11c CD317(+) and CD8α(+) DC. Analysis of RNA in human CD4(+) memory T cells showed up-regulation of type 1 IFN-stimulated genes and inhibition with histone methyltransferase inhibitor. We suggest the paradigm that stress-induced tonic stimulation might be responsible for the robust persistence of the immune response in vaccination and that epigenetic changes are involved in maintaining CD4(+) T cell memory.
Collapse
Affiliation(s)
- Yufei Wang
- Mucosal Immunology Unit, Kings College London, London SE1 1UL, United Kingdom
| | - Paul Lavender
- MRC, and Asthma UK Centre in Allergic Mechanisms of Asthma, Kings College London, London SE1 1UL, United Kingdom
| | - Julie Watson
- MRC, and Asthma UK Centre in Allergic Mechanisms of Asthma, Kings College London, London SE1 1UL, United Kingdom
| | - Matthew Arno
- Genomics Centre, Kings College London, London SE1 1UL, United Kingdom
| | - Thomas Lehner
- Mucosal Immunology Unit, Kings College London, London SE1 1UL, United Kingdom.
| |
Collapse
|
110
|
Ghosh AK, Sinha D, Mukherjee S, Biswas R, Biswas T. LPS stimulates and Hsp70 down-regulates TLR4 to orchestrate differential cytokine response of culture-differentiated innate memory CD8(+) T cells. Cytokine 2015; 73:44-52. [PMID: 25697138 DOI: 10.1016/j.cyto.2015.01.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 01/13/2015] [Accepted: 01/15/2015] [Indexed: 01/04/2023]
Abstract
Nonconventional innate memory CD8(+) T cells characteristically expressing CD44, CD122, eomesodermin (Eomes) and promyelocytic leukemia zinc finger (PLZF) were derived in culture from CD4(+)CD8(+) double positive (DP) thymocytes of normal BALB/c and C57BL/6 mice. These culture-differentiated cells constitutively express toll-like receptor (TLR)4 and release interferon (IFN)-γ and interleukin (IL)-10. We show the TLR4-ligand lipopolysaccharide (LPS) stimulate the TLR and up-regulate IFN-γ skewing the cells towards type 1 polarization. In presence of LPS these cells also express suppressor of cytokine signaling (SOCS)1 and thus suppress IL-10 expression. In contrast, heat shock protein (Hsp)70 down-regulated TLR4 augmenting the anti-inflammatory cytokine IL-10. In association with IL-10 release IFN-γ was abrogated. The programmed cell death (PD)-1 mostly present in regulatory T cells was stimulated in these IL-10 producing cells by Hsp70 and not LPS indicating the cells can be driven to two contrast outcomes by the two TLR4 ligands. Our work provides a scope for in vitro monitoring of CD8(+) T cells to decipher important immune therapeutic option during infection or sepsis.
Collapse
Affiliation(s)
- Amlan Kanti Ghosh
- Division of Immunology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Debolina Sinha
- Division of Immunology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Subhadeep Mukherjee
- Division of Immunology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Ratna Biswas
- Division of Immunology, National Institute of Cholera and Enteric Diseases, Kolkata, India.
| | - Tapas Biswas
- Division of Immunology, National Institute of Cholera and Enteric Diseases, Kolkata, India.
| |
Collapse
|
111
|
Abstract
Interleukin-15 (IL-15) exerts many biological functions essential for the maintenance and function of multiple cell types. Although its expression is tightly regulated, IL-15 upregulation has been reported in many organ-specific autoimmune disorders. In celiac disease, an intestinal inflammatory disorder driven by gluten exposure, the upregulation of IL-15 expression in the intestinal mucosa has become a hallmark of the disease. Interestingly, because it is overexpressed both in the gut epithelium and in the lamina propria, IL-15 acts on distinct cell types and impacts distinct immune components and pathways to disrupt intestinal immune homeostasis. In this article, we review our current knowledge of the multifaceted roles of IL-15 with regard to the main immunological processes involved in the pathogenesis of celiac disease.
Collapse
Affiliation(s)
- Valérie Abadie
- Sainte-Justine Hospital Research Center, Department of Microbiology and Immunology, Faculty of Medicine, University of Montreal, Montreal, Canada
| | | |
Collapse
|
112
|
Hendricks DW, Min-Oo G, Lanier LL. Sweet Is the Memory of Past Troubles: NK Cells Remember. Curr Top Microbiol Immunol 2015; 395:147-71. [PMID: 26099194 DOI: 10.1007/82_2015_447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Natural killer (NK) cells are important in host defense against tumors and microbial pathogens. Recent studies indicate that NK cells share many features with the adaptive immune system, and like B cells and T cells, NK cells can acquire immunological memory. Here, we review evidence for NK cell memory and the molecules involved in the generation and maintenance of these self-renewing NK cells that provide enhanced protection of the host.
Collapse
Affiliation(s)
- Deborah W Hendricks
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, 94143-0414, USA
| | - Gundula Min-Oo
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, 94143-0414, USA
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, 94143-0414, USA.
| |
Collapse
|
113
|
Sharma RK, Yolcu ES, Shirwan H. SA-4-1BBL as a novel adjuvant for the development of therapeutic cancer vaccines. Expert Rev Vaccines 2014; 13:387-98. [PMID: 24521311 DOI: 10.1586/14760584.2014.880340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor associated antigen (TAA)-based therapeutic vaccines have great potential as a safe, practical, and cost-efficient alternative to standard treatments for cancer. Clinical efficacy of TAA-based vaccines, however, has yet to be realized and will require adjuvants with pleiotropic functions on immune cells. Such adjuvants need not only to generate/boost T cell responses, but also reverse intrinsic/extrinsic tumor immune evasion mechanisms for therapeutic efficacy. This review focuses on a novel agonistic ligand, SA-4-1BBL, for 4-1BB costimulatory receptor as an adjuvant of choice because of its ability to: i) serve as a vehicle to deliver TAAs to dendritic cells (DCs) for antigen uptake and cross-presentation to CD8(+) T cells; ii) augment adaptive Th1 and innate immune responses; and iii) overcome various immune evasion mechanisms, cumulatively translating into therapeutic efficacy in preclinical tumor models.
Collapse
Affiliation(s)
- Rajesh K Sharma
- Department of Microbiology and Immunology, Institute for Cellular Therapeutics, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | | | | |
Collapse
|
114
|
Downregulation of endogenous STAT3 augments tumoricidal activity of interleukin 15 activated dendritic cell against lymphoma and leukemia via TRAIL. Exp Cell Res 2014; 327:192-208. [DOI: 10.1016/j.yexcr.2014.08.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 08/04/2014] [Accepted: 08/08/2014] [Indexed: 12/22/2022]
|
115
|
Krzych U, Zarling S, Pichugin A. Memory T cells maintain protracted protection against malaria. Immunol Lett 2014; 161:189-95. [PMID: 24709142 PMCID: PMC6499475 DOI: 10.1016/j.imlet.2014.03.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 03/25/2014] [Accepted: 03/27/2014] [Indexed: 10/25/2022]
Abstract
Immunologic memory is one of the cardinal features of antigen-specific immune responses, and the persistence of memory cells contributes to prophylactic immunizations against infectious agents. Adequately maintained memory T and B cell pools assure a fast, effective and specific response against re-infections. However, many aspects of immunologic memory are still poorly understood, particularly immunologic memory inducible by parasites, for example, Plasmodium spp., the causative agents of malaria. For example, memory responses to Plasmodium antigens amongst residents of malaria endemic areas appear to be either inadequately developed or maintained, because persons who survive episodes of childhood malaria remain vulnerable to intermittent malaria infections. By contrast, multiple exposures of humans and laboratory rodents to radiation-attenuated Plasmodium sporozoites (γ-spz) induce sterile and long-lasting protection against experimental sporozoite challenge. Multifactorial immune mechanisms maintain this protracted and sterile protection. While the presence of memory CD4 T cell subsets has been associated with lasting protection in humans exposed to multiple bites from Anopheles mosquitoes infected with attenuated Plasmodium falciparum, memory CD8 T cells maintain protection induced with Plasmodium yoelii and Plasmodium berghei γ-spz in murine models. In this review, we discuss our observations that show memory CD8 T cells specific for antigens expressed by P. berghei liver stage parasites as an indispensable component for the maintenance of protracted protective immunity against experimental malaria infection; moreover, the provision of an Ag-depot assures a quick recall of memory T cells as IFN-γ-producing effector CD8 T cells and IL-4- producing CD4 T cells that collaborate with B cells for an effective antibody response.
Collapse
Affiliation(s)
- Urszula Krzych
- Department of Cellular Immunology, Branch of Malaria Vaccine Development, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States.
| | - Stasya Zarling
- Department of Cellular Immunology, Branch of Malaria Vaccine Development, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States
| | - Alexander Pichugin
- Department of Cellular Immunology, Branch of Malaria Vaccine Development, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States
| |
Collapse
|
116
|
Chen X, Ni J, Meng H, Li D, Wei Y, Luo Y, Wu Y. Interleukin‑15: a potent adjuvant enhancing the efficacy of an autologous whole‑cell tumor vaccine against Lewis lung carcinoma. Mol Med Rep 2014; 10:1828-34. [PMID: 25109355 DOI: 10.3892/mmr.2014.2474] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 02/18/2014] [Indexed: 02/05/2023] Open
Abstract
Lung cancer is a major cause of cancer‑associated mortality worldwide due to its limited response rate to current chemotherapy and radiation, thus immunotherapy is rapidly becoming the most promising approach. Although the highly specific tumor‑associated antigen of lung cancer has been found, autologous whole‑cell tumor vaccines remain indispensable in the development of therapeutic cancer vaccines. Interleukin (IL)‑15 is a T helper type 1 cytokine that has been demonstrated to have a marked antitumor immune response and the potential ability to reverse the host tolerance of tumor antigens in certain preclinical trials. In the present study, a cationic liposome encapsulating IL‑15 gene‑loaded plasmid acted as an adjuvant of an autologous whole‑cell tumor vaccine by subcutaneous injection. The combination immunotherapy resulted in significant inhibition of tumor growth without side effects in the preventive tumor inhibition and adoptive therapy study. Cytotoxic lymphocyte assay detection of the serum antigen and cytokines using an enzyme‑linked immunosorbent assay suggested that the IL‑15 gene can significantly improve the cellular immune response and humoral immune response provoked by autologous whole‑cell tumor vaccines. These results demonstrated that the IL‑15 gene was an effective adjuvant of autologous whole‑cell tumor vaccines against mouse lung cancer and may provide an attractive vaccine strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Xi Chen
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jie Ni
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hui Meng
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Dandan Li
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan Luo
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yang Wu
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
117
|
Díaz-Torné C, Ortiz de Juana MA, Geli C, Cantó E, Laiz A, Corominas H, Casademont J, de Llobet JM, Juárez C, Díaz-López C, Vidal S. Rituximab-induced interleukin-15 reduction associated with clinical improvement in rheumatoid arthritis. Immunology 2014; 142:354-362. [PMID: 24219764 PMCID: PMC4080951 DOI: 10.1111/imm.12212] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Revised: 10/24/2013] [Accepted: 11/07/2013] [Indexed: 12/21/2022] Open
Abstract
Rituximab therapy alters all aspects of B-cell participation in the disturbed immune response of rheumatoid arthritis patients. To determine the impact of B-cell depletion on other immune compartments, we analysed levels of soluble and surface interleukin-15 (IL-15) along with the frequency of IL-15-related subsets after rituximab treatment. We then studied the correlation of observed changes with clinical activity. Heparinized blood samples from 33 rheumatoid arthritis patients were collected on days 0, 30, 90 and 180 after each of three rituximab cycles. Serum cytokine levels were determined by ELISA. Interleukin-15 trans-presentation was analysed by cytometry. Flow cytometry with monoclonal antibodies was performed to analyse circulating cell subsets. Interleukin-15 was detected in the serum of 25 patients before initiating the treatment. Rituximab then progressively reduced serum IL-15 (138 ± 21 pg/ml at baseline, 48 ± 18 pg/ml after third cycle, P = 0·03) along with IL-17 (1197 ± 203 pg/ml at baseline, 623 ± 213 pg/ml after third cycle, P = 0·03) and tended to increase the frequency of circulating regulatory T cells (3·1 ± 1 cells/μl at baseline, 7·7 ± 2 cells/μl after third cycle). Rituximab also significantly decreased IL-15 trans-presentation on surface monocytes of patients negative for IL-15 serum (mean fluorescence intensity: 4·82 ± 1·30 at baseline, 1·42 ± 0·69 after third cycle P = 0·05). Reduction of serum IL-15 was associated with decrease in CD8(+) CD45RO(+) /RA(+) ratio (1·17 ± 0·21 at baseline, 0·36 ± 0·06 at third cycle, P = 0·02). DAS28, erythrocyte sedimentation rate and C-reactive protein correlated significantly with CD8(+) CD45RO(+) /RA(+) ratio (R = 0·323, R = 0·357, R = 0·369 respectively, P < 0·001). Our results suggest that sustained clinical improvement after rituximab treatment is associated with IL-15/memory T-cell-related mechanisms beyond circulating B cells.
Collapse
Affiliation(s)
- César Díaz-Torné
- Rheumatology Unit, Internal Medicine Department Hospital Sant Pau, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Li S, Xie Q, Zeng Y, Zou C, Liu X, Wu S, Deng H, Xu Y, Li XC, Dai Z. A naturally occurring CD8(+)CD122(+) T-cell subset as a memory-like Treg family. Cell Mol Immunol 2014; 11:326-31. [PMID: 24793406 PMCID: PMC4085522 DOI: 10.1038/cmi.2014.25] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 03/19/2014] [Accepted: 03/19/2014] [Indexed: 01/07/2023] Open
Abstract
Despite extensive studies on CD4(+)CD25(+) regulatory T cells (Tregs) during the past decade, the progress on their clinical translation remains stagnant. Mounting evidence suggests that naturally occurring CD8(+)CD122(+) T cells are also Tregs with the capacity to inhibit T-cell responses and suppress autoimmunity as well as alloimmunity. In fact, they are memory-like Tregs that resemble a central memory T cell (TCM) phenotype. The mechanisms underlying their suppression are still not well understood, although they may include IL-10 production. We have recently demonstrated that programmed death-1 (PD-1) expression distinguishes between regulatory and memory CD8(+)CD122(+) T cells and that CD8(+)CD122(+) Tregs undergo faster homeostatic proliferation and are more potent in the suppression of allograft rejection than conventional CD4(+)CD25(+) Tregs. These findings may open a new line of investigation for accelerating effective Treg therapies in the clinic. In this review, we summarize the significant progress in this promising field of CD8(+)CD122(+) Treg research and discuss their phenotypes, suppressive roles in autoimmunity and alloimmunity, functional requirements, mechanisms of action and potential applications in the clinic.
Collapse
Affiliation(s)
- Shanshan Li
- Section of Immunology, Center for Regenerative and Translational Medicine
| | - Qingfeng Xie
- Section of Immunology, Center for Regenerative and Translational Medicine
| | - Yuqun Zeng
- Department of Nephrology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Chuan Zou
- Department of Nephrology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Xusheng Liu
- Department of Nephrology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Shouhai Wu
- Section of Immunology, Center for Regenerative and Translational Medicine
| | - Haixia Deng
- Section of Immunology, Center for Regenerative and Translational Medicine
| | - Yang Xu
- Section of Immunology, Center for Regenerative and Translational Medicine
| | - Xian C Li
- Immunobiology and Transplantation Research Center, Houston Methodist Research Institute, Houston, TX, USA
| | - Zhenhua Dai
- Section of Immunology, Center for Regenerative and Translational Medicine
| |
Collapse
|
119
|
Abstract
Interleukin-15 (IL-15) exerts many biological functions essential for the maintenance and function of multiple cell types. Although its expression is tightly regulated, IL-15 upregulation has been reported in many organ-specific autoimmune disorders. In celiac disease, an intestinal inflammatory disorder driven by gluten exposure, the upregulation of IL-15 expression in the intestinal mucosa has become a hallmark of the disease. Interestingly, because it is overexpressed both in the gut epithelium and in the lamina propria, IL-15 acts on distinct cell types and impacts distinct immune components and pathways to disrupt intestinal immune homeostasis. In this article, we review our current knowledge of the multifaceted roles of IL-15 with regard to the main immunological processes involved in the pathogenesis of celiac disease.
Collapse
Affiliation(s)
- Valérie Abadie
- Sainte-Justine Hospital Research Center, Department of Microbiology and Immunology, Faculty of Medicine, University of Montreal, Montreal, Canada
| | | |
Collapse
|
120
|
Duga B, Czako M, Komlosi K, Hadzsiev K, Torok K, Sumegi K, Kisfali P, Kosztolanyi G, Melegh B. Deletion of 4q28.3-31.23 in the background of multiple malformations with pulmonary hypertension. Mol Cytogenet 2014; 7:36. [PMID: 24959202 PMCID: PMC4066825 DOI: 10.1186/1755-8166-7-36] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 05/23/2014] [Indexed: 11/16/2022] Open
Abstract
The 4q deletion syndrome shows a broad spectrum of clinical manifestations consisting of key features comprising growth failure, developmental delay, craniofacial dysmorphism, digital anomalies, and cardiac and skeletal defects. We have identified a de novo interstitial distal deletion in a 9 month-old girl with growth failure, developmental delay, ventricular septum defect in the subaortic region, patent foramen ovale and patent ductus arteriosus, vascular malformation of the lung, dysgenesis of the corpus callosum and craniofacial dysmorphism using array-comparative genomic hybridization. This de novo deletion is located at 4q28.3-31.23 (136,127,048 - 150,690,325), its size is 14.56 Mb, and contains 8 relevant genes (PCDH18, SETD7, ELMOD2, IL15, GAB1, HHIP, SMAD1, NR3C2) with possible contributions to the phenotype. Among other functions, a role in lung morphogenesis and tubulogenesis can be attributed to the deleted genes in our patient, which may explain the unique feature of vascular malformation of the lung leading to pulmonary hypertension. With the detailed molecular characterization of our case with 4q- syndrome we hope to contribute to the elucidation of the genetic spectrum of this disorder.
Collapse
Affiliation(s)
- Balazs Duga
- Department of Medical Genetics, Clinical Centre, University of Pecs, Szigeti 12, Pecs H-7624, Hungary ; Szentágothai Research Centre, University of Pecs, Ifjusag 20, Pecs H-7624, Hungary
| | - Marta Czako
- Department of Medical Genetics, Clinical Centre, University of Pecs, Szigeti 12, Pecs H-7624, Hungary ; Szentágothai Research Centre, University of Pecs, Ifjusag 20, Pecs H-7624, Hungary
| | - Katalin Komlosi
- Department of Medical Genetics, Clinical Centre, University of Pecs, Szigeti 12, Pecs H-7624, Hungary ; Szentágothai Research Centre, University of Pecs, Ifjusag 20, Pecs H-7624, Hungary
| | - Kinga Hadzsiev
- Department of Medical Genetics, Clinical Centre, University of Pecs, Szigeti 12, Pecs H-7624, Hungary ; Szentágothai Research Centre, University of Pecs, Ifjusag 20, Pecs H-7624, Hungary
| | - Katalin Torok
- Department of Pediatrics, Clinical Centre, University of Pecs, Jozsef Attila 7, Pecs H-7623, Hungary
| | - Katalin Sumegi
- Department of Medical Genetics, Clinical Centre, University of Pecs, Szigeti 12, Pecs H-7624, Hungary ; Szentágothai Research Centre, University of Pecs, Ifjusag 20, Pecs H-7624, Hungary
| | - Peter Kisfali
- Department of Medical Genetics, Clinical Centre, University of Pecs, Szigeti 12, Pecs H-7624, Hungary ; Szentágothai Research Centre, University of Pecs, Ifjusag 20, Pecs H-7624, Hungary
| | - Gyorgy Kosztolanyi
- Department of Medical Genetics, Clinical Centre, University of Pecs, Szigeti 12, Pecs H-7624, Hungary ; Szentágothai Research Centre, University of Pecs, Ifjusag 20, Pecs H-7624, Hungary
| | - Bela Melegh
- Department of Medical Genetics, Clinical Centre, University of Pecs, Szigeti 12, Pecs H-7624, Hungary ; Szentágothai Research Centre, University of Pecs, Ifjusag 20, Pecs H-7624, Hungary
| |
Collapse
|
121
|
Traitanon O, Gorbachev A, Bechtel JJ, Keslar KS, Baldwin WM, Poggio ED, Fairchild RL. IL-15 induces alloreactive CD28(-) memory CD8 T cell proliferation and CTLA4-Ig resistant memory CD8 T cell activation. Am J Transplant 2014; 14:1277-89. [PMID: 24842641 PMCID: PMC6083870 DOI: 10.1111/ajt.12719] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/07/2014] [Accepted: 02/20/2014] [Indexed: 01/25/2023]
Abstract
The presence of CD28(-) memory CD8 T cells in the peripheral blood of renal transplant patients is a risk factor for graft rejection and resistance to CTLA-4Ig induction therapy. In vitro analyses have indicated poor alloantigen-induced CD28(-) memory CD8 T cell proliferation, raising questions about mechanisms mediating their clonal expansion in kidney grafts to mediate injury. Candidate proliferative cytokines were tested for synergy with alloantigen in stimulating CD28(-) memory CD8 T cell proliferation. Addition of IL-15, but not IL-2 or IL-7, to co-cultures of CD28(-) or CD28(+) memory CD8 T cells and allogeneic B cells rescued proliferation of the CD28(-) and enhanced CD28(+) memory T cell proliferation. Proliferating CD28(-) memory CD8 T cells produced high amounts of interferon gamma and tumor necrosis factor alpha and expressed higher levels of the cytolytic marker CD107a than CD28(+) memory CD8 T cells. CTLA-4Ig inhibited alloantigen-induced proliferation of CD28(+) memory CD8 T cell proliferation but had no effect on alloantigen plus IL-15-induced proliferation of either CD28(-) or CD28(+) memory CD8 T cells. These results indicate the ability of IL-15, a cytokine produced by renal epithelial during inflammation, to provoke CD28(-) memory CD8 T cell proliferation and to confer memory CD8 T cell resistance to CTLA-4Ig-mediated costimulation blockade.
Collapse
Affiliation(s)
- Opas Traitanon
- Department of Nephrology and Hypertension, Cleveland Clinic, Cleveland, OH 44195,Division of Nephrology, Department of Internal Medicine, Thammasart University, Pathumthani, Thailand
| | - Anton Gorbachev
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Jennifer J. Bechtel
- Department of Nephrology and Hypertension, Cleveland Clinic, Cleveland, OH 44195
| | - Karen S. Keslar
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - William M. Baldwin
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195,Glickman Urological Institute, Cleveland Clinic, Cleveland, OH 44195,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Emilio D. Poggio
- Department of Nephrology and Hypertension, Cleveland Clinic, Cleveland, OH 44195,Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195,Glickman Urological Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Robert L. Fairchild
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195,Glickman Urological Institute, Cleveland Clinic, Cleveland, OH 44195,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| |
Collapse
|
122
|
Abstract
Tissues such as the genital tract, skin, and lung act as barriers against invading pathogens. To protect the host, incoming microbes must be quickly and efficiently controlled by the immune system at the portal of entry. Memory is a hallmark of the adaptive immune system, which confers long-term protection and is the basis for efficacious vaccines. While the majority of existing vaccines rely on circulating antibody for protection, struggles to develop antibody-based vaccines against infections such as herpes simplex virus (HSV) and human immunodeficiency virus (HIV) have underscored the need to generate memory T cells for robust antiviral control. The circulating memory T-cell population is generally divided into two subsets: effector memory (TEM ) and central memory (TCM ). These two subsets can be distinguished by their localization, as TCM home to secondary lymphoid organs and TEM circulate through non-lymphoid tissues. More recently, studies have identified a third subset, called tissue-resident memory (TRM ) cells, based on its migratory properties. This subset is found in peripheral tissues that require expression of specific chemoattractants and homing receptors for T-cell recruitment and retention, including barrier sites such as the skin and genital tract. In this review, we categorize different tissues in the body based on patterns of memory T-cell migration and tissue residency. This review also describes the rules for TRM generation and the properties that distinguish them from circulating TEM and TCM cells. Finally, based on the failure of recent T-cell-based vaccines to provide optimal protection, we also discuss the potential role of TRM cells in vaccine design against microbes that invade through the peripheral tissues and highlight new vaccination strategies that take advantage of this newly described memory T-cell subset.
Collapse
Affiliation(s)
- Haina Shin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | |
Collapse
|
123
|
Yin J, Liu C, Wang X, Wang L, Shi Y, Tang W, Ding G, Liu R, Chen S, Gu H, Zheng L. Interleukin 15 receptor alpha rs2228059 A > C polymorphism decreased risk of gastric cardiac adenocarcinoma in a Chinese population. Tumour Biol 2014; 35:6593-600. [PMID: 24696261 DOI: 10.1007/s13277-014-1872-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 03/19/2014] [Indexed: 11/28/2022] Open
Abstract
Gastric cardiac adenocarcinoma (GCA) is one of the common malignant tumors in the world and has a high incidence in China. Both environmental risk factors and genetic factors might play an essential role in the GCA carcinogenesis. We performed a hospital-based case-control study to evaluate the genetic effects of interleukin 15 (IL15) and IL15 receptor alpha (IL15RA) functional single nucleotide polymorphisms (SNPs) on the pathogenesis of GCA. A total of 243 GCA cases and 476 controls were enrolled in this study. The genotypes were determined using a custom-by-design 48-Plex SNPscan(TM) Kit. When the IL15RA rs2228059 AA homozygote genotype was used as the reference group, the CC genotype was correlated with a significantly decreased risk for GCA (CC vs. AA: adjusted OR = 0.61, 95 % CI = 0.37-0.98, p = 0.042). Our results revealed that functional variant IL15RA rs2228059 A > C might attenuate individual's risk of GCA. However, there was no significant association between the other five IL15 SNPs and GCA susceptibility. This present study demonstrated that IL15RA rs2228059 A > C polymorphism might modify GCA susceptibility. The results were based on a limited sample size; future larger studies with more rigorous designs are warranted to validate our findings.
Collapse
Affiliation(s)
- Jun Yin
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Larrea E, Riezu-Boj JI, Aldabe R, Guembe L, Echeverria I, Balasiddaiah A, Gastaminza P, Civeira MP, Sarobe P, Prieto J. Dysregulation of interferon regulatory factors impairs the expression of immunostimulatory molecules in hepatitis C virus genotype 1-infected hepatocytes. Gut 2014; 63:665-73. [PMID: 23787026 DOI: 10.1136/gutjnl-2012-304377] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
BACKGROUND IL-7 and IL-15 are produced by hepatocytes and are critical for the expansion and function of CD8 T cells. IL-15 needs to be presented by IL-15Rα for efficient stimulation of CD8 T cells. METHODS We analysed the hepatic levels of IL-7, IL-15, IL-15Rα and interferon regulatory factors (IRF) in patients with chronic hepatitis C (CHC) (78% genotype 1) and the role of IRF1 and IRF2 on IL-7 and IL-15Rα expression in Huh7 cells with or without hepatitis C virus (HCV) replicon. RESULTS Hepatic expression of both IL-7 and IL-15Rα, but not of IL-15, was reduced in CHC. These patients exhibited decreased hepatic IRF2 messenger RNA levels and diminished IRF2 staining in hepatocyte nuclei. We found that IRF2 controls basal expression of both IL-7 and IL-15Rα in Huh7 cells. IRF2, but not IRF1, is downregulated in cells with HCV genotype 1b replicon and this was accompanied by decreased expression of IL-7 and IL-15Rα, a defect reversed by overexpressing IRF2. Treating Huh7 cells with IFNα plus oncostatin M increased IL-7 and IL-15Rα mRNA more intensely than either cytokine alone. This effect was mediated by strong upregulation of IRF1 triggered by the combined treatment. Induction of IRF1, IL-7 and IL-15Rα by IFNα plus oncostatin M was dampened in replicon cells but the combination was more effective than either cytokine alone. CONCLUSIONS HCV genotype 1 infection downregulates IRF2 in hepatocytes attenuating hepatocellular expression of IL-7 and IL-15Rα. Our data reveal a new mechanism by which HCV abrogates specific T-cell responses and point to a novel therapeutic approach to stimulate anti-HCV immunity.
Collapse
Affiliation(s)
- Esther Larrea
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research (CIMA), , Pamplona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
İnal A. Immunology of liver transplantation. EXP CLIN TRANSPLANT 2014; 12 Suppl 1:5-10. [PMID: 24635783 DOI: 10.6002/ect.25liver.l9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In comparison with other solid-organ transplants, liver allografts are immunologically privileged. Allografts are rejected by immune reactions of the host, and clinical therapy for liver allografts includes immunosuppression to prevent rejection. Orthotopic liver transplant causes systemic donor-specific T-cell tolerance. In addition, antigens introduced into hepatocytes or the portal vein cause tolerance. The basic mechanism in liver tolerance may include continuous exposure of diverse liver cell types to endotoxin derived from intestinal bacteria. This exposure promotes the expression of cytokines, antigen-presenting molecules, and costimulatory signals that inactivate T cells, partly by effects on liver antigen-presenting cells. A simple, reliable, noninvasive assay to evaluate antidonor alloreactivity may be important in implementing these approaches in the laboratory and clinic.
Collapse
Affiliation(s)
- Ali İnal
- Department of Immunology, Baskent University Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
126
|
Increasing the biological activity of IL-2 and IL-15 through complexing with anti-IL-2 mAbs and IL-15Rα-Fc chimera. Immunol Lett 2014; 159:1-10. [PMID: 24512738 DOI: 10.1016/j.imlet.2014.01.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/23/2014] [Accepted: 01/31/2014] [Indexed: 11/22/2022]
Abstract
IL-2 and IL-15 are structurally relative cytokines that share two receptor subunits, CD132 (γ(c) chain) and CD122 (β chain). However, the expression pattern and physiological role of IL-2 and IL-15 private receptor α chains CD25 and IL-15Rα, respectively, are strikingly different. CD25, together with CD122 and CD132, forms a trimeric high affinity IL-2 receptor that is expressed and functions on cells acquiring an IL-2 signal. Conversely, IL-15Rα is expressed and binds IL-15 with high affinity per se already in the endoplasmic reticulum of the IL-15 producing cells and it presents IL-15 to cells expressing CD122/CD132 dimeric receptor in trans. Thus, while IL-2 is secreted almost exclusively by activated T cells and acts as a free molecule, IL-15 is expressed mostly by myeloid cells and works as a cell surface-associated cytokine. Interestingly, the in vivo biological activity of IL-2 can be dramatically increased through complexing with certain anti-IL-2 mAbs; such IL-2/anti-IL-2 mAbs immunocomplexes selectively stimulate the proliferation of a distinct population of immune cells, depending on the clone of the anti-IL-2 mAb used. IL-2/S4B6 mAb immunocomplexes are highly stimulatory for CD122(high) populations (memory CD8(+) T and NK cells) and intermediately also for CD25(high) populations (Treg and activated T cells), while IL-2/JES6-1 mAb immunocomplexes enormously expand only CD25(high) cells. Although IL-2 immunocomplexes are much more potent than IL-2 in vivo, they show comparable to slightly lower activity in vitro. The in vivo biological activity of IL-15 can be dramatically increased through complexing with recombinant IL-15Rα-Fc chimera; however, IL-15/IL-15Rα-Fc complexes are significantly more potent than IL-15 both in vivo and in vitro. In this review we summarize and discuss the features and biological relevance of IL-2/anti-IL-2 mAbs and IL-15/IL-15Rα-Fc complexes, and try to foreshadow their potential in immunological research and immunotherapy.
Collapse
|
127
|
Sckisel GD, Tietze JK, Zamora AE, Hsiao HH, Priest SO, Wilkins DEC, Lanier LL, Blazar BR, Baumgarth N, Murphy WJ. Influenza infection results in local expansion of memory CD8(+) T cells with antigen non-specific phenotype and function. Clin Exp Immunol 2014; 175:79-91. [PMID: 23937663 DOI: 10.1111/cei.12186] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2013] [Indexed: 12/30/2022] Open
Abstract
Primary viral infections induce activation of CD8(+) T cells responsible for effective resistance. We sought to characterize the nature of the CD8(+) T cell expansion observed after primary viral infection with influenza. Infection of naive mice with different strains of influenza resulted in the rapid expansion of memory CD8(+) T cells exhibiting a unique bystander phenotype with significant up-regulation of natural killer group 2D (NKG2D), but not CD25, on the CD44(high) CD8(+) T cells, suggesting an antigen non-specific phenotype. We further confirmed the non-specificity of this phenotype on ovalbumin-specific (OT-I) CD8(+) T cells, which are not specific to influenza. These non-specific CD8(+) T cells also displayed increased lytic capabilities and were observed primarily in the lung. Thus, influenza infection was shown to induce a rapid, antigen non-specific memory T cell expansion which is restricted to the specific site of inflammation. In contrast, CD8(+) T cells of a similar phenotype could be observed in other organs following administration of systemic agonistic anti-CD40 and interleukin-2 immunotherapy, demonstrating that bystander expansion in multiple sites is possible depending on whether the nature of activation is either acute or systemic. Finally, intranasal blockade of NKG2D resulted in a significant increase in viral replication early during the course of infection, suggesting that NKG2D is a critical mediator of anti-influenza responses prior to the initiation of adaptive immunity. These results characterize further the local bystander expansion of tissue-resident, memory CD8(+) T cells which, due to their early induction, may play an important NKG2D-mediated, antigen non-specific role during the early stages of viral infection.
Collapse
Affiliation(s)
- Gail D Sckisel
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA; Graduate Group in Immunology, University of California, Davis, Davis, CA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Abstract
We are entering an exciting time in the study of immunologic tolerance. Several cellular and molecular strategies have been developed that show promise in nonhuman transplant models and these approaches are just now appearing in clinical trials. Tolerance strategies that prevent immune rejection and obviate the need for immunosuppressive medications (with inherent risk of cancer, infection, and organ toxicity) would improve both graft and patient survival. Each tolerance protocol brings its own set of associated risks. As the results of these trials become available, we must continue to evaluate their successes and failures. The balance of these outcomes will help us answer the question: "Tolerance-Is it worth it?"
Collapse
Affiliation(s)
- Erik B Finger
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota 55455
| | | | | |
Collapse
|
129
|
Yin J, Wang L, Shi Y, Shao A, Tang W, Wang X, Zhang W, Ding G, Liu C, Chen Y, Chen S, Gu H. IL-15 receptor alpha rs2228059 A>C polymorphism was associated with a decreased risk of esophageal cancer in a Chinese population. Mol Biol Rep 2014; 41:1951-7. [PMID: 24464181 DOI: 10.1007/s11033-014-3042-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 01/04/2014] [Indexed: 02/02/2023]
|
130
|
Dai Z, Zhang S, Xie Q, Wu S, Su J, Li S, Xu Y, Li XC. Natural CD8+CD122+ T cells are more potent in suppression of allograft rejection than CD4+CD25+ regulatory T cells. Am J Transplant 2014; 14:39-48. [PMID: 24219162 DOI: 10.1111/ajt.12515] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 08/12/2013] [Accepted: 08/13/2013] [Indexed: 01/25/2023]
Abstract
Despite extensive studies on CD4+CD25+ regulatory T cells (Tregs), their application in adoptive transfer therapies is still not optimal in immune-competent wild-type (WT) animal models. Therefore, it is compelling to search for more potent Tregs for potential clinical application. Mounting evidence has shown that naturally occurring CD8+CD122+ T cells are also Tregs. However, their suppression in allograft rejection, efficiency in suppression and underlying mechanisms remain unclear. Using a murine allotransplantation model, we reported here that CD8+CD122+ Tregs were actually more potent in suppression of allograft rejection and underwent more rapid homeostatic proliferation than their CD4+CD25+ counterparts. Moreover, they produced more IL-10 and were more potent in suppressing T cell proliferation in vitro. Deficiency in IL-10 in CD4+CD25+ and CD8+CD122+ Tregs resulted in their reduced but equal suppression in vivo and in vitro, suggesting that IL-10 is responsible for more effective suppression by CD8+CD122+ than CD4+CD25+ Tregs. Importantly, transfer of CD8+CD122+ Tregs together with the administration of recombinant IL-15 significantly prolonged allograft survival in WT mice. Thus, for the first time, we demonstrate that naturally arising CD8+CD122+ Tregs not only inhibit allograft rejection but also exert this suppression more potently than their CD4+CD25+ counterparts. This novel finding may have important implications for tolerance induction.
Collapse
Affiliation(s)
- Z Dai
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine and the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
131
|
Litvinova LS, Sokhonevich NA, Gutsol AA, Kofanova KA. The influence of immunoregulatory cytokines IL-2, IL-7, and IL-15 upon activation, proliferation, and apoptosis of immune memory T-cells in vitro. ACTA ACUST UNITED AC 2013. [DOI: 10.1134/s1990519x13060072] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
132
|
Dendritic cell-specific delivery of Flt3L by coronavirus vectors secures induction of therapeutic antitumor immunity. PLoS One 2013; 8:e81442. [PMID: 24312302 PMCID: PMC3842931 DOI: 10.1371/journal.pone.0081442] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/22/2013] [Indexed: 12/22/2022] Open
Abstract
Efficacy of antitumor vaccination depends to a large extent on antigen targeting to dendritic cells (DCs). Here, we assessed antitumor immunity induced by attenuated coronavirus vectors which exclusively target DCs in vivo and express either lymphocyte- or DC-activating cytokines in combination with a GFP-tagged model antigen. Tracking of in vivo transduced DCs revealed that vectors encoding for Fms-like tyrosine kinase 3 ligand (Flt3L) exhibited a higher capacity to induce DC maturation compared to vectors delivering IL-2 or IL-15. Moreover, Flt3L vectors more efficiently induced tumor-specific CD8+ T cells, expanded the epitope repertoire, and provided both prophylactic and therapeutic tumor immunity. In contrast, IL-2- or IL-15-encoding vectors showed a substantially lower efficacy in CD8+ T cell priming and failed to protect the host once tumors had been established. Thus, specific in vivo targeting of DCs with coronavirus vectors in conjunction with appropriate conditioning of the microenvironment through Flt3L represents an efficient strategy for the generation of therapeutic antitumor immunity.
Collapse
|
133
|
Abstract
Improved outcomes for patients with cancer hinge on the development of new targeted therapies with acceptable short-term and long-term toxicity. Progress in basic, preclinical, and clinical arenas spanning cellular immunology, synthetic biology, and cell-processing technologies has paved the way for clinical applications of chimeric antigen receptor-based therapies. This new form of targeted immunotherapy merges the exquisite targeting specificity of monoclonal antibodies with the potent cytotoxicity and long-term persistence provided by cytotoxic T cells. Although this field is still in its infancy, clinical trials have already shown clinically significant antitumor activity in neuroblastoma, chronic lymphocytic leukemia, and B cell lymphoma, and trials targeting a variety of other adult and pediatric malignancies are under way. Ongoing work is focused on identifying optimal tumor targets and on elucidating and manipulating both cell- and host-associated factors to support expansion and persistence of the genetically engineered cells in vivo. The potential to target essentially any tumor-associated cell-surface antigen for which a monoclonal antibody can be made opens up an entirely new arena for targeted therapy of cancer.
Collapse
Affiliation(s)
- David M Barrett
- Abramson Cancer Center and the Departments of Medicine, Pediatrics, and Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104;
| | | | | | | | | |
Collapse
|
134
|
Kalos M, June CH. Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. Immunity 2013; 39:49-60. [PMID: 23890063 DOI: 10.1016/j.immuni.2013.07.002] [Citation(s) in RCA: 364] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Indexed: 01/12/2023]
Abstract
Adoptive T cell transfer for cancer and chronic infection is an emerging field that shows promise in recent trials. Synthetic-biology-based engineering of T lymphocytes to express high-affinity antigen receptors can overcome immune tolerance, which has been a major limitation of immunotherapy-based strategies. Advances in cell engineering and culture approaches to enable efficient gene transfer and ex vivo cell expansion have facilitated broader evaluation of this technology, moving adoptive transfer from a "boutique" application to the cusp of a mainstream technology. The major challenge currently facing the field is to increase the specificity of engineered T cells for tumors, because targeting shared antigens has the potential to lead to on-target off-tumor toxicities, as observed in recent trials. As the field of adoptive transfer technology matures, the major engineering challenge is the development of automated cell culture systems, so that the approach can extend beyond specialized academic centers and become widely available.
Collapse
Affiliation(s)
- Michael Kalos
- Abramson Cancer Center and the Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5156, USA.
| | | |
Collapse
|
135
|
Alcantar-Orozco EM, Gornall H, Baldan V, Hawkins RE, Gilham DE. Potential limitations of the NSG humanized mouse as a model system to optimize engineered human T cell therapy for cancer. Hum Gene Ther Methods 2013; 24:310-20. [PMID: 23931270 DOI: 10.1089/hgtb.2013.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The genetic modification of peripheral blood lymphocytes using retroviral vectors to redirect T cells against tumor cells has been recently used as a means to generate large numbers of antigen-specific T cells for adoptive cell therapy protocols. However, commonly used retroviral vector-based genetic modification requires T cells to be driven into cell division; this potent mitogenic stimulus is associated with the development of an effector phenotype that may adversely impact upon the long-term engraftment potential and subsequent antitumor effects of T cells. To investigate whether the cytokines used during culture impact upon the engraftment potential of gene-modified T cells, a humanized model employing T cells engrafted with a MART-1-specific T cell receptor adoptively transferred into NOD/Shi-scid IL-2rγ(-/-) (NSG) immune-deficient mice bearing established melanoma tumors was used to compare the effects of the common γ chain cytokines IL-2, IL-7, and IL-15 upon gene-modified T cell activity. MART-1-specific T cells cultured in IL-7 and IL-15 demonstrated greater relative in vitro proliferation and viability of T cells compared with the extensively used IL-2. Moreover, the IL-15 culture prolonged the survival of animals bearing melanoma tumors after adoptive transfer. However, the combination of IL-7 and IL-15 produced T cells with improved engraftment potential compared with IL-15 alone; however, a high rate of xenogeneic graft-versus-host disease prevented the identification of a clear improvement in antitumor effect of these T cells. These results clearly demonstrate modulation of gene-modified T cell engraftment in the NSG mouse, which supports the future testing of the combination of IL-7 and IL-15 in adoptive cell therapy protocols; however, this improved engraftment is also associated with the long-term maintenance of xenoreactive T cells, which limits the ultimate usefulness of the NSG mouse model in this situation.
Collapse
Affiliation(s)
- Erik M Alcantar-Orozco
- Clinical and Experimental Immunotherapy Group, Department of Medical Oncology, The Institute of Cancer Sciences, Manchester Academic Healthcare Science Centre, The University of Manchester , Manchester M20 4BX, United Kingdom
| | | | | | | | | |
Collapse
|
136
|
Proliferation-linked apoptosis of adoptively transferred T cells after IL-15 administration in macaques. PLoS One 2013; 8:e56268. [PMID: 23418547 PMCID: PMC3572023 DOI: 10.1371/journal.pone.0056268] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 01/07/2013] [Indexed: 11/19/2022] Open
Abstract
The adoptive transfer of antigen-specific effector T cells is being used to treat human infections and malignancy. T cell persistence is a prerequisite for therapeutic efficacy, but reliably establishing a high-level and durable T cell response by transferring cultured CD8+ T cells remains challenging. Thus, strategies that promote a transferred high-level T cell response may improve the efficacy of T cell therapy. Lymphodepletion enhances persistence of transferred T cells in mice in part by reducing competition for IL-15, a common γ-chain cytokine that promotes T cell memory, but lymphodepleting regimens have toxicity. IL-15 can be safely administered and has minimal effects on CD4+ regulatory T cells at low doses, making it an attractive adjunct in adoptive T cell therapy. Here, we show in lymphoreplete macaca nemestrina, that proliferation of adoptively transferred central memory-derived CD8+ effector T (TCM/E) cells is enhanced in vivo by administering IL-15. TCM/E cells migrated to memory niches, persisted, and acquired both central memory and effector memory phenotypes regardless of the cytokine treatment. Unexpectedly, despite maintaining T cell proliferation, IL-15 did not augment the magnitude of the transferred T cell response in blood, bone marrow, or lymph nodes. T cells induced to proliferate by IL-15 displayed increased apoptosis demonstrating that enhanced cycling was balanced by cell death. These results suggest that homeostatic mechanisms that regulate T cell numbers may interfere with strategies to augment a high-level T cell response by adoptive transfer of CD8+ TCM/E cells in lymphoreplete hosts.
Collapse
|
137
|
Santegoets SJAM, Turksma AW, Suhoski MM, Stam AGM, Albelda SM, Hooijberg E, Scheper RJ, van den Eertwegh AJM, Gerritsen WR, Powell DJ, June CH, de Gruijl TD. IL-21 promotes the expansion of CD27+ CD28+ tumor infiltrating lymphocytes with high cytotoxic potential and low collateral expansion of regulatory T cells. J Transl Med 2013; 11:37. [PMID: 23402380 PMCID: PMC3626797 DOI: 10.1186/1479-5876-11-37] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 01/14/2013] [Indexed: 11/10/2022] Open
Abstract
Background Adoptive cell transfer of tumor infiltrating lymphocytes has shown clinical efficacy in the treatment of melanoma and is now also being explored in other tumor types. Generation of sufficient numbers of effector T cells requires extensive ex vivo expansion, often at the cost of T cell differentiation and potency. For the past 20 years, IL-2 has been the key cytokine applied in the expansion of TIL for ACT. However, the use of IL-2 has also led to collateral expansion of regulatory T cells (Tregs) and progressive T cell differentiation, factors known to limit in vivo persistence and activity of transferred TIL. The use of alternative T cell growth factors is therefore warranted. Here, we have compared the effects of IL-2, -15 and −21 cytokines on the expansion and activation of TIL from single-cell suspensions of non-small cell lung cancer, ovarian cancer and melanoma. Methods We applied the K562-based artificial APC (aAPC) platform for the direct and rapid expansion of tumor infiltrating lymphocytes isolated from primary cancer specimens. These aAPC were engineered to express the Fc-γ receptor CD32 (for anti-CD3 antibody binding), the co-stimulatory molecule 4-1BBL, and to secrete either IL-2, IL-15 or IL-21 cytokine. Results Although IL-2 aAPC induced the greatest overall TIL expansion, IL-21 aAPC induced superior expansion of CD8+ T cells with a CD27+CD28+ “young” phenotype and superior functional cytotoxic effector characteristics, without collateral expansion of Tregs. Conclusion Our data rationalize the clinical application of IL-21-secreting aAPC as a standardized cell-based platform in the expansion of “young” effector TIL for ACT.
Collapse
|
138
|
Yang X, Kallarakal A, Saptharishi N, Jiang H, Yang Z, Xie Y, Mitra G, Zheng XX, Strom TB, Soman G. Molecular characterization and functional activity of an IL-15 antagonist MutIL-15/Fc human fusion protein. Mol Pharm 2013; 10:717-27. [PMID: 23311475 PMCID: PMC3573692 DOI: 10.1021/mp300513j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Fc fusion proteins are a new emerging class of molecules for immune-targeted delivery of therapeutic proteins. Biophysical and bioanalytical characterization is critical for clinical development and delivery of therapeutic proteins. Here we report molecular and functional characterization of a recombinant human fusion protein Mutant IL-15/Fc. MutIL-15/Fc has a molecular weight of ∼95 kDa as determined by multiangle laser light scattering with online size exclusion chromatography and migrated at a faster rate (lower retention time) in gel filtration column. The kinetics of binding of MutIL-15/Fc to Fcγ receptor is best fitted in a bivalent modal with K(D1) 5 μM and K(D2) 9 μM determined by surface plasmon resonance (BIAcore). N-Glycoprofiling analysis revealed extensive glycosylation of MutIL-15/Fc. The Fc and IL-15 components in the MutIL-15/Fc are detected using the dual mode ELISA. The HT-2 cell proliferation inhibition assay is qualified as a quantitative in vitro marker functional assay. Molecular state changes associated with forced stress analyzed by SEC-MALS resulted in changes in bioactivity and Fc:Fcγ receptor interaction affinity. These data provide a systematic approach to molecular and functional characterization of the MutIL-15/Fc to establish product consistency and stability monitoring during storage and under drug delivery conditions.
Collapse
Affiliation(s)
- Xiaoyi Yang
- Biopharmaceutical Development Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Abraham Kallarakal
- Biopharmaceutical Development Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Nirmala Saptharishi
- Biopharmaceutical Development Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Hengguang Jiang
- Biopharmaceutical Development Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Zhiwen Yang
- Biopharmaceutical Development Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Yueqing Xie
- Biopharmaceutical Development Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - George Mitra
- Biopharmaceutical Development Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Xin Xiao Zheng
- Thomas Starzl Transplant Institute, University of Pittsburgh Medical Center, Pittsburgh, PA 15261
| | - Terry B. Strom
- Harvard Medical School, Department of Surgery and Medicine, Transplant Institute at Beth Israel Deaconess Medical Center, Massachusetts General Hospital, Boston, MA 02215
| | - Gopalan Soman
- Biopharmaceutical Development Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| |
Collapse
|
139
|
Mathematical models of memory CD8+ T-cell repertoire dynamics in response to viral infections. Bull Math Biol 2013; 75:491-522. [PMID: 23377628 PMCID: PMC7088647 DOI: 10.1007/s11538-013-9817-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 01/17/2013] [Indexed: 01/29/2023]
Abstract
Immunity to diseases is conferred by pathogen-specific memory cells that prevent disease reoccurrences. A broad repertoire of memory T-cells must be developed and maintained to effectively protect against viral invasions; yet, the total number of memory T-cells is constrained between infections. Thus, creating memory to new infections can require attrition of some existing memory cells. Furthermore, some viruses induce memory T-cell death early in an infection, after which surviving cells proliferate to refill the memory compartment.We develop mathematical models of cellular attrition and proliferation in order to examine how new viral infections impact existing immunity. With these probabilistic models, we qualitatively and quantitatively predict how the composition and diversity of the memory repertoire changes as a result of viral infections. In addition, we calculate how often immunity to prior diseases is lost due to new infections. Comparing our results across multiple general infection types allows us to draw conclusions about, which types of viral effects most drastically alter existing immunity. We find that early memory attrition does not permanently alter the repertoire composition, while infections that spark substantial new memory generation drastically shift the repertoire and hasten the decline of existing immunity.
Collapse
|
140
|
Heikamp EB, Powell JD. Sensing the immune microenvironment to coordinate T cell metabolism, differentiation & function. Semin Immunol 2013; 24:414-20. [PMID: 23332779 DOI: 10.1016/j.smim.2012.12.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 12/11/2012] [Indexed: 12/14/2022]
Abstract
Mounting an adaptive immune response is bioenergetically demanding. As a result, T cell activation coincides with profound changes in cellular metabolism that must be coordinated with instructive signals from cytokine and costimulatory receptors to generate an immune response. Studies examining the intimate link between metabolism and immune function have revealed that different types of T cells have distinct metabolic profiles. Data is emerging that place mTOR, an evolutionarily conserved serine-threonine kinase, as a central integrator of these processes. In this review, we will discuss the role of mTOR in determining both CD4 and CD8 T cell metabolism, differentiation, and trafficking.
Collapse
Affiliation(s)
- Emily B Heikamp
- Sidney Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | | |
Collapse
|
141
|
Keshavarz Valian H, Nateghi Rostami M, Tasbihi M, Miramin Mohammadi A, Eskandari SE, Sarrafnejad A, Khamesipour A. CCR7+ central and CCR7- effector memory CD4+ T cells in human cutaneous leishmaniasis. J Clin Immunol 2013; 33:220-34. [PMID: 22990666 DOI: 10.1007/s10875-012-9788-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 08/28/2012] [Indexed: 01/02/2023]
Abstract
PURPOSE The profile of central (=T(CM)) and effector (=T(EM)) memory CD4(+) T cell subsets and the possible role as surrogate markers of protection is studied in the volunteers with history of cutaneous leishmaniasis (HCL). METHODS Profile of T cell subsets based on CCR7/CD45RA expressions and phenotypic changes after soluble Leishmania antigen (SLA) stimulation were analyzed. Then, sorted CD4(+)CD45RO(-)CD45RA(+) naïve T, CD4(+)CD45RO(+)CD45RA(-)CCR7(-) T(EM,) CD4(+)CD45RO(+)CD45RA(-)CCR7(+) T(CM) subsets were cultured with SLA for proliferation, cytokine production and intracellular cytokine assays. RESULTS In the HCL and control volunteers, the mean frequencies of CD4(+)CD45RA(+)CCR7(+) naïve T cells and CD4(+)CD45RA(-)CCR7(-) T(EM) cells were higher than the other subsets before culture. Frequency of naïve T cells and CD4(+)CD45RA(-)CCR7(+) T(CM) cells was significantly decreased (P=0.01 for naïve T and P<0.05 for T(CM) cells) and frequency of T(EM) cells was significantly increased after SLA stimulation compared to before culture (P<0.001). By CFSE labeling, CD4(+)CD45RO(+)CD45RA(-)CCR7(+) T(CM) cells showed more proliferation potential than CD4(+)CD45RO(+)CD45RA(-)CCR7(-) T(EM) cells. Stimulation of the T(EM) cells in HCL volunteers induced a significantly higher IFN-γ production (P=0.04) with higher number of intracellular IFN-γ positive cells (P=0.032) than the same cells from controls. A significantly higher number of T(CM) cells produced IL-2 in HCL volunteers compared with controls (P<0.05). Most of the intracellular IFN-γ positive T(EM) cells were proliferating CFSE-dim populations (P<0.05). CONCLUSIONS A combination of Leishmania-reactive IFN-γ producing CD4(+)CD45RO(+)CD45RA(-)CCR7(-) T(EM) and Leishmania-reactive IL-2 producing CD4(+)CD45RO(+)CD45RA(-)CCR7(+) T(CM) are identified in individuals with history of CL which might play a role in protective recall immune response against Leishmania infection.
Collapse
Affiliation(s)
- Hossein Keshavarz Valian
- Medical Parasitology and Mycology Department, School of Public Health, Tehran University of Medical Sciences, P.O. Box 14155-6446, Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
142
|
Rathakrishnan A, Wang SM, Hu Y, Khan AM, Ponnampalavanar S, Lum LCS, Manikam R, Sekaran SD. Cytokine expression profile of dengue patients at different phases of illness. PLoS One 2012; 7:e52215. [PMID: 23284941 PMCID: PMC3527385 DOI: 10.1371/journal.pone.0052215] [Citation(s) in RCA: 136] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 11/12/2012] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Dengue is an important medical problem, with symptoms ranging from mild dengue fever to severe forms of the disease, where vascular leakage leads to hypovolemic shock. Cytokines have been implicated to play a role in the progression of severe dengue disease; however, their profile in dengue patients and the synergy that leads to continued plasma leakage is not clearly understood. Herein, we investigated the cytokine kinetics and profiles of dengue patients at different phases of illness to further understand the role of cytokines in dengue disease. METHODS AND FINDINGS Circulating levels of 29 different types of cytokines were assessed by bead-based ELISA method in dengue patients at the 3 different phases of illness. The association between significant changes in the levels of cytokines and clinical parameters were analyzed. At the febrile phase, IP-10 was significant in dengue patients with and without warning signs. However, MIP-1β was found to be significant in only patients with warning signs at this phase. IP-10 was also significant in both with and without warning signs patients during defervescence. At this phase, MIP-1β and G-CSF were significant in patients without warning signs, whereas MCP-1 was noted to be elevated significantly in patients with warning signs. Significant correlations between the levels of VEGF, RANTES, IL-7, IL-12, PDGF and IL-5 with platelets; VEGF with lymphocytes and neutrophils; G-CSF and IP-10 with atypical lymphocytes and various other cytokines with the liver enzymes were observed in this study. CONCLUSIONS The cytokine profile patterns discovered between the different phases of illness indicate an essential role in dengue pathogenesis and with further studies may serve as predictive markers for progression to dengue with warning signs.
Collapse
Affiliation(s)
- Anusyah Rathakrishnan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Seok Mui Wang
- Institute of Medical Molecular Biotechnology, Faculty of Medicine, Universiti Teknologi MARA, Selangor, Malaysia
| | - Yongli Hu
- Perdana University Graduate School of Medicine, Serdang, Selangor, Malaysia
| | - Asif M. Khan
- Perdana University Graduate School of Medicine, Serdang, Selangor, Malaysia
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | | | - Lucy Chai See Lum
- Department of Paediatrics, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Rishya Manikam
- Department of Trauma and Emergency Medicine, University Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Shamala Devi Sekaran
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
143
|
Connor LM, Kohlmeier JE, Ryan L, Roberts AD, Cookenham T, Blackman MA, Woodland DL. Early dysregulation of the memory CD8+ T cell repertoire leads to compromised immune responses to secondary viral infection in the aged. IMMUNITY & AGEING 2012; 9:28. [PMID: 23244347 PMCID: PMC3564731 DOI: 10.1186/1742-4933-9-28] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 12/08/2012] [Indexed: 11/10/2022]
Abstract
UNLABELLED BACKGROUND Virus-specific memory CD8+ T cells persist long after infection is resolved and are important for mediating recall responses to secondary infection. Although the number of memory T cells remains relatively constant over time, little is known about the overall stability of the memory T cell pool, particularly with respect to T cell clonal diversity. In this study we developed a novel assay to measure the composition of the memory T cell pool in large cohorts of mice over time following respiratory virus infection. RESULTS We find that the clonal composition of the virus-specific memory CD8+ T cell pool begins to change within months of the initial infection. These early clonal perturbations eventually result in large clonal expansions that have been associated with ageing. CONCLUSIONS Maintenance of clonal diversity is important for effective long-term memory responses and dysregulation of the memory response begins early after infection.
Collapse
|
144
|
Krzych U, Dalai S, Zarling S, Pichugin A. Memory CD8 T cells specific for plasmodia liver-stage antigens maintain protracted protection against malaria. Front Immunol 2012; 3:370. [PMID: 23233854 PMCID: PMC3517952 DOI: 10.3389/fimmu.2012.00370] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 11/20/2012] [Indexed: 01/15/2023] Open
Abstract
Immunologic memory induced by pathogenic agents or vaccinations is inextricably linked to long-lasting protection. Adequately maintained memory T and B cell pools assure a fast, effective, and specific response against re-infections. Studies of immune responses amongst residents of malaria endemic areas suggest that memory responses to Plasmodia antigens appear to be neither adequately developed nor maintained, because persons who survive episodes of childhood malaria remain vulnerable to persistent or intermittent malaria infections. By contrast, multiple exposures of humans and laboratory rodents to radiation-attenuated Plasmodia sporozoites (γ-spz) induces sterile and long-lasting protection against experimental sporozoite challenge. Protection is associated with MHC-class I-dependent CD8 T cells, the key effectors against pre-erythrocytic stage infection. We have adopted the P. berghei γ-spz mouse model to study memory CD8 T cells that are specific for antigens expressed by Pb liver-stage (LS) parasites and are found predominantly in the liver. On the basis of phenotypic and functional characteristics, we have demonstrated that liver CD8 T cells form two subsets: CD44hiCD62LloKLRG-1+CD107+CD127−CD122loCD8 T effector/effector memory (TE/EM) cells that are the dominant IFN-γ producers and CD44hiCD62LhiKLRG-1−CD107−CD127+CD122hiCD8 T central memory (TCM) cells. In this review, we discuss our observations concerning the role of CD8 TE/EM and CD8 TCM cells in the maintenance of protracted protective immunity against experimental malaria infection. Finally, we present a hypothesis consistent with a model whereby intrahepatic CD8 TCM cells, that are maintained in part by LS-Ag depot and by IL-15-mediated survival and homeostatic proliferation, form a reservoir of cells ready for conscription to CD8 TE/EM cells needed to prevent re-infections.
Collapse
Affiliation(s)
- Urszula Krzych
- Department of Cellular Immunology, Branch of Military Malaria Vaccine Development, Walter Reed Army Institute of Research Silver Spring, MD, USA
| | | | | | | |
Collapse
|
145
|
Zhang M, Ju W, Yao Z, Yu P, Wei BR, Simpson RM, Waitz R, Fassò M, Allison JP, Waldmann TA. Augmented IL-15Rα expression by CD40 activation is critical in synergistic CD8 T cell-mediated antitumor activity of anti-CD40 antibody with IL-15 in TRAMP-C2 tumors in mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 188:6156-64. [PMID: 22593619 PMCID: PMC3370156 DOI: 10.4049/jimmunol.1102604] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
IL-15 has potential as an immunotherapeutic agent for cancer treatment because it is a critical factor for the proliferation and activation of NK and CD8(+) T cells. However, monotherapy of patients with malignancy with IL-15 that has been initiated may not be optimal, because of the limited expression of the private receptor, IL-15Rα. We demonstrated greater CD8 T cell-mediated therapeutic efficacy using a combination regimen of murine IL-15 administered with an agonistic anti-CD40 Ab (FGK4.5) that led to increased IL-15Rα expression on dendritic cells (DCs), as well as other cell types, in a syngeneic established TRAMP-C2 tumor model. Seventy to one hundred percent of TRAMP-C2 tumor-bearing wild-type C57BL/6 mice in the combination group manifested sustained remissions, whereas only 0-30% in the anti-CD40-alone group and none in the murine IL-15-alone group became tumor free (p < 0.001). However, the combination regimen showed less efficacy in TRAMP-C2 tumor-bearing IL-15Rα(-/-) mice than in wild-type mice. The combination regimen significantly increased the numbers of TRAMP-C2 tumor-specific SPAS-1/SNC9-H(8) tetramer(+)CD8(+) T cells, which were associated with the protection from tumor development on rechallenge with TRAMP-C2 tumor cells. Using an in vitro cytolytic assay that involved NK cells primed by wild-type or IL-15Rα(-/-) bone marrow-derived DCs, we demonstrated that the expression of IL-15Rα by DCs appeared to be required for optimal IL-15-induced NK priming and killing. These findings support the view that anti-CD40-mediated augmented IL-15Rα expression was critical in IL-15-associated sustained remissions observed in TRAMP-C2 tumor-bearing mice receiving combination therapy.
Collapse
Affiliation(s)
- Meili Zhang
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol 2012; 12:180-90. [PMID: 22343569 DOI: 10.1038/nri3156] [Citation(s) in RCA: 1229] [Impact Index Per Article: 94.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Interleukin-2 (IL-2) signals influence various lymphocyte subsets during differentiation, immune responses and homeostasis. As discussed in this Review, stimulation with IL-2 is crucial for the maintenance of regulatory T (T(Reg)) cells and for the differentiation of CD4(+) T cells into defined effector T cell subsets following antigen-mediated activation. For CD8(+) T cells, IL-2 signals optimize both effector T cell generation and differentiation into memory cells. IL-2 is presented in soluble form or bound to dendritic cells and the extracellular matrix. Use of IL-2 - either alone or in complex with particular neutralizing IL-2-specific antibodies - can amplify CD8(+) T cell responses or induce the expansion of the T(Reg) cell population, thus favouring either immune stimulation or suppression.
Collapse
|
147
|
Akue AD, Lee JY, Jameson SC. Derivation and maintenance of virtual memory CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:2516-23. [PMID: 22308307 DOI: 10.4049/jimmunol.1102213] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Memory CD8(+) T cells are an important component of the adaptive immune response against many infections, and understanding how Ag-specific memory CD8(+) T cells are generated and maintained is crucial for the development of vaccines. We recently reported the existence of memory-phenotype, Ag-specific CD8(+) T cells in unimmunized mice (virtual memory or VM cells). However, it was not clear when and where these cells are generated during normal development, nor the factors required for their production and maintenance. This issue is especially pertinent given recent data showing that memory-like CD8 T cells can be generated in the thymus, in a bystander response to IL-4. In this study, we show that the size of the VM population is reduced in IL-4R-deficient animals. However, the VM population appears first in the periphery and not the thymus of normal animals, suggesting this role of IL-4 is manifest following thymic egress. We also show that the VM pool is durable, showing basal proliferation and long-term maintenance in normal animals, and also being retained during responses to unrelated infection.
Collapse
Affiliation(s)
- Adovi D Akue
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55414, USA
| | | | | |
Collapse
|
148
|
Siegel AM, Heimall J, Freeman AF, Hsu AP, Brittain E, Brenchley JM, Douek DC, Fahle GH, Cohen JI, Holland SM, Milner JD. A critical role for STAT3 transcription factor signaling in the development and maintenance of human T cell memory. Immunity 2012; 35:806-18. [PMID: 22118528 DOI: 10.1016/j.immuni.2011.09.016] [Citation(s) in RCA: 230] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 07/26/2011] [Accepted: 09/16/2011] [Indexed: 10/15/2022]
Abstract
STAT3 transcription factor signaling in specific T helper cell differentiation has been well described, although the broader roles for STAT3 in lymphocyte memory are less clear. Patients with autosomal-dominant hyper-IgE syndrome (AD-HIES) carry dominant-negative STAT3 mutations and are susceptible to a variety of bacterial and fungal infections. We found that AD-HIES patients have a cell-intrinsic defect in the number of central memory CD4(+) and CD8(+) T cells compared to healthy controls. Naive T cells from AD-HIES patients had lower expression of memory-related transcription factors BCL6 and SOCS3, a primary proliferation defect, and they failed to acquire central memory-like surface phenotypes in vitro. AD-HIES patients showed a decreased ability to control varicella zoster virus (VZV) and Epstein-Barr virus (EBV) latency, and T cell memory to both of these viruses was compromised. These data point to a specific role for STAT3 in human central memory T cell formation and in control of certain chronic viruses.
Collapse
Affiliation(s)
- Andrea M Siegel
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Delineation of antigen-specific and antigen-nonspecific CD8(+) memory T-cell responses after cytokine-based cancer immunotherapy. Blood 2012; 119:3073-83. [PMID: 22251483 DOI: 10.1182/blood-2011-07-369736] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Memory T cells exhibit tremendous antigen specificity within the immune system and accumulate with age. Our studies reveal an antigen-independent expansion of memory, but not naive, CD8(+) T cells after several immunotherapeutic regimens for cancer resulting in a distinctive phenotype. Signaling through T-cell receptors (TCRs) or CD3 in both mouse and human memory CD8(+) T cells markedly up-regulated programmed death-1 (PD-1) and CD25 (IL-2 receptor α chain), and led to antigen-specific tumor cell killing. In contrast, exposure to cytokine alone in vitro or with immunotherapy in vivo did not up-regulate these markers but resulted in expanded memory CD8(+) T cells expressing NKG2D, granzyme B, and possessing broadly lytic capabilities. Blockade of NKG2D in mice also resulted in significantly diminished antitumor effects after immunotherapy. Treatment of TCR-transgenic mice bearing nonantigen expressing tumors with immunotherapy still resulted in significant antitumor effects. Human melanoma tissue biopsies obtained from patients after topically applied immunodulatory treatment resulted in increased numbers of these CD8(+) CD25(-) cells within the tumor site. These findings demonstrate that memory CD8(+) T cells can express differential phenotypes indicative of adaptive or innate effectors based on the nature of the stimuli in a process conserved across species.
Collapse
|
150
|
van der Windt GJW, Everts B, Chang CH, Curtis JD, Freitas TC, Amiel E, Pearce EJ, Pearce EL. Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity 2011; 36:68-78. [PMID: 22206904 DOI: 10.1016/j.immuni.2011.12.007] [Citation(s) in RCA: 1184] [Impact Index Per Article: 84.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 10/25/2011] [Accepted: 12/08/2011] [Indexed: 11/29/2022]
Abstract
CD8(+) T cells undergo major metabolic changes upon activation, but how metabolism influences the establishment of long-lived memory T cells after infection remains a key question. We have shown here that CD8(+) memory T cells, but not CD8(+) T effector (Teff) cells, possessed substantial mitochondrial spare respiratory capacity (SRC). SRC is the extra capacity available in cells to produce energy in response to increased stress or work and as such is associated with cellular survival. We found that interleukin-15 (IL-15), a cytokine critical for CD8(+) memory T cells, regulated SRC and oxidative metabolism by promoting mitochondrial biogenesis and expression of carnitine palmitoyl transferase (CPT1a), a metabolic enzyme that controls the rate-limiting step to mitochondrial fatty acid oxidation (FAO). These results show how cytokines control the bioenergetic stability of memory T cells after infection by regulating mitochondrial metabolism.
Collapse
|