1
|
Berton RR, Heidarian M, Kannan SK, Shah M, Butler NS, Harty JT, Badovinac VP. Accurate enumeration of pathogen-specific and virtual memory CD8 T cells after infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf007. [PMID: 40167212 DOI: 10.1093/jimmun/vkaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 01/04/2025] [Indexed: 04/02/2025]
Abstract
Establishing the magnitude and kinetics of polyclonal Ag-specific CD8 T-cell responses, in addition to their functional fitness, is critical for evaluating a host's ability to respond to different kinds of infections and/or immunizations. To track CD8 T-cell responses during infection, a surrogate-activation-marker approach (CD8αloCD11ahi) is used to distinguish naïve and Ag-experienced effector/memory CD8 T cells in vivo. However, semidifferentiated virtual memory (Tvm) CD8 T cells have recently been identified in uninfected/unmanipulated mice that display a phenotype similar to Ag-experienced cells. Therefore, magnitude and breadth of CD8 T-cell responses may be overestimated when responses are profiled using only CD8α/CD11a markers. Thus, to precisely define and distinguish Tvm from pathogen-specific CD8 T cells during bacterial, parasitic, and viral infections, pathogen-specific sensor TCR-Tg cells were adoptively transferred prior to challenge. We demonstrate that Tvm CD8 T cells are found in CD8αloCD11ahi-defined Ag-experienced CD8 T cells but can be parsed out in infected host with their CD49d-CD44hiCD122hi expression pattern. However, this approach presents potential limitations as CD49d+ Ag-specific CD8 T cells can lose CD49d expression and adopt a Tvm-like phenotype depending on their Ag-stimulation history, age, and naïve CD8 T-cell precursor frequency before the infection. Importantly, Tvm cells contribute to the breadth of the CD8 T-cell response, and their contribution depends on type of infection, time after infection, and tissue examined. Thus, these data define limitations in our ability to resolve between pathogen/Ag-specific and Tvm CD8 T-cell responses during infection, a notion of direct relevance for experimental murine studies designed to follow CD8 T-cell responses in vivo.
Collapse
Affiliation(s)
- Roger R Berton
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| | - Mohammad Heidarian
- Department of Pathology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology Graduate Programs, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| | - Shravan Kumar Kannan
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| | - Manan Shah
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Microbiology and Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| | - Noah S Butler
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Microbiology and Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| | - John T Harty
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology Graduate Programs, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| | - Vladimir P Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
- Department of Pathology Graduate Programs, University of Iowa, 500 Newton Rd, 1020 ML, Iowa City, IA 52242, USA
| |
Collapse
|
2
|
Liu J, Liu Y, Panda S, Liu A, Lei J, Burd I. Type 1 Cytotoxic T Cells Increase in Placenta after Intrauterine Inflammation. Front Immunol 2021; 12:718563. [PMID: 34566975 PMCID: PMC8456007 DOI: 10.3389/fimmu.2021.718563] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/23/2021] [Indexed: 12/16/2022] Open
Abstract
CD8+ T cells recognize non-self antigen by MHC class I molecules and kill the target cells by the release of proinflammatory cytokines such as interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α). Our group previously reported an increase of CD8+ T‐cell trafficking in the placenta with exposure to Lipopolysaccharides (LPS). CD8+ cytotoxic T cells have been classified into distinct subsets based upon cytokine production: Tc1 cells produce IFN-γ, Tc2 cells produce interleukin 4 (IL-4). Accordingly, the purpose of this research is to analyze the subsets of placenta CD8+ T cells. We hypothesized that LPS injection would induce a change of properties of CD8+ T cell and Tc1/Tc2 ratio. We investigated the subsets of CD8+ T cell infiltration to placenta and their specific function in response to LPS-induced inflammation in a mouse model. At embryonic (E) day 17, pregnant CD-1 dams received an intrauterine injection of 25 µg LPS in100 μl PBS or 100 μl of PBS only. Flow cytometry was used to quantify CD8+ T cells, evaluate the phenotype and subtypes, and detect markers of Tc1 and Tc2 cells in placenta, at 6 hours and 24 hours post injection (hpi). Intracellular staining and flow cytometry were performed to characterize cytokines produced by CD8+ T cells. Standard statistical analysis were employed. After 6 and 24 hours of LPS injection, total CD8 T cells increased (P<0.05). Tc1 cells expanded (P<0.05) in LPS-treated dams compared with the PBS group. The Tc1/Tc2 ratio was significantly higher in the LPS group than the PBS group (P<0.05). The expression of TNF-α and IFN-γ were increased in LPS group both at 6hpi and 24 hpi (P<0.05). We identified functional placental CD8+ T cell subtypes and found a significant increase ratio of Tc1/Tc2. Following IUI, CD8+ T cells induced inflammatory response in the placenta primarily via the production of Type 1 cytokines such as IFN-γ and TNF-α. We have provided evidence of a Tc1-bias response and cytokines in the mouse model of IUI.
Collapse
Affiliation(s)
- Jin Liu
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yang Liu
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Snigdha Panda
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Anguo Liu
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jun Lei
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
3
|
The appropriate frequency and function of decidual Tim-3 +CTLA-4 +CD8 + T cells are important in maintaining normal pregnancy. Cell Death Dis 2019; 10:407. [PMID: 31138782 PMCID: PMC6538701 DOI: 10.1038/s41419-019-1642-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 12/21/2022]
Abstract
Maternal decidual CD8+ T (dCD8+ T) cells must integrate the antithetical demands of maternal–fetal tolerance and anti-viral immunity to establish a successful pregnancy. T-cell immunoglobulin mucin-3 (Tim-3) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are two important co-inhibitory molecules that regulating CD8+ T cells responses during infection and tumor. In the present study, we examined the co-expression of Tim-3 and CTLA-4 on CD8+ T cells during pregnancy and found the higher frequency of Tim-3+CTLA-4+dCD8+ T cells in response to trophoblasts. This Tim-3+CTLA-4+dCD8+ T cells subset showed an active status and produced more anti-inflammatory cytokines. Furthermore, the decreased number and altered function of Tim-3+CTLA-4+dCD8+ T cells correlated to miscarriage. Combined blocking Tim-3 and CTLA-4 pathways were highly effective in inhibiting the production of anti-inflammatory cytokines and were detrimental to the maintenance of pregnancy. Together, these findings supported that Tim-3 and CTLA-4 pathways might play positive roles in the establishment and/or maintenance of maternal–fetal tolerance so to promote the maintenance of normal pregnancy. So the reproductive safety must be considered, especially when anti-Tim-3/CTLA-4 antibody (and other immune checkpoint inhibitors) are used in pregnancy.
Collapse
|
4
|
Li N, Zhang L, Zheng B, Li W, Liu J, Zhang H, Zeng R. RSV recombinant candidate vaccine G1F/M2 with CpG as an adjuvant prevents vaccine-associated lung inflammation, which may be associated with the appropriate types of immune memory in spleens and lungs. Hum Vaccin Immunother 2019; 15:2684-2694. [PMID: 31021703 DOI: 10.1080/21645515.2019.1596710] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major respiratory pathogen in infants. The early formalin-inactivated RSV not only failed to protect infants against infection, but also was associated with enhanced pulmonary inflammatory disease upon natural infection. A safe and effective vaccine should prevent the inflammatory disease and provide protection. Immune memory is the cornerstone of vaccines. In this study, we evaluated three types of immune memory T cells, antibodies, and lung inflammation of a vaccine candidate G1F/M2, which includes a neutralizing epitope fragment of RSV G protein and a cytotoxic T lymphocyte epitope of M2 protein, with toll-like receptor 9 agonist CpG2006 as an adjuvant by intranasal (i.n.) and intraperitoneal (i.p.) immunization protocols. The results indicated that immunization of mice with G1F/M2 + CpG i.p. induced significantly higher level of CD4+ or CD8+ central memory (TCM), Th1-type effector memory (TEM), and balanced ratio of IgG1/IgG2a, but lower level of lung tissue-resident memory (TRM), compared with immunization with G1F/M2 + CpG i.n., G1F/M2 i.n., or G1F/M2 i.p. Following RSV challenge, the mice immunized with G1F/M2 + CpG i.p. showed higher level of Th1-type responses, remarkably suppressed inflammatory cytokines and histopathology in lungs, compared with mice immunized with G1F/M2 + CpG i.n., G1F/M2 i.n., or G1F/M2 i.p. These results suggested that high level of TCM and Th1 type of TEM in spleens may contribute to inhibition of lung inflammation, while high level of TRM in lungs and lack of or weak Th1-type immune memory in spleens may promote lung inflammation following RSV challenge.
Collapse
Affiliation(s)
- Na Li
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, PR China.,Department of Microbiology and Immunology, Xingtai Medical College, Xingtai, Hebei, PR China
| | - Ling Zhang
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Boyang Zheng
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Wenjian Li
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Jianxun Liu
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, PR China.,Department of Microbiology and Immunology, Xingtai Medical College, Xingtai, Hebei, PR China
| | - Huixian Zhang
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Ruihong Zeng
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, PR China.,Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei, China
| |
Collapse
|
5
|
Jahn ML, Steffensen MA, Christensen JP, Thomsen AR. Analysis of adenovirus-induced immunity to infection with Listeria monocytogenes: Fading protection coincides with declining CD8 T cell numbers and phenotypic changes. Vaccine 2018; 36:2825-2832. [PMID: 29627230 DOI: 10.1016/j.vaccine.2018.03.080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 03/23/2018] [Accepted: 03/28/2018] [Indexed: 01/09/2023]
Abstract
Defining correlates of T cell mediated protection is important in order to accelerate the development of efficient T cell based vaccines conferring long-term immunity. Extensive studies have provided important insight regarding the characteristics and functional properties of the effector and memory CD8 T cells induced by viral vector based vaccines. However, long-term protection has been difficult to achieve with T cell inducing vaccines, and the determinants underlying this loss in protection over time are still not fully defined. In this study we analyzed different parameters of the CD8 T cell response as a function of time after vaccination with a human serotype 5 adenovector expressing the glycoprotein (GP) of LCMV tethered to the MHC class II-associated invariant chain. Using this vector we have previously found that CD8 T cells mediate protection from challenge with GP-expressing Listeria monocytogenes at 60 days post vaccination, but only little protection after further 60 days, and we now confirm this observation. A comparison of vaccine-primed CD8 T cells early and late after vaccination revealed a minor decline in the overall numbers of antigen specific memory CD8 T cells during this interval. More importantly, we also observed phenotypic changes over time with a distinct decline in the frequency and number of KLRG1+ CD8 T cells, and, notably, adoptive transfer studies confirmed that memory CD8 T cells expressing KLRG1 are central to protection from systemic L. monocytogenes infection. Together these findings imply that multiple factors including changes in memory T cell numbers and phenotypic composition over time influence the longevity of CD8 T-cell mediated protection.
Collapse
Affiliation(s)
- Marie Louise Jahn
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Allan Randrup Thomsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
6
|
Erkes DA, Smith CJ, Wilski NA, Caldeira-Dantas S, Mohgbeli T, Snyder CM. Virus-Specific CD8 + T Cells Infiltrate Melanoma Lesions and Retain Function Independently of PD-1 Expression. THE JOURNAL OF IMMUNOLOGY 2017; 198:2979-2988. [PMID: 28202614 DOI: 10.4049/jimmunol.1601064] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 01/20/2017] [Indexed: 12/27/2022]
Abstract
It is well known that CD8+ tumor-infiltrating lymphocytes (TILs) are correlated with positive prognoses in cancer patients and are used to determine the efficacy of immune therapies. Although it is generally assumed that CD8+ TILs will be tumor-associated Ag (TAA) specific, it is unknown whether CD8+ T cells with specificity for common pathogens also infiltrate tumors. If so, the presence of these T cells could alter the interpretation of prognostic and diagnostic TIL assays. We compared TAA-specific and virus-specific CD8+ T cells in the same tumors using murine CMV, a herpesvirus that causes a persistent/latent infection, and vaccinia virus, a poxvirus that is cleared by the host. Virus-specific CD8+ TILs migrated into cutaneous melanoma lesions during acute infection with either virus, after a cleared vaccinia virus infection, and during a persistent/latent murine CMV infection. Virus-specific TILs developed independently of viral Ag in the tumor and, interestingly, expressed low or intermediate levels of full-length PD-1 in the tumor environment. Importantly, PD-1 expression could be markedly induced by Ag but did not correlate with dysfunction for virus-specific TILs, in sharp contrast to TAA-specific TILs in the same tumors. These data suggest that CD8+ TILs can reflect an individual's immune status, rather than exclusively representing TAA-specific T cells, and that PD-1 expression on CD8+ TILs is not always associated with repeated Ag encounter or dysfunction. Thus, functional virus-specific CD8+ TILs could skew the results of prognostic or diagnostic TIL assays.
Collapse
Affiliation(s)
- Dan A Erkes
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Corinne J Smith
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Nicole A Wilski
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Sofia Caldeira-Dantas
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107.,Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal; and.,ICVS/3Bs, PT Government Associated Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Toktam Mohgbeli
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Christopher M Snyder
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107;
| |
Collapse
|
7
|
Maggioli MF, Palmer MV, Thacker TC, Vordermeier HM, McGill JL, Whelan AO, Larsen MH, Jacobs WR, Waters WR. Increased TNF-α/IFN-γ/IL-2 and Decreased TNF-α/IFN-γ Production by Central Memory T Cells Are Associated with Protective Responses against Bovine Tuberculosis Following BCG Vaccination. Front Immunol 2016; 7:421. [PMID: 27799930 PMCID: PMC5066095 DOI: 10.3389/fimmu.2016.00421] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/27/2016] [Indexed: 12/30/2022] Open
Abstract
Central memory T cell (Tcm) and polyfunctional CD4 T cell responses contribute to vaccine-elicited protection with both human and bovine tuberculosis (TB); however, their combined role in protective immunity to TB is unclear. To address this question, we evaluated polyfunctional cytokine responses by CD4 T cell effector/memory populations from bacille Calmette–Guerin (BCG) vaccinated and non-vaccinated calves by flow cytometry prior to and after aerosol challenge with virulent Mycobacterium bovis. Polyfunctional cytokine expression patterns in the response by Tcm, effector memory, and effector T cell subsets were similar between BCG-vaccinated and M. bovis-infected calves, only differing in magnitude (i.e., infected > vaccinated). BCG vaccination, however, did alter the kinetics of the ensuing response to virulent M. bovis infection. Early after challenge (3 weeks post-infection), non-vaccinates had greater antigen-specific interferon-γ (IFN-γ)/tumor necrosis factor-α (TNF-α) and lesser IFN-γ/TNF-α/IL-2 responses by Tcm cells than did vaccinated animals. Importantly, these differences were also associated with mycobacterial burden upon necropsy. Polyfunctional responses to ESAT-6:CFP10 (antigens not synthesized by BCG strains) were detected in memory subsets, as well as in effector cells, as early as 3 weeks after challenge. These findings suggest that cell fate divergence may occur early after antigen priming in the response to bovine TB and that memory and effector T cells may expand concurrently during the initial phase of the immune response. In summary, robust IFN-γ/TNF-α response by Tcm cells is associated with greater mycobacterial burden, while IFN-γ/TNF-α/IL-2 response by Tcm cells are indicative of a protective response to bovine TB.
Collapse
Affiliation(s)
- Mayara F Maggioli
- Infectious Bacterial Diseases of Livestock Research Unit, National Animal Disease Center, Ames, IA, USA; Imbio, Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Mitchell V Palmer
- Infectious Bacterial Diseases of Livestock Research Unit, National Animal Disease Center , Ames, IA , USA
| | - Tyler C Thacker
- Infectious Bacterial Diseases of Livestock Research Unit, National Animal Disease Center , Ames, IA , USA
| | | | - Jodi L McGill
- Department of Diagnostic Medicine and Pathology, College of Veterinary Medicine, Kansas State University , Manhattan, KS , USA
| | - Adam O Whelan
- Defense Science and Technology Laboratory, Porton Down , Wiltshire , UK
| | - Michelle H Larsen
- Department of Microbiology and Immunology, Albert Einstein College of Medicine , Bronx, NY , USA
| | - William R Jacobs
- Department of Microbiology and Immunology, Albert Einstein College of Medicine , Bronx, NY , USA
| | - W Ray Waters
- Infectious Bacterial Diseases of Livestock Research Unit, National Animal Disease Center , Ames, IA , USA
| |
Collapse
|
8
|
Cabinian A, Sinsimer D, Tang M, Zumba O, Mehta H, Toma A, Sant’Angelo D, Laouar Y, Laouar A. Transfer of Maternal Immune Cells by Breastfeeding: Maternal Cytotoxic T Lymphocytes Present in Breast Milk Localize in the Peyer's Patches of the Nursed Infant. PLoS One 2016; 11:e0156762. [PMID: 27285085 PMCID: PMC4902239 DOI: 10.1371/journal.pone.0156762] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/19/2016] [Indexed: 12/22/2022] Open
Abstract
Despite our knowledge of the protective role of antibodies passed to infants through breast milk, our understanding of immunity transfer via maternal leukocytes is still limited. To emulate the immunological interface between the mother and her infant while breast-feeding, we used murine pups fostered after birth onto MHC-matched and MHC-mismatched dams. Overall, data revealed that: 1) Survival of breast milk leukocytes in suckling infants is possible, but not significant after the foster-nursing ceases; 2) Most breast milk lymphocytes establish themselves in specific areas of the intestine termed Peyer’s patches (PPs); 3) While most leukocytes in the milk bolus were myeloid cells, the majority of breast milk leukocytes localized to PPs were T lymphocytes, and cytotoxic T cells (CTLs) in particular; 4) These CTLs exhibit high levels of the gut-homing molecules α4β7 and CCR9, but a reduced expression of the systemic homing marker CD62L; 5) Under the same activation conditions, transferred CD8 T cells through breast milk have a superior capacity to produce potent cytolytic and inflammatory mediators when compared to those generated by the breastfed infant. It is therefore possible that maternal CTLs found in breast milk are directed to the PPs to compensate for the immature adaptive immune system of the infant in order to protect it against constant oral infectious risks during the postnatal phase.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Animals, Suckling
- Cells, Cultured
- Chemotaxis, Leukocyte/physiology
- Female
- Immunity, Maternally-Acquired/immunology
- Immunization, Passive/methods
- Lactation/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Milk/cytology
- Milk/immunology
- Mothers
- Peyer's Patches/cytology
- Peyer's Patches/immunology
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/physiology
Collapse
Affiliation(s)
- Allison Cabinian
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Daniel Sinsimer
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - May Tang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Osvaldo Zumba
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Hetali Mehta
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Annmarie Toma
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Derek Sant’Angelo
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Yasmina Laouar
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
- * E-mail: (AL); (YL)
| | - Amale Laouar
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
- * E-mail: (AL); (YL)
| |
Collapse
|
9
|
Gonzalez-Perez G, Hicks AL, Tekieli TM, Radens CM, Williams BL, Lamousé-Smith ESN. Maternal Antibiotic Treatment Impacts Development of the Neonatal Intestinal Microbiome and Antiviral Immunity. THE JOURNAL OF IMMUNOLOGY 2016; 196:3768-79. [PMID: 27036912 DOI: 10.4049/jimmunol.1502322] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/29/2016] [Indexed: 12/20/2022]
Abstract
Microbial colonization of the infant gastrointestinal tract (GIT) begins at birth, is shaped by the maternal microbiota, and is profoundly altered by antibiotic treatment. Antibiotic treatment of mothers during pregnancy influences colonization of the GIT microbiota of their infants. The role of the GIT microbiota in regulating adaptive immune function against systemic viral infections during infancy remains undefined. We used a mouse model of perinatal antibiotic exposure to examine the effect of GIT microbial dysbiosis on infant CD8(+) T cell-mediated antiviral immunity. Maternal antibiotic treatment/treated (MAT) during pregnancy and lactation resulted in profound alterations in the composition of the GIT microbiota in mothers and infants. Streptococcus spp. dominated the GIT microbiota of MAT mothers, whereas Enterococcus faecalis predominated within the MAT infant GIT. MAT infant mice subsequently exhibited increased and accelerated mortality following vaccinia virus infection. Ag-specific IFN-γ-producing CD8(+) T cells were reduced in sublethally infected MAT infant mice. MAT CD8(+) T cells from uninfected infant mice also demonstrated a reduced capacity to sustain IFN-γ production following in vitro activation. We additionally determined that control infant mice became more susceptible to infection if they were born in an animal facility using stricter standards of hygiene. These data indicate that undisturbed colonization and progression of the GIT microbiota during infancy are necessary to promote robust adaptive antiviral immune responses.
Collapse
Affiliation(s)
- Gabriela Gonzalez-Perez
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Columbia University, New York, NY 10032; and
| | - Allison L Hicks
- Center for Infection and Immunity, Columbia University, New York, NY 10032
| | - Tessa M Tekieli
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Columbia University, New York, NY 10032; and
| | - Caleb M Radens
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Columbia University, New York, NY 10032; and
| | - Brent L Williams
- Center for Infection and Immunity, Columbia University, New York, NY 10032
| | - Esi S N Lamousé-Smith
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Columbia University, New York, NY 10032; and
| |
Collapse
|
10
|
Ye C, Choi JG, Abraham S, Shankar P, Manjunath N. Targeting DNA vaccines to myeloid cells using a small peptide. Eur J Immunol 2014; 45:82-8. [PMID: 25270431 DOI: 10.1002/eji.201445010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 08/25/2014] [Accepted: 09/29/2014] [Indexed: 01/02/2023]
Abstract
Targeting DNA vaccines to dendritic cells (DCs) greatly enhances immunity. Although several approaches have been used to target protein Ags to DCs, currently there is no method that targets DNA vaccines directly to DCs. Here, we show that a small peptide derived from the rabies virus glycoprotein fused to protamine residues (RVG-P) can target DNA to myeloid cells, including DCs, which results in enhanced humoral and T-cell responses. DCs targeted with a DNA vaccine encoding the immunodominant vaccinia B8R gene via RVG-P were able to restimulate vaccinia-specific memory T cells in vitro. Importantly, a single i.v. injection of B8R gene bound to RVG-P was able to prime a vaccinia-specific T-cell response that was able to rapidly clear a subsequent vaccinia challenge in mice. Moreover, delivery of DNA in DCs was enough to induce DC maturation and efficient Ag presentation without the need for adjuvants. Finally, immunization of mice with a DNA-vaccine encoding West Nile virus (WNV) prM and E proteins via RVG-P elicited high titers of WNV-neutralizing Abs that protected mice from lethal WNV challenge. Thus, RVG-P provides a reagent to target DNA vaccines to myeloid cells and elicit robust T-cell and humoral immune responses.
Collapse
Affiliation(s)
- Chunting Ye
- Center of Excellence in Infectious Disease Research, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | | | | | | | | |
Collapse
|
11
|
Phenotype of the anti-Rickettsia CD8(+) T cell response suggests cellular correlates of protection for the assessment of novel antigens. Vaccine 2014; 32:4960-7. [PMID: 25043277 DOI: 10.1016/j.vaccine.2014.07.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 06/19/2014] [Accepted: 07/08/2014] [Indexed: 02/03/2023]
Abstract
The obligately intracellular bacteria Rickettsia infect endothelial cells and cause systemic febrile diseases that are potentially lethal. No vaccines are currently available and current knowledge of the effective immune response is limited. Natural and experimental rickettsial infections provide strong and cross-protective cellular immunity if the infected individual survives the acute infection. Although resistance to rickettsial infections is attributed to the induction of antigen-specific T cells, particularly CD8(+) T cells, the identification and validation of correlates of protective cellular immunity against rickettsial infections, an important step toward vaccine validation, remains a gap in this field. Here, we show that after a primary challenge with Rickettsia typhi in the C3H mouse model, the peak of anti-Rickettsia CD8(+) T cell-mediated responses occurs 7 days post-infection (dpi), which coincides with the beginning of rickettsial clearance. At this time point, both effector-type and memory-type CD8(+) T cells are present, suggesting that 7 dpi is a valid time point for the assessment of CD8(+) T cell responses of mice previously immunized with protective antigens. Based on our results, we suggest four correlates of cellular protection for the assessment of protective rickettsial antigens: (1) production of IFN-γ by antigen-experienced CD3(+)CD8(+)CD44(high) cells, (2) production of Granzyme B by CD27(low)CD43(low) antigen-experienced CD8(+) T cells, (3) generation of memory-type CD8(+) T cells [Memory Precursor Effector Cells (MPECs), as well as CD127(high)CD43(low), and CD27(high)CD43(low) CD8(+) T cells], and (4) generation of effector-like memory CD8(+) T cells (CD27(low)CD43(low)). We propose that these correlates could be useful for the general assessment of the quality of the CD8(+) T cell immune response induced by novel antigens with potential use in a vaccine against Rickettsia.
Collapse
|
12
|
Villarreal DO, Wise MC, Walters JN, Reuschel EL, Choi MJ, Obeng-Adjei N, Yan J, Morrow MP, Weiner DB. Alarmin IL-33 acts as an immunoadjuvant to enhance antigen-specific tumor immunity. Cancer Res 2014; 74:1789-800. [PMID: 24448242 DOI: 10.1158/0008-5472.can-13-2729] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Studies of interleukin (IL)-33 reveal a number of pleiotropic properties. Here, we report that IL-33 has immunoadjuvant effects in a human papilloma virus (HPV)-associated model for cancer immunotherapy where cell-mediated immunity is critical for protection. Two biologically active isoforms of IL-33 exist that are full-length or mature, but the ability of either isoform to function as a vaccine adjuvant that influences CD4 T helper 1 or CD8 T-cell immune responses is not defined. We showed that both IL-33 isoforms are capable of enhancing potent antigen-specific effector and memory T-cell immunity in vivo in a DNA vaccine setting. In addition, although both IL-33 isoforms drove robust IFN-γ responses, neither elevated secretion of IL-4 or immunoglobulin E levels. Further, both isoforms augmented vaccine-induced antigen-specific polyfunctional CD4(+) and CD8(+) T-cell responses, with a large proportion of CD8(+) T cells undergoing plurifunctional cytolytic degranulation. Therapeutic studies indicated that vaccination with either IL-33 isoform in conjunction with an HPV DNA vaccine caused rapid and complete regressions in vivo. Moreover, IL-33 could expand the magnitude of antigen-specific CD8(+) T-cell responses and elicit effector-memory CD8(+) T cells. Taken together, our results support the development of these IL-33 isoforms as immunoadjuvants in vaccinations against pathogens, including in the context of antitumor immunotherapy.
Collapse
Affiliation(s)
- Daniel O Villarreal
- Authors' Affiliations: Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia; Inovio Pharmaceuticals, Inc., Blue Bell, Pennsylvania; and Korea Food and Drug Administration, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Aiello FB, Graciotti L, Procopio AD, Keller JR, Durum SK. Stemness of T cells and the hematopoietic stem cells: fate, memory, niche, cytokines. Cytokine Growth Factor Rev 2013; 24:485-501. [PMID: 24231048 PMCID: PMC6390295 DOI: 10.1016/j.cytogfr.2013.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stem cells are able to generate both cells that differentiate and cells that remain undifferentiated but potentially have the same developmental program. The prolonged duration of the protective immune memory for infectious diseases such as polio, small pox, and measles, suggested that memory T cells may have stem cell properties. Understanding the molecular basis for the life-long persistence of memory T cells may be useful to project targeted therapies for immune deficiencies and infectious diseases and to formulate vaccines. In the last decade evidence from different laboratories shows that memory T cells may share self-renewal pathways with bone marrow hematopoietic stem cells. In stem cells the intrinsic self-renewal activity, which depends on gene expression, is known to be modulated by extrinsic signals from the environment that may be tissue specific. These extrinsic signals for stemness of memory T cells include cytokines such as IL-7 and IL-15 and there are other cytokine signals for maintaining the cytokine signature (TH1, TH2, etc.) of memory T cells. Intrinsic and extrinsic pathways that might be common to bone marrow hematopoietic stem cells and memory T lymphocytes are discussed and related to self-renewal functions.
Collapse
Affiliation(s)
- Francesca B Aiello
- Laboratory of Molecular Immunoregulation, Frederick, MD 21702, USA; Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66013 Chieti, Italy.
| | | | | | | | | |
Collapse
|
14
|
CD4 and CD8 T cells participate in the immune memory response against Vaccinia virus after a previous natural infection. RESULTS IN IMMUNOLOGY 2013; 3:104-13. [PMID: 24600565 DOI: 10.1016/j.rinim.2013.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 10/25/2013] [Accepted: 10/28/2013] [Indexed: 11/23/2022]
Abstract
The present study evaluates the immune response of memory CD4(+) and CD8(+) T cells from patients following a natural Vaccinia virus (VACV) infection. A total of 42 individuals were involved in the study being: 22 previously infected individuals (vaccinated or not against smallpox) and 20 non-infected individuals (vaccinated or not). A short-term in vitro stimulation with UV-inactivated VACV of whole blood cells was performed. Our study showed that previously infected individuals have a lower percentage of CD4(+) T cells expressing lymph-node homing receptors (CD4(+)CD62L(+)CCR7(+)) and higher percentage of memory CD4(+) T cells subsets (CD4(+)CD45RO(High)) when compared with non-infected subjects, after in vitro viral stimulation. We also showed that infected individuals presented higher percentages of CD4(+) and CD8(+) memory T lymphocytes expressing IFN-γ when compared to non-infected individuals. We verified that the percentage of CD4(+) and CD8(+) T memory cells expressing TNF-α was higher in infected and non-infected vaccinated subjects when compared with non-infected unvaccinated individual. We also observed that previously infected individuals have higher percentages of CD8(+) T cells expressing lymph-node homing receptors (CCR7(+) and CD62L(+)) and that the memory T cells expressing IFN-γ and TNF-α were at higher percentages in the whole blood cells from infected and non-infected vaccinated individuals, when compared to unvaccinated non-infected subjects. Thus, our findings suggest that CD4(+) and CD8(+) T cells are involved in the immune memory response against Vaccinia virus natural infection.
Collapse
|
15
|
Silaeva YY, Kalinina AA, Vagida MS, Khromykh LM, Deikin AV, Ermolkevich TG, Sadchikova ER, Goldman IL, Kazansky DB. Decrease in pool of T lymphocytes with surface phenotypes of effector and central memory cells under influence of TCR transgenic β-chain expression. BIOCHEMISTRY (MOSCOW) 2013; 78:549-59. [PMID: 23848158 DOI: 10.1134/s0006297913050143] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Peripheral T lymphocytes can be subdivided into naïve and antigen-experienced T cells. The latter, in turn, are represented by effector and central memory cells that are identified by different profiles of activation markers expression, such as CD44 and CD62L in mice. These markers determine different traffic of T lymphocytes in the organism, but hardly reproduce real antigenic experience of a T lymphocyte. Mechanisms of homeostasis maintenance of T lymphocytes with different activation phenotypes remain largely unknown. To investigate impact of T cell receptor (TCR) transgenic chains on formation of T lymphocytes, their peripheral survival and activation surface phenotypes, we have generated the transgenic mouse strain expressing transgenic β-chain of TCR 1D1 (belonging to the Vβ6 family) on the genetic background B10.D2(R101). Intrathymic development of T cells in these transgenic mice is not impaired. The repertoire of peripheral T lymphocytes in these mice contains 70-80% of T cells expressing transgenic β-chain and 20-30% of T cells expressing endogenous β-chains. The ratio of peripheral CD4⁺CD8⁻ and CD4⁻CD8⁺ T lymphocytes remained unchanged in the transgenic animals, but the percent of T lymphocytes with the "naïve" phenotype CD44⁻CD62L⁺ was significantly increased, whereas the levels of effector memory CD44⁺CD62L⁻ and central memory CD44⁺CD62L⁺ T lymphocytes were markedly decreased in both subpopulations. On the contrary, T lymphocytes expressing endogenous β-chains had surface phenotype of activated T cells CD44⁺. Thus, for the first time we have shown that the pool of T lymphocytes with different activation phenotypes depends on the structure of T cell receptors.
Collapse
Affiliation(s)
- Yu Yu Silaeva
- Blokhin Cancer Research Center, Russian Academy of Medical Sciences, Kashirskoe Shosse 24, 115478 Moscow, Russia
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Takai S, Schlom J, Tucker J, Tsang KY, Greiner JW. Inhibition of TGF-β1 signaling promotes central memory T cell differentiation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:2299-307. [PMID: 23904158 PMCID: PMC3889640 DOI: 10.4049/jimmunol.1300472] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
This study affirmed that isolated CD8(+) T cells express mRNA and produce TGF-β following cognate peptide recognition. Blockage of endogenous TGF-β with either a TGF-β-blocking Ab or a small molecule inhibitor of TGF-βRI enhances the generation of CD62L(high)/CD44(high) central memory CD8(+) T cells accompanied with a robust recall response. Interestingly, the augmentation within the central memory T cell pool occurs in lieu of cellular proliferation or activation, but with the expected increase in the ratio of the Eomesoderm/T-bet transcriptional factors. Yet, the signal transduction pathway(s) seems to be noncanonical, independent of SMAD or mammalian target of rapamycin signaling. Enhancement of central memory generation by TGF-β blockade is also confirmed in human PBMCs. The findings underscore the role(s) that autocrine TGF-β plays in T cell homeostasis and, in particular, the balance of effector/memory and central/memory T cells. These results may provide a rationale to targeting TGF-β signaling to enhance Ag-specific CD8(+) T cell memory against a lethal infection or cancer.
Collapse
Affiliation(s)
- Shinji Takai
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
17
|
Effector-like CD8⁺ T cells in the memory population mediate potent protective immunity. Immunity 2013; 38:1250-60. [PMID: 23746652 DOI: 10.1016/j.immuni.2013.05.009] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 05/10/2013] [Accepted: 05/16/2013] [Indexed: 11/21/2022]
Abstract
The CD8⁺ memory T cell population is heterogeneous, and it is unclear which subset(s) optimally mediate the central goal of the immune system-protection against infection. Here we investigate the protective capacities of CD8⁺ T cell subsets present at the memory stage of the immune response. We show that a population of CD8⁺ T cells bearing markers associated with effector cells (KLRG1(hi), CD27(lo), T-bet(hi), Eomes(lo)) persisted to the memory phase and provided optimal control of Listeria monocytogenes and vaccinia virus, despite weak recall proliferative responses. After antigen-specific boosting, this population formed the predominant secondary memory subset and maintained superior pathogen control. The effector-like memory subset displayed a distinct pattern of tissue distribution and localization within the spleen, and their enhanced capacity to eliminate Listeria involved specialized utilization of cytolysis. Together, these data suggest that long-lived effector CD8⁺ T cells are optimal for protective immunity against certain pathogens.
Collapse
|
18
|
Charlton JJ, Chatzidakis I, Tsoukatou D, Boumpas DT, Garinis GA, Mamalaki C. Programmed death-1 shapes memory phenotype CD8 T cell subsets in a cell-intrinsic manner. THE JOURNAL OF IMMUNOLOGY 2013; 190:6104-14. [PMID: 23686498 DOI: 10.4049/jimmunol.1201617] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Memory phenotype T cells, found in unimmunized mice, display phenotypic and functional traits of memory cells and provide essential protection against infections, playing a role in both innate and adaptive immune responses. Mechanisms governing homeostasis of these memory phenotype T cells remain ill-defined. In this study, we reveal a crucial role of the negative costimulator programmed death-1 (PD-1) in regulating developmental fates of memory phenotype cells. Thus, in lymphoid organs and tissues of PD-1 knockout (KO) mice a marked accumulation of functional effector memory (T(EM)) phenotype CD8 T cells was observed. T(EM) phenotype cells from PD-1 KO mice exhibit decreased proliferation but increased survival potential. These cells could produce effector molecules constitutively, in response to phorbol esters or through bystander activation by innate stimuli. Similarly, in lymphopenia-induced proliferating CD8 T cells, whereby normally naive T cells acquire a memory phenotype, skewing toward a T(EM) phenotype was prominent in the absence of PD-1. Acquisition of the T(EM) phenotype was a CD8 T cell-intrinsic phenomenon as demonstrated by mixed bone marrow transfer experiments. Importantly, adoptively transferred PD-1 KO CD8 central memory T (T(CM)) cells converted into the T(EM) phenotype, indicating that PD-1 sets a major checkpoint in the T(CM) to T(EM) phenotype differentiation process. This was reflected by distinct patterns of gene expression of PD-1 KO T(CM) phenotype cells revealed by global transcriptional analysis. Additionally, adoptively transferred PD-1 KO T(EM) phenotype cells converted to a lesser degree to a T(CM) phenotype. Collectively, these data suggest that PD-1 shapes memory phenotype CD8 T cell subsets.
Collapse
Affiliation(s)
- Joanna J Charlton
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, GR-70013 Heraklion, Crete, Greece
| | | | | | | | | | | |
Collapse
|
19
|
Lassen LB, Füchtbauer A, Schmitz A, Sørensen AB, Pedersen FS, Füchtbauer EM. Septin9 is involved in T-cell development and CD8+ T-cell homeostasis. Cell Tissue Res 2013; 352:695-705. [DOI: 10.1007/s00441-013-1618-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 03/11/2013] [Indexed: 12/18/2022]
|
20
|
Hamilton SE, Jameson SC. CD8 T cell memory: it takes all kinds. Front Immunol 2012; 3:353. [PMID: 23230436 PMCID: PMC3515884 DOI: 10.3389/fimmu.2012.00353] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 11/06/2012] [Indexed: 12/12/2022] Open
Abstract
Understanding the mechanisms that regulate the differentiation and maintenance of CD8+ memory T cells is fundamental to the development of effective T cell-based vaccines. Memory cell differentiation is influenced by the cytokines that accompany T cell priming, the history of previous antigen encounters, and the tissue sites into which memory cells migrate. These cues combine to influence the developing CD8+ memory pool, and recent work has revealed the importance of multiple transcription factors, metabolic molecules, and surface receptors in revealing the type of memory cell that is generated. Paired with increasingly meticulous subsetting and sorting of memory populations, we now know the CD8+ memory pool to be phenotypically and functionally heterogeneous in nature. This includes both recirculating and tissue-resident memory populations, and cells with varying degrees of inherent longevity and protective function. These data point to the importance of tailored vaccine design. Here we discuss how the diversity of the memory CD8+ T cell pool challenges the notion that “one size fits all” for pathogen control, and how distinct memory subsets may be suited for distinct aspects of protective immunity.
Collapse
Affiliation(s)
- Sara E Hamilton
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School Minneapolis, MN, USA
| | | |
Collapse
|
21
|
Taylor AP, Makabi-Panzu B, Chen X, Gold DV, Goldenberg DM. Evaluation of a non-viral vaccine in smallpox-vaccinated individuals and immunized HLA-transgenic mice. Hum Immunol 2012; 73:612-9. [PMID: 22504409 DOI: 10.1016/j.humimm.2012.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 02/28/2012] [Accepted: 03/19/2012] [Indexed: 11/18/2022]
Abstract
The current poxvirus vaccine is associated with rare, but serious adverse events. Therefore, we investigated a non-replicating approach to vaccine design. Peptides encoding potential HLA-binding motifs were derived from the orthopoxvirus genes, D8L, A27L, and C12L (the IL-18-binding protein [vIL18BP105]), all of which are preserved among poxviruses that infect humans, and which may be a target of host immunity. The peptides were tested with poxvirus-vaccinated human PBMC and serum for eliciting memory responses, as well as with splenocytes and serum from peptide-immunized, human HLA-DR04 transgenic (HLA tg) mice. vIL18BP105 induced 5-fold proliferation of vaccinated-donor PBMC over non-vaccinated (P<0.001), including IL-2-producing CD8+ cells. Serum IgG recognizing vIL18BP105 was detected (P<0.002 vs non-vaccinated) by ELISA. Viral peptides were conjugated to the HLA-targeting mAb, L243, for immunization of HLA tg mice. Splenocytes from vIL18BP105-L243-immunized mice proliferated upon exposure to vIL18BP105 (P<0.001). Proliferating splenocytes were interferon-γ-producing CD4(+)CD45RA(neg). vIL18BP105-L243-immunized mice generated IgG more rapidly than free-peptide-immunized mice. Peptide-specific antibody was also detected when different L243-peptide conjugates were combined. vIL18BP, by eliciting human memory responses, is a viable antigen for inclusion in a virus-free vaccine. The immunogenicity of peptides was boosted by conjugation to L243, whether administered alone or combined.
Collapse
Affiliation(s)
- Alice P Taylor
- Center for Molecular Medicine and Immunology/Garden State Cancer Center (CMMI/GSCC), 300 The American Road, Morris Plains, NJ 07950, USA.
| | | | | | | | | |
Collapse
|
22
|
Takai S, Sabzevari H, Farsaci B, Schlom J, Greiner JW. Distinct effects of saracatinib on memory CD8+ T cell differentiation. THE JOURNAL OF IMMUNOLOGY 2012; 188:4323-33. [PMID: 22450814 DOI: 10.4049/jimmunol.1101439] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Immunologic memory involving CD8(+) T cells is a hallmark of an adaptive Ag-specific immune response and constitutes a critical component of protective immunity. Designing approaches that enhance long-term T cell memory would, for the most part, fortify vaccines and enhance host protection against infectious diseases and, perhaps, cancer immunotherapy. A better understanding of the cellular programs involved in the Ag-specific T cell response has led to new approaches that target the magnitude and quality of the memory T cell response. In this article, we show that T cells from TCR transgenic mice for the nucleoprotein of influenza virus NP68 exhibit the distinct phases--priming, expansion, contraction, and memory--of an Ag-specific T cell response when exposed in vitro to the cognate peptide. Saracatinib, a specific inhibitor of Src family kinases, administered at low doses during the expansion or contraction phases, increased CD62L(high)/CD44(high) central memory CD8(+) T cells and IFN-γ production but suppressed immunity when added during the priming phase. These effects by saracatinib were not accompanied by the expected decline of Src family kinases but were accompanied by Akt-mammalian target of rapamycin suppression and/or mediated via another pathway. Increased central memory cells by saracatinib were recapitulated in mice using a poxvirus-based influenza vaccine, thus underscoring the importance of dose and timing of the inhibitor in the context of memory T cell differentiation. Finally, vaccine plus saracatinib treatment showed better protection against tumor challenge. The immune-potentiating effects on CD8(+) T cells by a low dose of saracatinib might afford better protection from pathogens or cancer when combined with vaccine.
Collapse
Affiliation(s)
- Shinji Takai
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
23
|
Bot A, Qiu Z, Wong R, Obrocea M, Smith KA. Programmed cell death-1 (PD-1) at the heart of heterologous prime-boost vaccines and regulation of CD8+ T cell immunity. J Transl Med 2010; 8:132. [PMID: 21144062 PMCID: PMC3012026 DOI: 10.1186/1479-5876-8-132] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 12/14/2010] [Indexed: 01/24/2023] Open
Abstract
Developing new vaccination strategies and optimizing current vaccines through heterologous prime-boost carries the promise of integrating the benefits of different yet synergistic vectors. It has been widely thought that the increased immunity afforded by heterologous prime-boost vaccination is mainly due to the minimization of immune responses to the carrier vectors, which allows a progressive build up of immunity against defined epitopes and the subsequent induction of broader immune responses against pathogens. Focusing on CD8+ T cells, we put forward a different yet complementary hypothesis based primarily on the systematic analysis of DNA vaccines as priming agents. This hypothesis relies on the finding that during the initiation of immune response, acquisition of co-inhibitory receptors such as programmed cell death-1 (PD-1) is determined by the pattern of antigen exposure in conjunction with Toll-like receptor (TLR)-dependent stimulation, critically affecting the magnitude and profile of secondary immunity. This hypothesis, based upon the acquisition and co-regulation of pivotal inhibitory receptors by CD8+ T cells, offers a rationale for gene-based immunization as an effective priming strategy and, in addition, outlines a new dimension to immune homeostasis during immune reaction to pathogens. Finally, this model implies that new and optimized immunization approaches for cancer and certain viral infections must induce highly efficacious T cells, refractory to a broad range of immune-inhibiting mechanisms, rather than solely or primarily focusing on the generation of large pools of vaccine-specific lymphocytes.
Collapse
Affiliation(s)
- Adrian Bot
- MannKind Corporation, 28903 North Avenue Paine, Valencia, CA 91355, USA.
| | | | | | | | | |
Collapse
|
24
|
Flutter B, Edwards N, Fallah-Arani F, Henderson S, Chai JG, Sivakumaran S, Ghorashian S, Bennett CL, Freeman GJ, Sykes M, Chakraverty R. Nonhematopoietic antigen blocks memory programming of alloreactive CD8+ T cells and drives their eventual exhaustion in mouse models of bone marrow transplantation. J Clin Invest 2010; 120:3855-68. [PMID: 20978352 DOI: 10.1172/jci41446] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Accepted: 09/02/2010] [Indexed: 12/31/2022] Open
Abstract
Allogeneic blood or BM transplantation (BMT) is the most commonly applied form of adoptive cellular therapy for cancer. In this context, the ability of donor T cells to respond to recipient antigens is coopted to generate graft-versus-tumor (GVT) responses. The major reason for treatment failure is tumor recurrence, which is linked to the eventual loss of functional, host-specific CTLs. In this study, we have explored the role of recipient antigen expression by nonhematopoietic cells in the failure to sustain effective CTL immunity. Using clinically relevant models, we found that nonhematopoietic antigen severely disrupts the formation of donor CD8+ T cell memory at 2 distinct levels that operate in the early and late phases of the response. First, initial and direct encounters between donor CD8+ T cells and nonhematopoietic cells blocked the programming of memory precursors essential for establishing recall immunity. Second, surviving CD8+ T cells became functionally exhausted with heightened expression of the coinhibitory receptor programmed death-1 (PD-1). These 2 factors acted together to induce even more profound failure in long-term immunosurveillance. Crucially, the functions of exhausted CD8+ T cells could be partially restored by late in vivo blockade of the interaction between PD-1 and its ligand, PD-L1, without induction of graft-versus-host disease, suggestive of a potential clinical strategy to prevent or treat relapse following allogeneic BMT.
Collapse
Affiliation(s)
- Barry Flutter
- Transplantation Immunology Group, Department of Haematology, University College London, London, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Szulc L, Boratynska A, Martyniszyn L, Niemialtowski MG. Antigen presenting and effector cell cluster formation in BALB/c mice during mousepox: model studies*. J Appl Microbiol 2010; 109:1817-28. [PMID: 20666870 DOI: 10.1111/j.1365-2672.2010.04813.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIMS The objective of this study was to access APC-effector cell cluster formation in genetically susceptible BALB/c (H-2(d) ) mice infected with highly virulent Moscow strain of ectromelia virus (ECTV-MOS) and estimate of lymphocyte activation based upon expression of CD62L and CD44 molecules. METHODS AND RESULTS APC-effector cell clusters were obtained by enzymatic digestion from draining lymph nodes (DLNs) and spleens of BALB/c mice. We found that APCs infected with ECTV-MOS form unstable clusters with effector cells, and thus may diminish T-cell activation at the early stage of mousepox. Different types of effector cells including T-cell subsets (CD4(+) and CD8(+) ), B cells and polymorphonuclear cells colocalize within individual clusters. Increase in CD19(+) B cells within APC-effector cell clusters during severe clinical mousepox may reflect B-cell activation. CONCLUSIONS Our studies indicated vigorous changes in APC-effector cell cluster formation in genetically susceptible BALB/c mice during mousepox (up to 2 weeks). ECTV-MOS can modulate APC interactions with effector cells and consequently may impair T-cell activation probably owing to unstable cluster formation and/or subsequent weak stimulation by infected APCs at the early stages of mousepox. SIGNIFICANCE AND IMPACT OF THE STUDY This is the first report of APC-effector cell cluster formation in BALB/c mice during mousepox. It gives us a new light on the mutual cell-cell interactions and development of the immune response during ECTV-MOS infection.
Collapse
Affiliation(s)
- L Szulc
- Division of Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Warsaw, Poland
| | | | | | | |
Collapse
|
26
|
Takayama E, Ono T, Carnero E, Umemoto S, Yamaguchi Y, Kanayama A, Oguma T, Takashima Y, Tadakuma T, García-Sastre A, Miyahira Y. Quantitative and qualitative features of heterologous virus-vector-induced antigen-specific CD8+ T cells against Trypanosoma cruzi infection. Int J Parasitol 2010; 40:1549-61. [PMID: 20620143 DOI: 10.1016/j.ijpara.2010.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 05/24/2010] [Accepted: 05/26/2010] [Indexed: 12/27/2022]
Abstract
We studied some aspects of the quantitative and qualitative features of heterologous recombinant (re) virus-vector-induced, antigen-specific CD8(+) T cells against Trypanosoma cruzi. We used three different, highly attenuated re-viruses, i.e., influenza virus, adenovirus and vaccinia virus, which all expressed a single, T. cruzi antigen-derived CD8(+) T-cell epitope. The use of two out of three vectors or the triple virus-vector vaccination regimen not only confirmed that the re-vaccinia virus, which was placed last in order for sequential immunisation, was an effective booster for the CD8(+) T-cell immunity in terms of the number of antigen-specific CD8(+) T cells, but also demonstrated that (i) the majority of cells exhibit the effector memory (T(EM)) phenotype, (ii) robustly secrete IFN-γ, (iii) express higher intensity of the CD122 molecule and (iv) present protective activity against T. cruzi infection. In contrast, placing the re-influenza virus last in sequential immunisation had a detrimental effect on the quantitative and qualitative features of CD8(+) T cells. The triple virus-vector vaccination was more effective at inducing a stronger CD8(+) T-cell immunity than using two re-viruses. The different quantitative and qualitative features of CD8(+) T cells induced by different immunisation regimens support the notion that the refinement of the best choice of multiple virus-vector combinations is indispensable for the induction of a maximum number of CD8(+) T cells of high quality.
Collapse
Affiliation(s)
- Eiji Takayama
- Department of Global Infectious Diseases and Tropical Medicine, National Defense Medical College, Tokorozawa City, Saitama, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Mattoo H, Faulkner M, Kandpal U, Das R, Lewis V, George A, Rath S, Durdik JM, Bal V. Naive CD4 T cells from aged mice show enhanced death upon primary activation. Int Immunol 2009; 21:1277-89. [PMID: 19748905 DOI: 10.1093/intimm/dxp094] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Poor T cell immunity is one of the many defects seen in elderly humans and aged (Ad) mice. We report that naive CD4 T cells from aged mice (ANCD4 cells) showed greater apoptosis upon primary activation than those from young (Yg) mice, with loss of mitochondrial membrane potential, poor activation of Rel family transcription factors and increased DNA damage. Their ability to enhance glycolysis, produce lactate and induce autophagy following activation was also compromised. ANCD4 cells remained susceptible to death beyond first cell division. Activated ANCD4 cells also showed poor transition to a 'central memory' (CM) CD44(high), CD62L(high) phenotype in vitro. This correlated with low proportions of CM cells in Ad mice in vivo. Functionally, too, IFN-gamma responses recalled from T cells of immunized Ad mice, poor to begin with, worsened with time as compared with Yg mice. Thus, ANCD4 cells handle activation-associated stress very poorly due to multiple defects, possibly contributing to poor formation of long-lasting memory.
Collapse
Affiliation(s)
- Hamid Mattoo
- National Institute of Immunology, Aruna Asaf Ali, New Delhi, India
| | | | | | | | | | | | | | | | | |
Collapse
|