51
|
Homeostatic cytokines orchestrate the segregation of CD4 and CD8 memory T-cell reservoirs in mice. Blood 2011; 118:3039-50. [PMID: 21791416 DOI: 10.1182/blood-2011-04-349746] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Memory T cells (T(M)s) have been detected in many tissues but their quantitative distribution remains largely undefined. We show that in mice there is a remarkably biased accumulation of long-term CD4 T(M)s into mucosal sites (mainly gut, especially Peyer patches), and CD8 T(M)s into lymph nodes and spleen (in particular, peripheral lymph nodes [PLNs]). This distinction correlates with their differentiated expression of PLN- and gut-homing markers. CD8 and CD4 T(M)s selectively require the expression of PLN-homing marker CCR7 or gut-homing marker α4β7 for maintenance. PLNs and gut supply CD8 and CD4 T(M)s with their individually favored homeostatic cytokine, IL-15, or IL-7. Cytokine stimulation in turn regulates the different gut-homing marker expression on CD4 and CD8 T(M)s. IL-15 plays a major role in vivo regulating CD8 T(M)s homing to PLNs. Thus, the reservoir segregation of CD4 and CD8 T(M)s meets their individual needs for homeostatic cytokines and is under feedback control of cytokine stimulation.
Collapse
|
52
|
Innate signals overcome acquired TCR signaling pathway regulation and govern the fate of human CD161(hi) CD8α⁺ semi-invariant T cells. Blood 2011; 118:2752-62. [PMID: 21791427 DOI: 10.1182/blood-2011-02-334698] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Type 17 programmed CD161(hi)CD8α(+) T cells contribute to mucosal immunity to bacteria and yeast. In early life, microbial colonization induces proliferation of CD161(hi) cells that is dependent on their expression of a semi-invariant Vα7.2(+) TCR. Although prevalent in adults, CD161(hi)CD8α(+) cells exhibit weak proliferative and cytokine responses to TCR ligation. The mechanisms responsible for the dichotomous response of neonatal and adult CD161(hi) cells, and the signals that enable their effector function, have not been established. We describe acquired regulation of TCR signaling in adult memory CD161(hi)CD8α(+) T cells that is absent in cord CD161(hi) cells and adult CD161(lo) cells. Regulated TCR signaling in CD161(hi) cells was due to profound alterations in TCR signaling pathway gene expression and could be overcome by costimulation through CD28 or innate cytokine receptors, which dictated the fate of their progeny. Costimulation with IL-1β during TCR ligation markedly increased proinflammatory IL-17 production, while IL-12-induced Tc1-like function and restored the response to TCR ligation without costimulation. CD161(hi) cells from umbilical cord blood and granulocyte colony stimulating factor-mobilized leukaphereses differed in frequency and function, suggesting future evaluation of the contribution of CD161(hi) cells in hematopoietic stem cell grafts to transplant outcomes is warranted.
Collapse
|
53
|
Abstract
After infection, most antigen-specific memory T cells reside in nonlymphoid tissues. Tissue-specific programming during priming leads to directed migration of T cells to the appropriate tissue, which promotes the development of tissue-resident memory in organs such as intestinal mucosa and skin. Mechanisms that regulate the retention of tissue-resident memory T cells include transforming growth factor-β (TGF-β)-mediated induction of the E-cadherin receptor CD103 and downregulation of the chemokine receptor CCR7. These pathways enhance protection in internal organs, such as the nervous system, and in the barrier tissues--the mucosa and skin. Memory T cells that reside at these surfaces provide a first line of defense against subsequent infection, and defining the factors that regulate their development is critical to understanding organ-based immunity.
Collapse
Affiliation(s)
- Brian S Sheridan
- Center for Integrated Immunology and Vaccine Research, Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA.
| | | |
Collapse
|
54
|
Abstract
Understanding the mechanisms underlying the induction of immunity in the gastrointestinal mucosa following oral immunization and the cross-talk between mucosal and systemic immunity should expedite the development of vaccines to diminish the global burden caused by enteric pathogens. Identifying an immunological correlate of protection in the course of field trials of efficacy, animal models (when available), or human challenge studies is also invaluable. In industrialized country populations, live attenuated vaccines (e.g. polio, typhoid, and rotavirus) mimic natural infection and generate robust protective immune responses. In contrast, a major challenge is to understand and overcome the barriers responsible for the diminished immunogenicity and efficacy of the same enteric vaccines in underprivileged populations in developing countries. Success in developing vaccines against some enteric pathogens has heretofore been elusive (e.g. Shigella). Different types of oral vaccines can selectively or inclusively elicit mucosal secretory immunoglobulin A and serum immunoglobulin G antibodies and a variety of cell-mediated immune responses. Areas of research that require acceleration include interaction between the gut innate immune system and the stimulation of adaptive immunity, development of safe yet effective mucosal adjuvants, better understanding of homing to the mucosa of immunologically relevant cells, and elicitation of mucosal immunologic memory. This review dissects the immune responses elicited in humans by enteric vaccines.
Collapse
Affiliation(s)
- Marcela F Pasetti
- Center for Vaccine Development, University of Maryland School of Medicine, 685 West Baltimore St., Room 480, Baltimore, MD 21201, USA.
| | | | | | | |
Collapse
|
55
|
Nadazdin O, Boskovic S, Murakami T, Tocco G, Smith RN, Colvin RB, Sachs DH, Allan J, Madsen JC, Kawai T, Cosimi AB, Benichou G. Host alloreactive memory T cells influence tolerance to kidney allografts in nonhuman primates. Sci Transl Med 2011; 3:86ra51. [PMID: 21653831 PMCID: PMC3261229 DOI: 10.1126/scitranslmed.3002093] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Transplant tolerance, defined as indefinite allograft survival without immunosuppression, has been regularly achieved in laboratory mice but not in nonhuman primates or humans. In contrast to laboratory mice, primates regularly have high frequencies of alloreactive memory T cells (TMEMs) before transplantation. These TMEMs are poorly sensitive to conventional immunosuppression and costimulation blockade, and the presence of donor-reactive TMEMs in primates may account for their resistance to transplant tolerance protocols that have proven consistently effective in mice. We measured the frequencies of anti-donor TMEMs before and after transplantation in a series of rejecting and tolerant monkeys that underwent nonmyeloablative conditioning, short-term immunosuppression, and combined allogeneic kidney/cell transplantation. Transplants were acutely rejected in all the monkeys with high numbers of donor-specific TMEMs before transplantation. In contrast, long-term survival was observed in the recipients harboring lower frequencies of anti-donor TMEMs before transplantation. Similar amounts of TMEM homeostatic expansion were recorded in all transplanted monkeys upon hematopoietic reconstitution; however, only the tolerant monkeys had no expansion or activation of donor-reactive TMEMs after transplantation. These results indicate that the presence of high frequencies of host donor-reactive TMEMs before transplantation impairs tolerance induction to kidney allografts in this nonhuman primate model. Indeed, recipients harboring a low anamnestic reactivity to their donor before transplantation were successfully rendered tolerant via infusion of donor cells and short-term immunosuppression. This suggests that selection of allogeneic donors with low memory responses in recipients may be essential to successful transplant tolerance induction in patients.
Collapse
Affiliation(s)
- Ognjenka Nadazdin
- Transplant Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Svjetlan Boskovic
- Transplant Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Toru Murakami
- Transplant Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Georges Tocco
- Transplant Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Rex-Neal Smith
- Pathology Department, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Robert B. Colvin
- Pathology Department, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - David H. Sachs
- Transplantation Biology Research Center, Massachusetts General Hospital, Boston, MA 02129, USA
| | - James Allan
- Transplant Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Joren C. Madsen
- Transplant Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Tatsuo Kawai
- Transplant Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - A. Benedict Cosimi
- Transplant Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Gilles Benichou
- Transplant Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
56
|
du Pré MF, van Berkel LA, Ráki M, van Leeuwen MA, de Ruiter LF, Broere F, ter Borg MN, Lund FE, Escher JC, Lundin KEA, Sollid LM, Kraal G, Nieuwenhuis EES, Samsom JN. CD62L(neg)CD38⁺ expression on circulating CD4⁺ T cells identifies mucosally differentiated cells in protein fed mice and in human celiac disease patients and controls. Am J Gastroenterol 2011; 106:1147-59. [PMID: 21386831 PMCID: PMC3696487 DOI: 10.1038/ajg.2011.24] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The aim of this study was to identify new markers of mucosal T cells to monitor ongoing intestinal immune responses in peripheral blood. METHODS Expression of cell-surface markers was studied in mice on ovalbumin (OVA)-specific T cells in the gut-draining mesenteric lymph nodes (MLN) after OVA feed. The effect of the local mucosal mediators retinoic acid (RA) and transforming growth factor-β (TGF-β) on the induction of a mucosal phenotype was determined in in vitro T-cell differentiation assays with murine and human T cells. Tetramer stainings were performed to study gluten-specific T cells in the circulation of patients with celiac disease, a chronic small-intestinal inflammation. RESULTS In mice, proliferating T cells in MLN were CD62L(neg)CD38(+) during both tolerance induction and abrogation of intestinal homeostasis. This mucosal CD62L(neg)CD38(+) T-cell phenotype was efficiently induced by RA and TGF-β in mice, whereas for human CD4(+) T cells RA alone was sufficient. The CD4(+)CD62L(neg)CD38(+) T-cell phenotype could be used to identify T cells with mucosal origin in human peripheral blood, as expression of the gut-homing chemokine receptor CCR9 and β(7) integrin were highly enriched in this subset whereas expression of cutaneous leukocyte-associated antigen was almost absent. Tetramer staining revealed that gluten-specific T cells appearing in blood of treated celiac disease patients after oral gluten challenge were predominantly CD4(+)CD62L(neg)CD38(+). The total percentage of circulating CD62L(neg)CD38(+) of CD4 T cells was not an indicator of intestinal inflammation as percentages did not differ between pediatric celiac disease patients, inflammatory bowel disease patients and respective controls. However, the phenotypic selection of mucosal T cells allowed cytokine profiling as upon restimulation of CD62L(neg)CD38(+) cells interleukin-10 (IL-10) and interferon-γ (IFN-γ) transcripts were readily detected in circulating mucosal T cells. CONCLUSIONS By selecting for CD62L(neg)CD38(+) expression that comprises 5-10% of the cells within the total CD4(+) T-cell pool we are able to highly enrich for effector T cells with specificity for mucosal antigens. This is of pivotal importance for functional studies as this purification enhances the sensitivity of cytokine detection and cellular activation.
Collapse
Affiliation(s)
- M. Fleur du Pré
- Dept. of Pediatrics, division Gastroenterology and nutrition, Erasmus MC – Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Lisette A. van Berkel
- Dept. of Pediatrics, division Gastroenterology and nutrition, Erasmus MC – Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Melinda Ráki
- Centre for Immune Regulation, Institute of Immunology, University of Oslo and Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Marieke A. van Leeuwen
- Dept. of Pediatrics, division Gastroenterology and nutrition, Erasmus MC – Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Lilian F. de Ruiter
- Dept. of Pediatrics, division Gastroenterology and nutrition, Erasmus MC – Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Femke Broere
- Div. of Immunology, Institute of Infectious Diseases and Immunology, Utrecht University, Utrecht, the Netherlands
| | - Mariëtte N.D. ter Borg
- Dept. of Pediatrics, division Gastroenterology and nutrition, Erasmus MC – Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Frances E. Lund
- Dept. of Medicine, University of Rochester, Rochester NY, USA
| | - Johanna C. Escher
- Dept. of Pediatrics, division Gastroenterology and nutrition, Erasmus MC – Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Knut E. A. Lundin
- Centre for Immune Regulation, Institute of Immunology, University of Oslo and Oslo University Hospital - Rikshospitalet, Oslo, Norway,Dept of Medicine, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Ludvig M. Sollid
- Centre for Immune Regulation, Institute of Immunology, University of Oslo and Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Georg Kraal
- Dept. of Molecular Cell Biology and Immunology, VUMC, Amsterdam, the Netherlands
| | - Edward E. S. Nieuwenhuis
- Dept. of Pediatrics, division Gastroenterology and nutrition, Erasmus MC – Sophia Children’s Hospital, Rotterdam, the Netherlands,Dept. of Pediatric Gastroenterology, University Medical Center Utrecht – Wilhelmina Children’s Hospital, Utrecht, the Netherlands
| | - Janneke N. Samsom
- Dept. of Pediatrics, division Gastroenterology and nutrition, Erasmus MC – Sophia Children’s Hospital, Rotterdam, the Netherlands
| |
Collapse
|
57
|
Altered T cell memory and effector cell development in chronic lymphatic filarial infection that is independent of persistent parasite antigen. PLoS One 2011; 6:e19197. [PMID: 21559422 PMCID: PMC3084782 DOI: 10.1371/journal.pone.0019197] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 03/22/2011] [Indexed: 02/08/2023] Open
Abstract
Chronic lymphatic filarial (LF) infection is associated with suppression of parasite-specific T cell responses that persist even following elimination of infection. While several mechanisms have been implicated in mediating this T cell specific downregulation, a role for alterations in the homeostasis of T effector and memory cell populations has not been explored. Using multiparameter flow cytometry, we investigated the role of persistent filarial infection on the maintenance of T cell memory in patients from the filarial-endemic Cook Islands. Compared to filarial-uninfected endemic normals (EN), microfilaria (mf) positive infected patients (Inf) had a reduced CD4 central memory (TCM) compartment. In addition, Inf patients tended to have more effector memory cells (TEM) and fewer effector cells (TEFF) than did ENs giving significantly smaller TEFF ∶ TEM ratios. These contracted TCM and TEFF populations were still evident in patients previously mf+ who had cleared their infection (CLInf). Moreover, the density of IL-7Rα, necessary for T memory cell maintenance (but decreased in T effector cells), was significantly higher on memory cells of Inf and CLInf patients, although there was no evidence for decreased IL-7 or increased soluble IL7-Rα, both possible mechanisms for signaling defects in memory cells. However, effector cells that were present in Inf and CLInf patients had lower percentages of HLA-DR suggesting impaired function. These changes in T cell populations appear to reflect chronicity of infection, as filarial-infected children, despite the presence of active infection, did not show alterations in the frequencies of these T cell phenotypes. These data indicate that filarial-infected patients have contracted TCM compartments and a defect in effector cell development, defects that persist even following clearance of infection. The fact that these global changes in memory and effector cell compartments do not yet occur in infected children makes early treatment of LF even more crucial.
Collapse
|
58
|
Wirth TC, Martin MD, Starbeck-Miller G, Harty JT, Badovinac VP. Secondary CD8+ T-cell responses are controlled by systemic inflammation. Eur J Immunol 2011; 41:1321-33. [PMID: 21425157 DOI: 10.1002/eji.201040730] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 12/21/2010] [Accepted: 01/26/2011] [Indexed: 11/06/2022]
Abstract
Repeated infections and experimental prime-boost regimens frequently result in the generation of secondary (2°) CD8(+) T-cell responses. In contrast to primary (1°) CD8(+) T cells, the parameters that influence the abundance and phenotype of 2° effector and memory CD8(+) T-cell populations are largely unknown. Here, we analyze the impact of different booster infections, Ag curtailment, and systemic inflammation on the quality and quantity of secondary CD8(+) T-cell responses. We show that similar to 1° CD8(+) T-cell responses, the phenotype of 2° effector and memory CD8(+) T-cell populations is critically dependent on the nature of the infectious pathogen and the inflammatory milieu early after infection. In addition, systemic inflammation increases the number of 2° effector and memory CD8(+) T cells after booster infections and immunizations. Therefore, our data reveal new means to boost the number of 2° effector and memory CD8(+) T cells in prime-boost regimens and show a surprisingly high degree of plasticity in 2° memory CD8(+) T-cell phenotype that is controlled by systemic inflammation.
Collapse
Affiliation(s)
- Thomas C Wirth
- Department of Microbiology, University of Iowa, Iowa City, IA, USA
| | | | | | | | | |
Collapse
|
59
|
Vasoactive intestinal peptide receptor 1 is downregulated during expansion of antigen-specific CD8 T cells following primary and secondary Listeria monocytogenes infections. J Neuroimmunol 2011; 234:40-8. [PMID: 21396722 DOI: 10.1016/j.jneuroim.2011.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 12/30/2010] [Accepted: 02/03/2011] [Indexed: 12/26/2022]
Abstract
As regulation of CD8 T cell homeostasis is incompletely understood, we investigated the expression profile of the vasoactive intestinal peptide (VIP) receptors, VPAC1 and VPAC2, on CD8 T cells throughout an in vivo immune response. Herein, we show that adoptively transferred CD8 T cells responding to a Listeria monocytogenes infection significantly downregulated, functionally active VPAC1 protein expression during primary and secondary expansion. VPAC1 mRNA expression was restored during contraction and regained naïve levels in primary, but remained low during secondary, memory generation. VIP co-administration with primary infection suppressed CD8 T cell expansion (≈ 50%). VPAC2 was not detected at any time points throughout primary and secondary infections. Collectively, our data demonstrate that functionally active VPAC1 is dynamically downregulated to render expanding CD8 T cells unresponsive to VIP.
Collapse
|
60
|
Cush SS, Flaño E. KLRG1+NKG2A+ CD8 T cells mediate protection and participate in memory responses during γ-herpesvirus infection. THE JOURNAL OF IMMUNOLOGY 2011; 186:4051-8. [PMID: 21346231 DOI: 10.4049/jimmunol.1003122] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Functional CD8 T cell effector and memory responses are generated and maintained during murine γ-herpesvirus 68 (γHV68) persistent infection despite continuous presentation of viral lytic Ags. However, the identity of the CD8 T cell subpopulations that mediate effective recall responses and that can participate in the generation of protective memory to a γ-herpesvirus infection remains unknown. During γHV68 persistence, ∼75% of γHV68-specific CD8 T cells coexpress the NK receptors killer cell lectin-like receptor G1 (KLRG1) and NKG2A. In this study, we take advantage of this unique phenotype to analyze the capacity of CD8 T cells expressing or not expressing KLRG1 and NKG2A to mediate effector and memory responses. Our results show that γHV68-specific KLRG1(+)NKG2A(+) CD8 T cells have an effector memory phenotype as well as characteristics of polyfunctional effector cells such us IFN-γ and TNF-α production, killing capacity, and are more efficient at protecting against a γHV68 challenge than their NKG2A(-)KLRG1(-) counterparts. Nevertheless, γHV68-specific NKG2A(+)KLRG1(+) CD8 T cells express IL-7 and IL-15 receptors, can survive long-term without cognate Ag, and subsequently mount a protective response during antigenic recall. These results highlight the plasticity of the immune system to generate protective effector and proliferative memory responses during virus persistence from a pool of KLRG1(+)NKG2A(+) effector memory CD8 T cells.
Collapse
Affiliation(s)
- Stephanie S Cush
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | | |
Collapse
|
61
|
Whitmire JK. Induction and function of virus-specific CD4+ T cell responses. Virology 2011; 411:216-28. [PMID: 21236461 DOI: 10.1016/j.virol.2010.12.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 12/08/2010] [Indexed: 12/18/2022]
Abstract
CD4+ T cells - often referred to as T-helper cells - play a central role in immune defense and pathogenesis. Virus infections and vaccines stimulate and expand populations of antigen-specific CD4+ T cells in mice and in man. These virus-specific CD4+ T cells are extremely important in antiviral protection: deficiencies in CD4+ T cells are associated with virus reactivation, generalized susceptibility to opportunistic infections, and poor vaccine efficacy. As described below, CD4+ T cells influence effector and memory CD8+ T cell responses, humoral immunity, and the antimicrobial activity of macrophages and are involved in recruiting cells to sites of infection. This review summarizes a few key points about the dynamics of the CD4+ T cell response to virus infection, the positive role of pro-inflammatory cytokines in the differentiation of virus-specific CD4+ T cells, and new areas of investigation to improve vaccines against virus infection.
Collapse
Affiliation(s)
- Jason K Whitmire
- Carolina Vaccine Institute, The University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
62
|
Amanna IJ, Slifka MK. Contributions of humoral and cellular immunity to vaccine-induced protection in humans. Virology 2011; 411:206-15. [PMID: 21216425 DOI: 10.1016/j.virol.2010.12.016] [Citation(s) in RCA: 179] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 12/08/2010] [Indexed: 12/17/2022]
Abstract
Vaccines play a vital role in protecting the host against infectious disease. The most effective licensed vaccines elicit long-term antigen-specific antibody responses by plasma cells in addition to the development of persisting T cell and B cell memory. The relative contributions of these different immune cell subsets are context-dependent and vary depending on the attributes of the vaccine (i.e., live/attenuated, inactivated, and subunit) as well as the biology of the pathogen in question. For relatively simple vaccines against bacterial antigens (e.g., tetanus toxin) or invariant viruses, the immunological correlates of protection are well-characterized. For more complex vaccines against viruses, especially those that mutate or cause latent infections, it is more difficult to define the specific correlates of immunity. This often requires observational/natural history studies, clinical trials, or experimental evaluation in relevant animal models in order for immunological correlates to be determined or extrapolated. In this review, we will discuss the relative contributions of virus-specific T cell and B cell responses to vaccine-mediated protection against disease.
Collapse
Affiliation(s)
- Ian J Amanna
- Najít Technologies, Inc., Beaverton, OR 97006, USA.
| | | |
Collapse
|
63
|
Wiesel M, Kratky W, Oxenius A. Type I IFN Substitutes for T Cell Help during Viral Infections. THE JOURNAL OF IMMUNOLOGY 2010; 186:754-63. [DOI: 10.4049/jimmunol.1003166] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
64
|
O'Connor AM, Crawley AM, Angel JB. Interleukin-7 enhances memory CD8(+) T-cell recall responses in health but its activity is impaired in human immunodeficiency virus infection. Immunology 2010; 131:525-36. [PMID: 20673240 DOI: 10.1111/j.1365-2567.2010.03325.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Memory CD8(+) T cells regain function during a recall response, but the requirement of signals in addition to antigen during a secondary immune response is unknown. In this study, the ability of interleukin-7 (IL-7) to enhance memory CD8(+ ) CD45RA(- ) CD127(+) T-cell responses in health and in human immunodeficiency virus (HIV) infection was investigated. CD8(+) T-cell-depleted peripheral blood mononuclear cells (PBMCs) from HIV(-) and untreated HIV(+) donors were pulsed with a cytomegalovirus/Epstein-Barr virus/influenza (CEF) peptide pool, and co-cultured with autologous memory CD8(+) T cells in the presence of IL-7. Cell survival and the function of memory CD8(+) T-cell subsets were then evaluated. Memory CD8(+) T-cell proliferation and interferon-γ (IFN-γ) production was enhanced by the presence of antigen, and the addition of IL-7 further enhanced antigen-induced proliferation. In HIV(+) individuals, the presence of antigen enhanced IFN-γ production to a small degree but did not enhance proliferation. Lastly, IL-7 did not enhance antigen-mediated proliferation of memory CD8(+) T cells from HIV(+) individuals. IL-7 therefore appears to have a role in secondary immune responses and its activity is impaired in memory CD8(+) T cells from HIV(+) individuals. These results further our understanding of the signals involved in secondary immune responses, and provide new insight into the loss of CD8(+) T-cell function in HIV infection.
Collapse
Affiliation(s)
- Alison M O'Connor
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | | | | |
Collapse
|
65
|
Abstract
The world is now experiencing an epidemic of obesity. Although the effects of obesity on the development of metabolic and cardiovascular problems are well studied, much less is known about the impact of obesity on immune function and infectious disease. Studies in obese humans and with obese animal models have repeatedly demonstrated impaired immune function, including decreased cytokine production, decreased response to antigen/mitogen stimulation, reduced macrophage and dendritic cell function, and natural killer cell impairment. Recent studies have demonstrated that the impaired immune response in the obese host leads to increased susceptibility to infection with a number of different pathogens such as community-acquired tuberculosis, influenza, Mycobacterium tuberculosis, coxsackievirus, Helicobacter pylori and encephalomyocarditis virus. While no specific mechanism has been defined for the decreased immune response to infectious disease in the obese host, several obesity-associated changes such as excessive inflammation, altered adipokine signaling, metabolic changes and even epigenetic regulation could affect the immune response. This review will discuss what is currently known about the relationship between obesity and infectious disease.
Collapse
Affiliation(s)
- Erik A Karlsson
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN 38105-3678
| | - Melinda A Beck
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599-7461, USA
| |
Collapse
|
66
|
Mucosal adjuvants and long-term memory development with special focus on CTA1-DD and other ADP-ribosylating toxins. Mucosal Immunol 2010; 3:556-66. [PMID: 20844480 DOI: 10.1038/mi.2010.54] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The ultimate goal for vaccination is to stimulate protective immunological memory. Protection against infectious diseases not only relies on the magnitude of the humoral immune response, but more importantly on the quality and longevity of it. Adjuvants are critical components of most non-living vaccines. Although little attention has been given to qualitative aspects of the choice of vaccine adjuvant, emerging data demonstrate that this function may be central to vaccine efficacy. In this review we describe efforts to understand more about how adjuvants influence qualitative aspects of memory development. We describe recent advances in understanding how vaccines induce long-lived plasma and memory B cells, and focus our presentation on the germinal center reaction. As mucosal vaccination requires powerful adjuvants, we have devoted much attention to the adenosine diphosphate (ADP)-ribosylating cholera toxin and the CTA1-DD adjuvants as examples of how mucosal adjuvants can influence induction of long-term memory.
Collapse
|
67
|
Wiesel M, Joller N, Ehlert AK, Crouse J, Spörri R, Bachmann MF, Oxenius A. Th cells act via two synergistic pathways to promote antiviral CD8+ T cell responses. THE JOURNAL OF IMMUNOLOGY 2010; 185:5188-97. [PMID: 20881183 DOI: 10.4049/jimmunol.1001990] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The mechanisms of how Th cells promote CD8(+) T cell responses during viral infections are largely unknown. In this study, we unraveled the mechanisms of T cell help for CD8(+) T cell responses during vaccinia virus infection. Our results demonstrate that Th cells promote vaccinia virus-specific CD8(+) T cell responses via two interconnected synergistic pathways: First, CD40L expressed by activated CD4(+) T cells instructs dendritic cells to produce bioactive IL-12p70, which is directly sensed by Ag-specific CD8(+) T cells, resulting in increased IL-2Rα expression. Second, Th cells provide CD8(+) T cells with IL-2, thereby enhancing their survival. Thus, Th cells are at the center of an important communication loop with a central role for IL-2/IL-2R and bioactive IL-12.
Collapse
Affiliation(s)
- Melanie Wiesel
- Institute for Microbiology, Swiss Federal Institute of Technology Zürich (ETH Zürich), Zürich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
68
|
Daniels MA, Teixeiro E. The persistence of T cell memory. Cell Mol Life Sci 2010; 67:2863-78. [PMID: 20364394 PMCID: PMC11115859 DOI: 10.1007/s00018-010-0362-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2010] [Accepted: 03/19/2010] [Indexed: 12/14/2022]
Abstract
T cell memory is a crucial feature of the adaptive immune system in the defense against pathogens. During the last years, numerous studies have focused their efforts on uncovering the signals, inflammatory cues, and extracellular factors that support memory differentiation. This research is beginning to decipher the complex gene network that controls memory programming. However, how the different signals, that a T cell receives during the process of differentiation, interplay to trigger memory programming is still poorly defined. In this review, we focus on the most recent advances in the field and discuss how T cell receptor signaling and inflammation control CD8 memory differentiation.
Collapse
Affiliation(s)
- Mark A Daniels
- Department of Molecular Microbiology and Immunology, School of Medicine, Center for Cellular and Molecular Immunology, University of Missouri, M616 Medical Sciences Bldg., One Hospital Dr., Columbia, MO 65212, USA.
| | | |
Collapse
|
69
|
Regulation of memory CD8 T-cell differentiation by cyclin-dependent kinase inhibitor p27Kip1. Mol Cell Biol 2010; 30:5145-59. [PMID: 20805358 DOI: 10.1128/mcb.01045-09] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Induction of potent T-cell memory is the goal of vaccinations, but the molecular mechanisms that regulate the formation of memory CD8 T cells are not well understood. Despite the recognition that controls of cellular proliferation and apoptosis govern the number of memory T cells, the cell cycle regulatory mechanisms that control these key cellular processes in CD8 T cells during an immune response are poorly defined. Here, we have identified the cyclin-dependent kinase inhibitor p27(Kip1) as a critical regulator of the CD8 T-cell homeostasis at all phases of the T-cell response to an acute viral infection in mice. By acting as a timer for cell cycle exit, p27(Kip1) curtailed the programmed expansion of interleukin-2-producing memory precursors and markedly limited the magnitude and quality of CD8 T-cell memory. In the absence of p27(Kip1), CD8 T cells showed superior recall responses shortly after vaccination with recombinant Listeria monocytogenes. Additionally, we show that p27(Kip1) constrains proliferative renewal of memory CD8 T cells, especially of the effector memory subset. These findings provide critical insights into the cell cycle regulation of CD8 T-cell homeostasis and suggest that modulation of p27(Kip1) could bolster vaccine-induced T-cell memory and protective immunity.
Collapse
|
70
|
Wirth TC, Xue HH, Rai D, Sabel JT, Bair T, Harty JT, Badovinac VP. Repetitive antigen stimulation induces stepwise transcriptome diversification but preserves a core signature of memory CD8(+) T cell differentiation. Immunity 2010; 33:128-40. [PMID: 20619696 DOI: 10.1016/j.immuni.2010.06.014] [Citation(s) in RCA: 217] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 04/26/2010] [Accepted: 05/11/2010] [Indexed: 12/21/2022]
Abstract
Repetitive antigen stimulation by prime-boost vaccination or pathogen reencounter increases memory CD8(+) T cell numbers, but the impact on memory CD8(+) T cell differentiation is unknown. Here we showed that repetitive antigen stimulations induced accumulation of memory CD8(+) T cells with uniform effector memory characteristics. However, genome-wide microarray analyses revealed that each additional antigen challenge resulted in the differential regulation of several hundred new genes in the ensuing memory CD8(+) T cell populations and, therefore, in stepwise diversification of CD8(+) T cell transcriptomes. Thus, primary and repeatedly stimulated (secondary, tertiary, and quaternary) memory CD8(+) T cells differed substantially in their molecular signature while sharing expression of a small group of genes and biological pathways, which may constitute a core signature of memory differentiation. These results reveal the complex regulation of memory CD8(+) T cell differentiation and identify potential new molecular targets to dissect the function of memory cells generated by repeated antigen stimulation.
Collapse
Affiliation(s)
- Thomas C Wirth
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | | | |
Collapse
|
71
|
Obar JJ, Lefrançois L. Early signals during CD8 T cell priming regulate the generation of central memory cells. THE JOURNAL OF IMMUNOLOGY 2010; 185:263-72. [PMID: 20519649 DOI: 10.4049/jimmunol.1000492] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The CD8(+) T cell response to infection is characterized by the appearance of short-lived (CD127(low) killer cell lectin-like receptor G 1-high) and memory-precursor (CD127(high) killer cell lectin-like receptor G 1-low) effector cells. How and when central-memory T (T(CM); CD62L(high) CCR7(+)) cell and effector-memory T(T(EM); CD62L(low) CCR7(-)) cell subsets are established remains unclear. We now show that the T(CM) cell lineage represents an early developmental branchpoint during the CD8(+) T cell response to infection. Central-memory CD8(+) T cells could be identified prior to the peak of the CD8(+) T cell response and were enriched in lymphoid organs. Moreover, the kinetics and magnitude of T(CM) cell development were dependent on the infectious agent. Furthermore, the extent of early Ag availability, which regulated programmed death-1 and CD25 expression levels, controlled the T(CM)/T(EM) cell lineage decision ultimately through IL-2 and IL-15 signaling levels. These observations identify key early signals that help establish the T(CM)/T(EM) cell dichotomy and provide the means to manipulate memory lineage choices.
Collapse
Affiliation(s)
- Joshua J Obar
- Department of Immunology, Center for Integrated Immunology and Vaccine Research, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | |
Collapse
|
72
|
Nadazdin O, Boskovic S, Murakami T, O'Connor DH, Wiseman RW, Karl JA, Tuscher JJ, Sachs DH, Madsen JC, Tocco G, Kawai T, Cosimi AB, Benichou G. Phenotype, distribution and alloreactive properties of memory T cells from cynomolgus monkeys. Am J Transplant 2010; 10:1375-84. [PMID: 20486921 PMCID: PMC2893326 DOI: 10.1111/j.1600-6143.2010.03119.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The high frequency of memory T cells present in primates is thought to represent a major barrier to tolerance induction in transplantation. Therefore, it is crucial to characterize these memory T cells and determine their functional properties. High numbers of memory T cells were detected in peripheral blood and all lymphoid tissues except lymph nodes, which were essentially the site of naïve T cells. The majority of CD8(+) memory T cells were effector memory cells located in the blood and bone marrow while most CD4(+) memory T cells were central memory cells present in the spleen. Next, memory T cells from over 100 monkeys were tested for their response to alloantigens by ELISPOT. Memory alloreactivity mediated via direct but not indirect allorecognition was detected in all animals. The frequency of allospecific memory T cells varied dramatically depending upon the nature of the responder/stimulator monkey combination tested. MHC gene matching was generally associated with a low-memory alloreactivity. Nevertheless, low anamnestic alloresponses were also found in a significant number of fully MHC-mismatched monkey combinations. These results show that selected donor/recipient combinations displaying a low memory alloresponsiveness can be found. These combinations may be more favorable for transplant tolerance induction.
Collapse
Affiliation(s)
- Ognjenka Nadazdin
- Department of Surgery, Transplantation Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Svjetlan Boskovic
- Department of Surgery, Transplantation Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Toru Murakami
- Department of Surgery, Transplantation Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - D. H. O'Connor
- Department of Pathology and Laboratory Medicine, Wisconsin Primate Research Center, University of Wisconsin at Madison, Madison, WI
| | - Roger W. Wiseman
- Department of Pathology and Laboratory Medicine, Wisconsin Primate Research Center, University of Wisconsin at Madison, Madison, WI
| | - J. A. Karl
- Department of Pathology and Laboratory Medicine, Wisconsin Primate Research Center, University of Wisconsin at Madison, Madison, WI
| | - J. J. Tuscher
- Department of Pathology and Laboratory Medicine, Wisconsin Primate Research Center, University of Wisconsin at Madison, Madison, WI
| | - D. H. Sachs
- Department of Surgery, Transplantation Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - J. C. Madsen
- Department of Surgery, Transplantation Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Georges Tocco
- Department of Surgery, Transplantation Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Tatsuo Kawai
- Department of Surgery, Transplantation Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - A. B. Cosimi
- Department of Surgery, Transplantation Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Gilles Benichou
- Department of Surgery, Transplantation Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA,Corresponding author: Gilles Benichou,
| |
Collapse
|
73
|
Decman V, Laidlaw BJ, Dimenna LJ, Abdulla S, Mozdzanowska K, Erikson J, Ertl HCJ, Wherry EJ. Cell-intrinsic defects in the proliferative response of antiviral memory CD8 T cells in aged mice upon secondary infection. THE JOURNAL OF IMMUNOLOGY 2010; 184:5151-9. [PMID: 20368274 DOI: 10.4049/jimmunol.0902063] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although previous studies have demonstrated delayed viral clearance and blunted effector T cell responses in aged mice during infection, memory CD8 T cells and especially secondary responses have received less attention. In this study, we show that modest differences in the number of memory CD8 T cells formed in aged versus young animals were associated with altered memory CD8 T cell differentiation. Aged immune mice had increased morbidity and mortality upon secondary viral challenge, suggesting changes in T cell immunity. Indeed, virus-specific memory CD8 T cells from aged mice showed substantially reduced proliferative expansion upon secondary infection using multiple challenge models. In addition, this defect in recall capacity of aged memory CD8 T cells was cell-intrinsic and persisted upon adoptive transfer into young mice. Thus, the poor proliferative potential of memory T cells and altered memory CD8 T cell differentiation could underlie age-related defects in antiviral immunity.
Collapse
Affiliation(s)
- Vilma Decman
- Immunology Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
74
|
Abstract
In response to infection or effective vaccination, naive antigen-specific CD8+ T cells undergo a dramatic highly orchestrated activation process. Initial encounter with an appropriately activated antigen-presenting cell leads to blastogenesis and an exponential increase in antigen-specific CD8+ T cell numbers. Simultaneously, a dynamic differentiation process occurs, resulting in formation of both primary effector and long-lived memory cells. Current findings have emphasized the heterogeneity of effector and memory cell populations with the description of multiple cellular subsets based on phenotype, function, and anatomic location. Yet, only recently have we begun to dissect the underlying factors mediating the temporal control of the development of distinct effector and memory CD8+ T cell sublineages. In this review we will focus on the requirements for mounting an effective CD8+ T cell response and highlight the elements regulating the differentiation of effector and memory subsets.
Collapse
Affiliation(s)
- Joshua J Obar
- Center for Integrated Immunology and Vaccine Research, Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut 06107, USA
| | | |
Collapse
|
75
|
Rai D, Pham NLL, Harty JT, Badovinac VP. Tracking the total CD8 T cell response to infection reveals substantial discordance in magnitude and kinetics between inbred and outbred hosts. THE JOURNAL OF IMMUNOLOGY 2010; 183:7672-81. [PMID: 19933864 DOI: 10.4049/jimmunol.0902874] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Determining the magnitude and kinetics, together with the phenotypic and functional characteristics of responding CD8 T cells, is critical for understanding the regulation of adaptive immunity as well as in evaluating vaccine candidates. Recent technical advances have allowed tracking of some CD8 T cells responding to infection, and a body of information now exists describing phenotypic changes that occur in CD8 T cells of known Ag-specificity during their activation, expansion, and memory generation in inbred mice. In this study, we demonstrate that Ag but not inflammation-driven changes in expression of CD11a and CD8alpha can be used to distinguish naive from Ag-experienced (effector and memory) CD8 T cells after infection or vaccination. Interestingly and in contrast to inbred mice, tracking polyclonal CD8 T cell responses with this approach after bacterial and viral infections revealed substantial discordance in the magnitude and kinetics of CD8 T cell responses in outbred hosts. These data reveal limitations to the use of inbred mouse strains as preclinical models at vaccine development and suggest the same dose of infection or vaccination can lead to substantial differences in the magnitude and timing of Ag-specific CD8 expansion as well in differences in protective memory CD8 T cell numbers in outbred individuals. This concept has direct relevance to development of vaccines in outbred humans.
Collapse
Affiliation(s)
- Deepa Rai
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
76
|
Kalia V, Sarkar S, Ahmed R. CD8 T-Cell Memory Differentiation during Acute and Chronic Viral Infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 684:79-95. [DOI: 10.1007/978-1-4419-6451-9_7] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
77
|
Waugh C, Sinclair L, Finlay D, Bayascas JR, Cantrell D. Phosphoinositide (3,4,5)-triphosphate binding to phosphoinositide-dependent kinase 1 regulates a protein kinase B/Akt signaling threshold that dictates T-cell migration, not proliferation. Mol Cell Biol 2009; 29:5952-62. [PMID: 19703999 PMCID: PMC2772752 DOI: 10.1128/mcb.00585-09] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 06/17/2009] [Accepted: 08/13/2009] [Indexed: 12/28/2022] Open
Abstract
The present study explored the consequences of phosphoinositide (3,4,5)-triphosphate [PI(3,4,5)P(3)] binding to the pleckstrin homology (PH) domain of the serine/threonine kinase 3-phosphoinositide-dependent kinase 1 (PDK1). The salient finding is that PDK1 directly transduces the PI(3,4,5)P(3) signaling that determines T-cell trafficking programs but not T-cell growth and proliferation. The integrity of the PDK1 PH domain thus is not required for PDK1 catalytic activity or to support cell survival and the proliferation of thymic and peripheral T cells. However, a PDK1 mutant that cannot bind PI(3,4,5)P(3) cannot trigger the signals that terminate the expression of the transcription factor KLF2 in activated T cells and cannot switch the chemokine and adhesion receptor profile of naive T cells to the profile of effector T cells. The PDK1 PH domain also is required for the maximal activation of Akt/protein kinase B (PKB) and for the maximal phosphorylation and inactivation of Foxo family transcription factors in T cells. PI(3,4,5)P(3) binding to PDK1 and the strength of PKB activity thus can dictate the nature of the T-cell response. Low levels of PKB activity can be sufficient for T-cell proliferation but insufficient to initiate the migratory program of effector T cells.
Collapse
Affiliation(s)
- Caryll Waugh
- Department of Cell Biology and Immunology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom, Institut de Neurociencies, Departament de Bioquimica i Biologia Molecular, Universitat Autonoma de Barcelona, Barcelona E-08193, Spain
| | - Linda Sinclair
- Department of Cell Biology and Immunology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom, Institut de Neurociencies, Departament de Bioquimica i Biologia Molecular, Universitat Autonoma de Barcelona, Barcelona E-08193, Spain
| | - David Finlay
- Department of Cell Biology and Immunology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom, Institut de Neurociencies, Departament de Bioquimica i Biologia Molecular, Universitat Autonoma de Barcelona, Barcelona E-08193, Spain
| | - Jose R. Bayascas
- Department of Cell Biology and Immunology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom, Institut de Neurociencies, Departament de Bioquimica i Biologia Molecular, Universitat Autonoma de Barcelona, Barcelona E-08193, Spain
| | - Doreen Cantrell
- Department of Cell Biology and Immunology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom, Institut de Neurociencies, Departament de Bioquimica i Biologia Molecular, Universitat Autonoma de Barcelona, Barcelona E-08193, Spain
| |
Collapse
|
78
|
Macrophage- and Dendritic-Cell-Derived Interleukin-15 Receptor Alpha Supports Homeostasis of Distinct CD8+ T Cell Subsets. Immunity 2009; 31:811-22. [DOI: 10.1016/j.immuni.2009.09.017] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 08/04/2009] [Accepted: 09/10/2009] [Indexed: 10/20/2022]
|
79
|
Steers NJ, Peachman KK, McClain S, Alving CR, Rao M. Liposome-encapsulated HIV-1 Gag p24 containing lipid A induces effector CD4+ T-cells, memory CD8+ T-cells, and pro-inflammatory cytokines. Vaccine 2009; 27:6939-49. [PMID: 19748578 DOI: 10.1016/j.vaccine.2009.08.105] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Revised: 08/26/2009] [Accepted: 08/27/2009] [Indexed: 11/26/2022]
Abstract
Liposomal lipid A is an effective adjuvant for the delivery of antigens and for the induction of both cellular and humoral immunity. In this study, we demonstrate that following the third immunization with HIV-1 Gag p24 encapsulated in liposomes containing lipid A [L(p24+LA)], central memory CD8+ T-cells were localized in the spleen and lymph nodes of mice while effector memory CD8+ T-cells and effector CD4+ T-cells were found in the PBMC. Effector CD4+ T-cells were also detected in the spleen and lymph nodes. The predominant cytokine secreted from splenic lymphocytes and lymph nodes was IFN-gamma. In contrast, IL-6 and IL-10 were the major cytokines produced from PBMC. The peptide stimulation indicated that the cytokine responses observed were T-cell specific. The results demonstrate the importance of the adjuvant liposomal lipid A for the induction of HIV-1 Gag p24 -specific CD8+ T-cells, effector CD4+ T-cells, and cytokines with a Th-1 type profile after immunization with L(p24+LA).
Collapse
Affiliation(s)
- Nicholas J Steers
- Division of Retrovirology, USMHRP, Walter Reed Army Institute of Research, Rockville, MD 20850, USA
| | | | | | | | | |
Collapse
|
80
|
Pham NLL, Badovinac VP, Harty JT. A default pathway of memory CD8 T cell differentiation after dendritic cell immunization is deflected by encounter with inflammatory cytokines during antigen-driven proliferation. THE JOURNAL OF IMMUNOLOGY 2009; 183:2337-48. [PMID: 19635915 DOI: 10.4049/jimmunol.0901203] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Inflammatory cytokines induced by infection or vaccination with adjuvant act directly or indirectly on CD8 T cells to modulate their expansion, contraction, and acquisition of memory characteristics. Importantly, the initial exposure of naive T cells to inflammatory cytokines may occur before, during, or after their interaction with stimulating dendritic cells (DC) and it is unknown whether and how the timing of cytokine exposure impacts the CD8 T cell response. In this study, we use an immunization strategy with peptide-coated mature DC that, in the absence of inflammatory cytokines, results in a transient effector phase followed by the accelerated acquisition of memory characteristics by the responding CD8 T cells. Induction of inflammatory cytokines by TLR agonists, at the time of DC immunization or 2-4 days after DC immunization, prevented the early acquisition of memory characteristics by the responding CD8 T cells. Interestingly, although induction of inflammatory cytokines at the time of DC immunization increased the effector response, induction of inflammatory cytokines after DC immunization did not promote further expansion of the responding CD8 T cells but still prevented their early acquisition of memory characteristics. In contrast, induction of inflammatory cytokines 2 days before DC immunization did not prevent the CD8 T cells from early acquisition of memory characteristics. Furthermore, TLR ligand-induced inflammatory cytokines had the most significant impact on the phenotype and function of proliferating CD8 T cells. These data suggest that a default pathway of memory CD8 T cell differentiation is deflected toward sustained effector commitment by encounter with inflammatory cytokines during Ag-driven proliferation.
Collapse
Affiliation(s)
- Nhat-Long L Pham
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
81
|
van Gisbergen KPJM, van Olffen RW, van Beek J, van der Sluijs KF, Arens R, Nolte MA, van Lier RA. Protective CD8 T cell memory is impaired during chronic CD70-driven costimulation. THE JOURNAL OF IMMUNOLOGY 2009; 182:5352-62. [PMID: 19380782 DOI: 10.4049/jimmunol.0802809] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chronic infection results in continuous formation and exhaustion of effector CD8 T cells and in failure of memory CD8 T cell development. Expression of CD70 and other molecules that provide costimulation to T cells is maintained during chronic infection. To analyze the impact of constitutive CD70-driven costimulation, we generated transgenic mice expressing CD70 specifically on T cells. We show that CD70 promoted accumulation of CD8 T cells with characteristics strikingly similar to exhausted effector CD8 T cells found during chronic infection. CD70 on T cells provided costimulation that enhanced primary CD8 T cell responses against influenza. In contrast, memory CD8 T cell maintenance and protection against secondary challenge with influenza was impaired. Interestingly, we found no effect on the formation of either effector or memory CD4 T cells. We conclude that constitutive expression of CD70 is sufficient to deregulate the CD8 T cell differentiation pathway of acute infection reminiscent of events in chronic infection.
Collapse
|
82
|
Zheng H, Matte-Martone C, Jain D, McNiff J, Shlomchik WD. Central memory CD8+ T cells induce graft-versus-host disease and mediate graft-versus-leukemia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:5938-48. [PMID: 19414745 PMCID: PMC9844260 DOI: 10.4049/jimmunol.0802212] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In allogeneic hemopoietic stem cell transplantation, mature donor alphabeta T cells in the allograft promote T cell reconstitution in the recipient and mediate the graft-vs-leukemia (GVL) effect. Unfortunately, donor T cells can attack nonmalignant host tissues and cause graft-vs-host disease (GVHD). It has previously been shown that effector memory T cells not primed to alloantigen do not cause GVHD yet transfer functional T cell memory and mediate GVL. Recently, central memory T cells (T(CM)) have also been reported to not cause GVHD. In contrast, in this study, we demonstrate that purified CD8(+) T(CM) not specifically primed to alloantigens mediate GVHD in the MHC-mismatched C57BL/6 (B6)-->BALB/c and the MHC-matched, multiple minor histocompatibility Ag-mismatched C3H.SW-->B6 strain pairings. CD8(+) T(CM) and naive T cells (T(N)) caused similar histological disease in liver, skin, and bowel. B6 CD8(+) T(CM) and T(N) similarly expanded in BALB/c recipients, and the majority of their progeny produced IFN-gamma upon restimulation. However, in both models, CD8(+) T(CM) induced milder clinical GVHD than did CD8(+) T(N). Nonetheless, CD8(+) T(CM) and T(N) were similarly potent mediators of GVL against a mouse model of chronic-phase chronic myelogenous leukemia. Thus, in contrast to what was previously thought, CD8(+) T(CM) are capable of inducing GVHD and are substantially different from T(EM) but only subtly so from T(N).
Collapse
Affiliation(s)
- Hong Zheng
- Penn State Milton S. Hershey Medical Center, Department of Medicine, Hershey, PA
| | - Catherine Matte-Martone
- Yale Cancer Center and Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Dhanpat Jain
- Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Jennifer McNiff
- Department of Dermatology, Yale University School of Medicine, New Haven, CT
| | - Warren D. Shlomchik
- Yale Cancer Center and Department of Immunobiology, Yale University School of Medicine, New Haven, CT,Correspondence: Warren D. Shlomchik, Yale Comprehensive Cancer Center, PO Box 208032, Yale University School of Medicine, New Haven, CT 06520-8032,
| |
Collapse
|
83
|
WIESEL MELANIE, WALTON SENTA, RICHTER KIRSTEN, OXENIUS ANNETTE. Virus-specific CD8 T cells: activation, differentiation and memory formation. APMIS 2009; 117:356-81. [DOI: 10.1111/j.1600-0463.2009.02459.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
84
|
Cush SS, Flaño E. Protective antigen-independent CD8 T cell memory is maintained during {gamma}-herpesvirus persistence. THE JOURNAL OF IMMUNOLOGY 2009; 182:3995-4004. [PMID: 19299697 DOI: 10.4049/jimmunol.0803625] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ag persistence during high-titer chronic viral infections induces CD8 T cell dysfunction and lack of Ag-independent CD8 T cell memory formation. However, we have a poor understanding of the generation and maintenance of CD8 T cell memory during asymptomatic persistent viral infections, particularly gamma-herpesvirus infections. In this study, we demonstrate that the continuous presence of cognate Ag in the host is not required for the maintenance of CD8 T cell memory during a persistent gamma-herpesvirus infection. Importantly, the Ag-independent CD8 T cell memory that is maintained during gamma-herpesvirus persistence has the capacity to survive long-term under homeostatic conditions and to mount a protective recall response to a secondary encounter with the pathogen. These data highlight the ability of the immune system to maintain a population of protective memory CD8 T cells with capacity for long-term Ag-independent survival in the presence of systemic virus persistence.
Collapse
Affiliation(s)
- Stephanie S Cush
- Center for Vaccines and Immunity, Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | | |
Collapse
|
85
|
|
86
|
T cells from lungs and livers of Francisella tularensis-immune mice control the growth of intracellular bacteria. Infect Immun 2009; 77:2010-21. [PMID: 19237526 DOI: 10.1128/iai.01322-08] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Parenteral and respiratory vaccinations with the intracellular bacterium Francisella tularensis have been studied using the live vaccine strain (LVS) in a mouse model, and spleen cells from immune mice are often used for immunological studies. However, mechanisms of host immunological responses may be different in nonlymphoid organs that are important sites of infection, such as lung and liver. Using parenteral (intradermal) or respiratory (cloud aerosol) vaccination, here we examine the functions of resulting LVS-immune liver or lung cells, respectively. Surprisingly, LVS was considerably more virulent when administered by cloud aerosol than by intranasal instillation, suggesting method-dependent differences in initial localization and/or dissemination patterns. Only low doses were sublethal, and resolution of sublethal cloud aerosol infection was dependent on gamma interferon (IFN-gamma), tumor necrosis factor alpha, and inducible nitric oxide synthase. Nonetheless, survival of cloud aerosol or parenteral infection resulted in the development of a protective immune response against lethal LVS intraperitoneal or aerosol challenge, reflecting development of systemic secondary immunity in both cases. Such immunity was further detected by directly examining the functions of LVS-immune lung or liver lymphocytes in vitro. Lung lymphocytes primed by respiratory infection, as well as liver lymphocytes primed by parenteral infection, clearly controlled in vitro intracellular bacterial growth primarily via mechanisms that were not dependent on IFN-gamma activity. Thus, our results indicate functional similarities between immune T cells residing in spleens, livers, and lungs of LVS-immune mice.
Collapse
|
87
|
van Loenen MM, Hagedoorn RS, Kester MGD, Hoogeboom M, Willemze R, Falkenburg JHF, Heemskerk MHM. Kinetic preservation of dual specificity of coprogrammed minor histocompatibility antigen-reactive virus-specific T cells. Cancer Res 2009; 69:2034-41. [PMID: 19223543 DOI: 10.1158/0008-5472.can-08-2523] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Adoptive transfer of antigen-specific T cells is an attractive strategy for the treatment of hematologic malignancies. It has been shown that T cells recognizing minor histocompatibility antigens (mHag) selectively expressed on hematopoietic cells mediate antileukemic reactivity after allogeneic stem cell transplantation. However, large numbers of T cells with defined specificity are difficult to attain. An attractive strategy to obtain large numbers of leukemia-reactive T cells is retroviral transfer of mHag-specific T-cell receptors (TCR). TCR transfer into T cells specific for persistent viruses may enable these T cells to proliferate both after encountering with viral antigens as well as mHags, increasing the possibility of in vivo survival. We analyzed whether the dual specificity of the TCR-transferred T cells after repetitive stimulation via either the introduced antileukemic HA-2-TCR or the endogenous cytomegalovirus (CMV) specific CMV-TCR was preserved. We show that after repetitive stimulation, T cells skew to a population predominantly expressing the triggered TCR. However, HA-2-TCR-transferred CMV-specific T cells with high antileukemic HA-2-TCR expression but low CMV-TCR expression were able to persist and proliferate after repetitive stimulation with pp65. Moreover, HA-2-TCR-transferred CMV-specific T cells remained dual specific after repetitive stimulation and TCR expression could be reverted after additional stimulation via the previously nonstimulated TCR, restoring high-avidity interactions. These data imply persistence of TCR-transferred virus-specific T cells with both antileukemic and antivirus reactivity in vivo.
Collapse
Affiliation(s)
- Marleen M van Loenen
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
88
|
Abstract
Over the last decade, significant advances have been made in the methodology for studying immune responses in vivo. It is now possible to follow almost every aspect of pathogen-specific immunity using in vivo models that incorporate physiological infectious doses and natural routes of infection. This new ability to study immunity in a relevant physiological context will greatly expand our understanding of the dynamic interplay between host and pathogen. Visualizing the resolution of primary infection and the development of long-term immunological memory should also aid the development of new vaccines and therapeutics for infectious diseases. In this review, we will describe the application of in vivo visualization technology to Salmonella infection, describe our current understanding of Salmonella-specific immunity, and discuss some unanswered questions that remain in this model.
Collapse
Affiliation(s)
- James J. Moon
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, MN, 55455
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455
| | - Stephen J. McSorley
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, 55455
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455
- Center for Infectious Diseases & Microbiology Translational Research, University of Minnesota Medical School, Minneapolis, MN, 55455
| |
Collapse
|
89
|
Luissint AC, Lutz PG, Calderwood DA, Couraud PO, Bourdoulous S. JAM-L-mediated leukocyte adhesion to endothelial cells is regulated in cis by alpha4beta1 integrin activation. ACTA ACUST UNITED AC 2008; 183:1159-73. [PMID: 19064666 PMCID: PMC2600739 DOI: 10.1083/jcb.200805061] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Junctional adhesion molecules (JAMs) are endothelial and epithelial adhesion molecules involved in the recruitment of circulating leukocytes to inflammatory sites. We show here that JAM-L, a protein related to the JAM family, is restricted to leukocytes and promotes their adhesion to endothelial cells. Cis dimerization of JAM-L is required to engage in heterophilic interactions with its cognate counter-receptor CAR (coxsackie and adenovirus receptor). Interestingly, JAM-L expressed on neutrophils binds CAR independently of integrin activation. However, on resting monocytes and T lymphocytes, which express the integrin VLA-4, JAM-L molecules engage in complexes with VLA-4 and mainly accumulate in their monomeric form. Integrin activation is required for the dissociation of JAM-L–VLA-4 complexes and the accumulation of functional JAM-L dimers, which indicates that the leukocyte integrin VLA-4 controls JAM-L function in cis by controlling its dimerization state. This provides a mechanism through which VLA-4 and JAM-L functions are coordinately regulated, allowing JAM-L to strengthen integrin-dependent adhesion of leukocytes to endothelial cells.
Collapse
Affiliation(s)
- Anny-Claude Luissint
- Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Paris F-75014, France
| | | | | | | | | |
Collapse
|
90
|
Wahid R, Salerno-Gonçalves R, Tacket CO, Levine MM, Sztein MB. Generation of specific effector and memory T cells with gut- and secondary lymphoid tissue- homing potential by oral attenuated CVD 909 typhoid vaccine in humans. Mucosal Immunol 2008; 1:389-98. [PMID: 19079203 PMCID: PMC3215293 DOI: 10.1038/mi.2008.30] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Induction of effective memory T cells is likely to be critical to the level and duration of protection elicited by novel live oral typhoid vaccines. Using cells from volunteers who ingested Salmonella Typhi vaccine strain CVD 909, we characterized the induction of interferon (IFN)-gamma-secreting central (T(CM), CD45RO(+)CD62L(+)) and effector (T(EM), CD45RO(+)CD62L(-)) memory T populations, and their gut-homing potential based on integrin alpha4/beta7 expression. Both CD4(+) T(EM) and T(CM) populations secreted IFN-gamma. However, although CD4(+) T(EM) expressed, or not, integrin alpha(4)/beta(7), CD4(+) T(CM) cells were predominantly integrin alpha(4)/beta(7)(+). In contrast, IFN-gamma-secreting CD8(+) cells were predominantly classical T(EM) and CD45RA(+) T(EM) (T(EMRA), CD45RO(-)CD62L(-)) subsets. However, although CD8(+) T(EM) expressed, or not, integrin alpha(4)/beta(7), CD8(+) T(EMRA) were predominantly integrin alpha(4)/beta(7)(+). This is the first demonstration that oral immunization of humans with S. Typhi elicits diverse IFN-gamma-secreting CD4(+) and CD8(+) T(CM) and T(EM) subsets able to migrate to the gut and other lymphoid tissues.
Collapse
Affiliation(s)
- R Wahid
- Department of Pediatrics, Center for Vaccine Development, University of Maryland, Baltimore, Maryland, USA
| | - R Salerno-Gonçalves
- Department of Pediatrics, Center for Vaccine Development, University of Maryland, Baltimore, Maryland, USA
| | - CO Tacket
- Department of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - MM Levine
- Department of Pediatrics, Center for Vaccine Development, University of Maryland, Baltimore, Maryland, USA,Department of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - MB Sztein
- Department of Pediatrics, Center for Vaccine Development, University of Maryland, Baltimore, Maryland, USA,Department of Medicine, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
91
|
Lapillonne H, Leclerc A, Ulinski T, Balu L, Garnier A, Dereuddre-Bosquet N, Watier H, Schlageter MH, Deschênes G. Stem cell mobilization in idiopathic steroid-sensitive nephrotic syndrome. Pediatr Nephrol 2008; 23:1251-6. [PMID: 18458957 DOI: 10.1007/s00467-008-0793-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Revised: 01/21/2008] [Accepted: 01/21/2008] [Indexed: 12/13/2022]
Abstract
Steroid-sensitive nephrotic syndrome (SSNS) is classically thought to be a T-cell disorder. The aim of this study was to examine whether or not thymus homeostasis was affected in SSNS. Mature and naive T cell recent thymic emigrants were quantified in the peripheral blood of nephrotic patients and controls. Because the generation of new T cells by the thymus ultimately depends on hematopoietic stem cells, CD34+ cells were also included in the study. Nineteen patients with SSNS during relapse, 13 with SSNS during proteinuria remission, and 18 controls were studied. Cell-surface markers (CD3, CD4, CD8, CD19, CD16, CD56, CD45RA, CD62L, CD34, and CD38) were analyzed by flow cytometric analysis. T-cell rearrangement excision circles (TRECs) were quantified in CD2+ cells by real-time polymerase chain reaction. Stroma cell-derived factor-1 (SDF-1) genotype and metalloproteinase-9 (MMP-9) plasma levels were also determined. Mature T cells (CD4+ and CD8+), circulating naive T cells (CD62L+ and CD3+ CD62L+), and recent thymic emigrants (CD45RA+) as well as TRECs, that measure thymus production, had a similar level in the three groups of patients. Conversely, CD34+ hematopoietic stem cells displayed a two-fold increase in SSNS patients during relapse either compared with controls or SSNS patients at remission. In addition, compared with controls, SSNS patients at remission displayed (1) a decrease in CD19+ cells (B cells) and (2) an increase in CD16CD56+ cells [natural killer (NK) cells]. In conclusion, thymus homeostasis is not significantly affected in nephrotic patients. Hematopoietic stem-cell mobilization at proteinuria relapse, as well as changes in B and NK cells during remission, suggest that SSNS might be due to a general disturbance of hematopoietic and immune cell trafficking.
Collapse
Affiliation(s)
- Hélène Lapillonne
- Laboratoire d'Hématologie, Hôpital Armand-Trousseau, Assistance Publique-Hôpitaux de Paris, 26 avenue du Docteur Arnold-Netter, 75571 Paris Cedex 12, France
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Schindler M, Schmökel J, Specht A, Li H, Münch J, Khalid M, Sodora DL, Hahn BH, Silvestri G, Kirchhoff F. Inefficient Nef-mediated downmodulation of CD3 and MHC-I correlates with loss of CD4+T cells in natural SIV infection. PLoS Pathog 2008; 4:e1000107. [PMID: 18636106 PMCID: PMC2444047 DOI: 10.1371/journal.ppat.1000107] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Accepted: 06/19/2008] [Indexed: 01/22/2023] Open
Abstract
Recent data suggest that Nef-mediated downmodulation of TCR-CD3 may protect SIVsmm-infected sooty mangabeys (SMs) against the loss of CD4+ T cells. However, the mechanisms underlying this protective effect remain unclear. To further assess the role of Nef in nonpathogenic SIV infection, we cloned nef alleles from 11 SIVsmm-infected SMs with high (>500) and 15 animals with low (<500) CD4+ T-cells/µl in bulk into proviral HIV-1 IRES/eGFP constructs and analyzed their effects on the phenotype, activation, and apoptosis of primary T cells. We found that not only efficient Nef-mediated downmodulation of TCR-CD3 but also of MHC-I correlated with preserved CD4+ T cell counts, as well as with high numbers of Ki67+CD4+ and CD8+CD28+ T cells and reduced CD95 expression by CD4+ T cells. Moreover, effective MHC-I downregulation correlated with low proportions of effector and high percentages of naïve and memory CD8+ T cells. We found that T cells infected with viruses expressing Nef alleles from the CD4low SM group expressed significantly higher levels of the CD69, interleukin (IL)-2 and programmed death (PD)-1 receptors than those expressing Nefs from the CD4high group. SIVsmm Nef alleles that were less active in downmodulating TCR-CD3 were also less potent in suppressing the activation of virally infected T cells and subsequent cell death. However, only nef alleles from a single animal with very low CD4+ T cell counts rendered T cells hyper-responsive to activation, similar to those of HIV-1. Our data suggest that Nef may protect the natural hosts of SIV against the loss of CD4+ T cells by at least two mechanisms: (i) downmodulation of TCR-CD3 to prevent activation-induced cell death and to suppress the induction of PD-1 that may impair T cell function and survival, and (ii) downmodulation of MHC-I to reduce CTL lysis of virally infected CD4+ T cells and/or bystander CD8+ T cell activation. The accessory Nef protein is commonly considered a “pathogenicity” factor of primate lentiviruses. However, SIVs do not cause disease in their natural hosts, although they all encode nef genes and sustain high levels of viremia. To better understand the role of Nef in natural nonpathogenic SIV infection, we compared the function of Nef alleles from two groups of SIVsmm-infected sooty mangabeys: (i) those that maintained normal CD4+ T cell counts and (ii) a small subset (10%–15%) of animals that exhibited a considerable loss of CD4+ helper T cells. We found that the efficiency of two specific Nef functions, i.e., downmodulation of TCR-CD3 and MHC-I, correlated with preserved CD4+ T cell homeostasis, as well as with other immunological features, such as high numbers of proliferating CD4+ Ki67+ T cells. Moreover, lack of CD3 surface expression was associated with low levels of apoptosis and PD-1 expression by virally infected T cells. Thus, the ability of Nef to remove TCR-CD3 and MHC-I from the cell surface may help the natural hosts of SIV to maintain normal CD4+ T cell counts despite high levels of viral replication by preventing activation-induced cell death and CTL lysis of infected T cells and/or CD8+ T cell activation.
Collapse
Affiliation(s)
| | - Jan Schmökel
- Institute of Virology, University of Ulm, Germany
| | - Anke Specht
- Institute of Virology, University of Ulm, Germany
| | - Hui Li
- Departments of Medicine and Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jan Münch
- Institute of Virology, University of Ulm, Germany
| | | | - Donald L. Sodora
- University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Beatrice H. Hahn
- Departments of Medicine and Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Guido Silvestri
- Yerkes Regional Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | | |
Collapse
|
93
|
van Stipdonk M, Sluijter M, Han W, Offringa R. Development of CTL memory despite arrested clonal expansion. Eur J Immunol 2008; 38:1839-46. [DOI: 10.1002/eji.200737974] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
94
|
Zhang Y, Ohkuri T, Wakita D, Narita Y, Chamoto K, Kitamura H, Nishimura T. Sialyl lewisx
antigen-expressing human CD4+
T and CD8+
T cells as initial immune responders in memory phenotype subsets. J Leukoc Biol 2008; 84:730-5. [DOI: 10.1189/jlb.0907599] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
95
|
Kodera M, Grailer JJ, Karalewitz APA, Subramanian H, Steeber DA. T lymphocyte migration to lymph nodes is maintained during homeostatic proliferation. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2008; 14:211-224. [PMID: 18312727 DOI: 10.1017/s1431927608080215] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The immune system maintains appropriate cell numbers through regulation of cell proliferation and death. Normal tissue distribution of lymphocytes is maintained through expression of specific adhesion molecules and chemokine receptors such as L-selectin and CCR7, respectively. Lymphocyte insufficiency or lymphopenia induces homeostatic proliferation of existing lymphocytes to increase cell numbers. Interestingly, homeostatic proliferation of T lymphocytes induces a phenotypic change from naïve- to memory-type cell. Naïve T cells recirculate between blood and lymphoid tissues whereas memory T cells migrate to nonlymphoid sites such as skin and gut. To assess effects of homeostatic proliferation on migratory ability of T cells, a murine model of lymphopenia-induced homeostatic proliferation was used. Carboxyfluorescein diacetate, succinimidyl ester-labeled wild-type splenocytes were adoptively transferred into recombination activation gene-1-deficient mice and analyzed by flow cytometry, in vitro chemotactic and in vivo migration assays, and immunofluorescence microscopy. Homeostatically proliferated T cells acquired a mixed memory-type CD44high L-selectinhigh CCR7low phenotype. Consistent with this, chemotaxis to secondary lymphoid tissue chemokine in vitro was reduced by 22%-34%. By contrast, no differences were found for migration or entry into lymph nodes during in vivo migration assays. Therefore, T lymphocytes that have undergone homeostatic proliferation recirculate using mechanisms similar to naïve T cells.
Collapse
Affiliation(s)
- Masanari Kodera
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | | | | | | | | |
Collapse
|
96
|
Obar JJ, Khanna KM, Lefrançois L. Endogenous naive CD8+ T cell precursor frequency regulates primary and memory responses to infection. Immunity 2008; 28:859-69. [PMID: 18499487 PMCID: PMC2836785 DOI: 10.1016/j.immuni.2008.04.010] [Citation(s) in RCA: 346] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 02/21/2008] [Accepted: 04/04/2008] [Indexed: 10/22/2022]
Abstract
Through genetic recombination, the adaptive immune system generates a diverse T cell repertoire allowing recognition of a vast spectrum of foreign antigens. Any given CD8+ T cell specificity is thought to be rare, but none have been directly quantified. Here, major histocompatibility complex tetramer and magnetic-bead technology were coupled to quantitate naive antigen-specific CD8+ T cells and the early response to infection. Among six specificities measured, the number of naive antigen-specific precursors ranged from approximately 80 to 1200 cells/mouse. After vesicular stomatitis virus infection, the antigen-specific CD8+ T cell response occurred in discrete phases: prolonged activation of a subset of cells over the first 72 hr followed by a rapid proliferative burst. Naive precursor frequency altered response kinetics and regulated immunodominance, as well as the time required for the responding population to shift toward CD62L(hi) memory cells. Thus, initial endogenous precursor frequencies were surprisingly diverse and not only regulated initial immune response characteristics but also controlled memory CD8+ T cell lineage decisions.
Collapse
Affiliation(s)
- Joshua J. Obar
- Department of Immunology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington CT 06030-1319, USA
| | - Kamal M. Khanna
- Department of Immunology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington CT 06030-1319, USA
| | - Leo Lefrançois
- Department of Immunology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington CT 06030-1319, USA
| |
Collapse
|
97
|
Sinclair LV, Finlay D, Feijoo C, Cornish GH, Gray A, Ager A, Okkenhaug K, Hagenbeek TJ, Spits H, Cantrell DA. Phosphatidylinositol-3-OH kinase and nutrient-sensing mTOR pathways control T lymphocyte trafficking. Nat Immunol 2008; 9:513-21. [PMID: 18391955 PMCID: PMC2857321 DOI: 10.1038/ni.1603] [Citation(s) in RCA: 306] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Accepted: 03/04/2008] [Indexed: 01/22/2023]
Abstract
Phosphatidylinositol-3-OH kinase (PI(3)K) and the nutrient sensor mTOR are evolutionarily conserved regulators of cell metabolism. Here we show that PI(3)K and mTOR determined the repertoire of adhesion and chemokine receptors expressed by T lymphocytes. The key lymph node-homing receptors CD62L (L-selectin) and CCR7 were highly expressed on naive T lymphocytes but were downregulated after immune activation. CD62L downregulation occurred through ectodomain proteolysis and suppression of gene transcription. The p110delta subunit of PI(3)K controlled CD62L proteolysis through mitogen-activated protein kinases, whereas control of CD62L transcription by p110delta was mediated by mTOR through regulation of the transcription factor KLF2. PI(3)K-mTOR nutrient-sensing pathways also determined expression of the chemokine receptor CCR7 and regulated lymphocyte trafficking in vivo. Hence, lymphocytes use PI(3)K and mTOR to match metabolism and trafficking.
Collapse
Affiliation(s)
- Linda V Sinclair
- Department of Cell Biology and Immunology, University of Dundee, DD1 5EH, UK
| | - David Finlay
- Department of Cell Biology and Immunology, University of Dundee, DD1 5EH, UK
| | - Carmen Feijoo
- Department of Cell Biology and Immunology, University of Dundee, DD1 5EH, UK
| | - Georgina H Cornish
- Immune Cell Biology, The National Institute for Medical Research, London, NW7 1AA, UK
| | - Alex Gray
- Division of Molecular Physiology, University of Dundee, DD1 5EH, UK
| | - Ann Ager
- Department of Medical Biochemistry and Immunology, Cardiff University, CF14 4XN, UK
| | - Klaus Okkenhaug
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB2 4AT, UK
| | - Thijs J Hagenbeek
- Department of Immunology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
- Department of Immunology Discovery, Genentech Inc., South San Francisco, California CA 94080, USA
| | - Hergen Spits
- Department of Immunology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
- Department of Immunology Discovery, Genentech Inc., South San Francisco, California CA 94080, USA
| | - Doreen A Cantrell
- Department of Cell Biology and Immunology, University of Dundee, DD1 5EH, UK
| |
Collapse
|
98
|
Whitmire JK, Eam B, Whitton JL. Tentative T cells: memory cells are quick to respond, but slow to divide. PLoS Pathog 2008; 4:e1000041. [PMID: 18404208 PMCID: PMC2275797 DOI: 10.1371/journal.ppat.1000041] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Accepted: 03/10/2008] [Indexed: 11/19/2022] Open
Abstract
T cell memory is a cornerstone of protective immunity, and is the key element in successful vaccination. Upon encountering the relevant pathogen, memory T cells are thought to initiate cell division much more rapidly than their naïve counterparts, and this is thought to confer a significant biological advantage upon an immune host. Here, we use traceable TCR-transgenic T cells to evaluate this proposed characteristic in CD4+ and CD8+ memory T cells. We find that, even in the presence of abundant antigen that was sufficient to induce in vivo IFNgamma production by memory T cells, both memory and naïve T cells show an extended, and indistinguishable, delay in the onset of proliferation. Although memory cells can detect, and respond to, virus infection within a few hours, their proliferation did not begin until approximately 3 days after infection, and occurred simultaneously in all anatomical compartments. Thereafter, cell division was extraordinarily rapid for both naïve and memory cells, with the latter showing a somewhat accelerated accumulation. We propose that, by permitting memory T cells to rapidly exert their effector functions while delaying the onset of their proliferation, evolution has provided a safeguard that balances the risk of infection against the consequences of severe T cell-mediated immunopathology.
Collapse
Affiliation(s)
- Jason K. Whitmire
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California, United States of America
| | - Boreth Eam
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California, United States of America
| | - J. Lindsay Whitton
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California, United States of America
| |
Collapse
|
99
|
Kalia V, Sarkar S, Ahmed R. Fine-tuning CD4+ central memory T cell heterogeneity by strength of stimulation. Eur J Immunol 2008; 38:15-9. [PMID: 18157815 DOI: 10.1002/eji.200738044] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The memory T cell pool serves as a relatively long-lived heterogeneous repository of antigen-experienced T cells that "remember" previous encounters with antigen. While heterogeneity in the memory T cell pool is now well established, signals regulating the generation of this memory T cell heterogeneity are not fully understood. Two articles in this issue of the European Journal of Immunology highlight the importance of the strength of antigenic stimulation in regulating the generation of phenotypically and functionally distinct CD4(+) T cell memory subsets. New insights are also provided into key molecular players that likely mediate differences in homeostatic and secondary expansion between the memory subsets.
Collapse
Affiliation(s)
- Vandana Kalia
- Emory Vaccine Center, Emory School of Medicine, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
100
|
Sarkar S, Kalia V, Haining WN, Konieczny BT, Subramaniam S, Ahmed R. Functional and genomic profiling of effector CD8 T cell subsets with distinct memory fates. ACTA ACUST UNITED AC 2008; 205:625-40. [PMID: 18316415 PMCID: PMC2275385 DOI: 10.1084/jem.20071641] [Citation(s) in RCA: 502] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
An important question in memory development is understanding the differences between effector CD8 T cells that die versus effector cells that survive and give rise to memory cells. In this study, we provide a comprehensive phenotypic, functional, and genomic profiling of terminal effectors and memory precursors. Using killer cell lectin-like receptor G1 as a marker to distinguish these effector subsets, we found that despite their diverse cell fates, both subsets possessed remarkably similar gene expression profiles and functioned as equally potent killer cells. However, only the memory precursors were capable of making interleukin (IL) 2, thus defining a novel effector cell that was cytotoxic, expressed granzyme B, and produced inflammatory cytokines in addition to IL-2. This effector population then differentiated into long-lived protective memory T cells capable of self-renewal and rapid recall responses. Experiments to understand the signals that regulate the generation of terminal effectors versus memory precursors showed that cells that continued to receive antigenic stimulation during the later stages of infection were more likely to become terminal effectors. Importantly, curtailing antigenic stimulation toward the tail end of the acute infection enhanced the generation of memory cells. These studies support the decreasing potential model of memory differentiation and show that the duration of antigenic stimulation is a critical regulator of memory formation.
Collapse
Affiliation(s)
- Surojit Sarkar
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|