Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Virology ; 564: 1-12, 2021 12.
Article in English | MEDLINE | ID: mdl-34560573

ABSTRACT

Many poxviruses produce proteins that are related to epidermal growth factor (EGF). Prior genome sequencing of ectromelia virus revealed a gene predicted to produce a protein with homology to EGF, which we refer to as ectromelia growth factor (ECGF). ECGF is truncated relative to vaccinia growth factor (VGF) because the former lacks a transmembrane domain. We show these proteins can experience differential N-linked glycosylation. Despite these differences, both proteins maintain the six conserved cysteine residues important for the function of EGF. Since ECGF has not been characterized, our objective was to determine if it can act as a growth factor. We added ECGF to cultured cells and found that the EGF receptor becomes activated, S-phase was induced, doubling time decreased, and in vitro wound healing occurred faster compared to untreated cells. In summary, we demonstrate that ECGF can act as a mitogen in a similar manner as VGF.


Subject(s)
Ectromelia virus/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Mitogens/metabolism , Viral Proteins/metabolism , Animals , Cell Division , Cell Line , Cell Movement , Epidermal Growth Factor/chemistry , Glycosylation , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Protein Binding , S Phase , Signal Transduction , Vaccinia virus/metabolism , Viral Proteins/chemistry , Wound Healing
2.
PLoS Pathog ; 16(8): e1008685, 2020 08.
Article in English | MEDLINE | ID: mdl-32745153

ABSTRACT

Smallpox and monkeypox pose severe threats to human health. Other orthopoxviruses are comparably virulent in their natural hosts, including ectromelia, the cause of mousepox. Disease severity is linked to an array of immunomodulatory proteins including the B22 family, which has homologs in all pathogenic orthopoxviruses but not attenuated vaccine strains. We demonstrate that the ectromelia B22 member, C15, is necessary and sufficient for selective inhibition of CD4+ but not CD8+ T cell activation by immunogenic peptide and superantigen. Inhibition is achieved not by down-regulation of surface MHC- II or co-stimulatory protein surface expression but rather by interference with antigen presentation. The appreciable outcome is interference with CD4+ T cell synapse formation as determined by imaging studies and lipid raft disruption. Consequently, CD4+ T cell activating stimulus shifts to uninfected antigen-presenting cells that have received antigen from infected cells. This work provides insight into the immunomodulatory strategies of orthopoxviruses by elucidating a mechanism for specific targeting of CD4+ T cell activation, reflecting the importance of this cell type in control of the virus.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Histocompatibility Antigens Class II/immunology , Viral Proteins/immunology , Animals , Ectromelia, Infectious/metabolism , Ectromelia, Infectious/virology , Female , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Viral Proteins/metabolism , Virulence
3.
Cancer Immunol Res ; 6(5): 509-516, 2018 05.
Article in English | MEDLINE | ID: mdl-29615399

ABSTRACT

One major hurdle to the success of adoptive T-cell therapy is the identification of antigens that permit effective targeting of tumors in the absence of toxicities to essential organs. Previous work has demonstrated that T cells engineered to express chimeric antigen receptors (CAR-T cells) targeting the murine homolog of the colorectal cancer antigen GUCY2C treat established colorectal cancer metastases, without toxicity to the normal GUCY2C-expressing intestinal epithelium, reflecting structural compartmentalization of endogenous GUCY2C to apical membranes comprising the intestinal lumen. Here, we examined the utility of a human-specific, GUCY2C-directed single-chain variable fragment as the basis for a CAR construct targeting human GUCY2C-expressing metastases. Human GUCY2C-targeted murine CAR-T cells promoted antigen-dependent T-cell activation quantified by activation marker upregulation, cytokine production, and killing of GUCY2C-expressing, but not GUCY2C-deficient, cancer cells in vitro GUCY2C CAR-T cells provided long-term protection against lung metastases of murine colorectal cancer cells engineered to express human GUCY2C in a syngeneic mouse model. GUCY2C murine CAR-T cells recognized and killed human colorectal cancer cells endogenously expressing GUCY2C, providing durable survival in a human xenograft model in immunodeficient mice. Thus, we have identified a human GUCY2C-specific CAR-T cell therapy approach that may be developed for the treatment of GUCY2C-expressing metastatic colorectal cancer. Cancer Immunol Res; 6(5); 509-16. ©2018 AACR.


Subject(s)
Colorectal Neoplasms/therapy , Cytotoxicity, Immunologic , Immunotherapy, Adoptive/methods , Lung Neoplasms/prevention & control , Receptors, Chimeric Antigen/metabolism , Receptors, Enterotoxin , T-Lymphocytes/transplantation , Animals , Cells, Cultured , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Metastasis , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Enterotoxin/genetics , Receptors, Enterotoxin/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
4.
Virology ; 518: 335-348, 2018 05.
Article in English | MEDLINE | ID: mdl-29602068

ABSTRACT

All known orthopoxviruses, including ectromelia virus (ECTV), contain a gene in the E3L family. The protein product of this gene, E3, is a double-stranded RNA-binding protein. It can impact host range and is used by orthopoxviruses to combat cellular defense pathways, such as PKR and RNase L. In this work, we constructed an ECTV mutant with a targeted disruption of the E3L open reading frame (ECTVΔE3L). Infection with this virus resulted in an abortive replication cycle in all cell lines tested. We detected limited transcription of late genes but no significant translation of these mRNAs. Notably, the replication defects of ECTVΔE3L were rescued in human and mouse cells lacking PKR. ECTVΔE3L was nonpathogenic in BALB/c mice, a strain susceptible to lethal mousepox disease. However, infection with ECTVΔE3L induced protective immunity upon subsequent challenge with wild-type virus. In summary, E3L is an essential gene for ECTV.


Subject(s)
Ectromelia virus/immunology , Ectromelia virus/physiology , Ectromelia, Infectious/prevention & control , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Vaccines/immunology , Virus Replication , Animals , Cell Line , Ectromelia virus/genetics , Ectromelia virus/pathogenicity , Gene Knockout Techniques , Humans , Mice, Inbred BALB C , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
5.
Virology ; 509: 98-111, 2017 09.
Article in English | MEDLINE | ID: mdl-28628829

ABSTRACT

Most orthopoxviruses, including vaccinia virus (VACV), contain genes in the E3L and K3L families. The protein products of these genes have been shown to combat PKR, a host defense pathway. Interestingly, ectromelia virus (ECTV) contains an E3L ortholog but does not possess an intact K3L gene. Here, we gained insight into how ECTV can still efficiently evade PKR despite lacking K3L. Relative to VACV, we found that ECTV-infected BS-C-1 cells accumulated considerably less double-stranded (ds) RNA, which was due to lower mRNA levels and less transcriptional read-through of some genes by ECTV. The abundance of dsRNA in VACV-infected cells, detected using a monoclonal antibody, was able to activate the RNase L pathway at late time points post-infection. Historically, the study of transcription by orthopoxviruses has largely focused on VACV as a model. Our data suggest that there could be more to learn by studying other members of this genus.


Subject(s)
Ectromelia virus/physiology , RNA, Double-Stranded/metabolism , Vaccinia virus/physiology , Virus Replication , Animals , Cell Line , Chlorocebus aethiops , Epithelial Cells/virology , Immune Evasion , RNA, Double-Stranded/immunology , RNA, Messenger/metabolism , RNA, Viral/metabolism , Transcription, Genetic , eIF-2 Kinase/antagonists & inhibitors
6.
PLoS One ; 10(3): e0119189, 2015.
Article in English | MEDLINE | ID: mdl-25734776

ABSTRACT

As a group, poxviruses have been shown to infect a wide variety of animal species. However, there is individual variability in the range of species able to be productively infected. In this study, we observed that ectromelia virus (ECTV) does not replicate efficiently in cultured rabbit RK13 cells. Conversely, vaccinia virus (VACV) replicates well in these cells. Upon infection of RK13 cells, the replication cycle of ECTV is abortive in nature, resulting in a greatly reduced ability to spread among cells in culture. We observed ample levels of early gene expression but reduced detection of virus factories and severely blunted production of enveloped virus at the cell surface. This work focused on two important host range genes, named E3L and K3L, in VACV. Both VACV and ECTV express a functional protein product from the E3L gene, but only VACV contains an intact K3L gene. To better understand the discrepancy in replication capacity of these viruses, we examined the ability of ECTV to replicate in wild-type RK13 cells compared to cells that constitutively express E3 and K3 from VACV. The role these proteins play in the ability of VACV to replicate in RK13 cells was also analyzed to determine their individual contribution to viral replication and PKR activation. Since E3L and K3L are two relevant host range genes, we hypothesized that expression of one or both of them may have a positive impact on the ability of ECTV to replicate in RK13 cells. Using various methods to assess virus growth, we did not detect any significant differences with respect to the replication of ECTV between wild-type RK13 compared to versions of this cell line that stably expressed VACV E3 alone or in combination with K3. Therefore, there remain unanswered questions related to the factors that limit the host range of ECTV.


Subject(s)
Ectromelia virus/genetics , Gene Expression Regulation, Viral , RNA-Binding Proteins/genetics , Vaccinia virus/genetics , Viral Proteins/genetics , Animals , Cell Line , Ectopic Gene Expression , Epithelial Cells/pathology , Epithelial Cells/virology , Host Specificity , Kidney/pathology , Kidney/virology , Mice , RNA-Binding Proteins/metabolism , Rabbits , Vaccinia virus/pathogenicity , Viral Proteins/metabolism , Virus Replication
7.
J Virol ; 88(17): 10078-91, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24965457

ABSTRACT

UNLABELLED: The factors that determine CD4+ T cell (TCD4+) specificities, functional capacity, and memory persistence in response to complex pathogens remain unclear. We explored these parameters in the C57BL/6 mouse through comparison of two highly related (>92% homology) poxviruses: ectromelia virus (ECTV), a natural mouse pathogen, and vaccinia virus (VACV), a heterologous virus that nevertheless elicits potent immune responses. In addition to elucidating several previously unidentified major histocompatibility complex class II (MHC-II)-restricted epitopes, we observed many qualitative and quantitative differences between the TCD4+ repertoires, including responses not elicited by VACV despite complete sequence conservation. In addition, we observed functional heterogeneity between ECTV- and VACV-specific TCD4+ at both a global and individual epitope level, particularly greater expression of the cytolytic marker CD107a from TCD4+ following ECTV infection. Most striking were differences during the late memory phase where, in contrast to ECTV, VACV infection failed to elicit measurable epitope-specific TCD4+ as determined by intracellular cytokine staining. These findings illustrate the strong influence of epitope-extrinsic factors on TCD4+ responses and memory. IMPORTANCE: Much of our understanding concerning host-pathogen relationships in the context of poxvirus infections stems from studies of VACV in mice. However, VACV is not a natural mouse pathogen, and therefore, the relevance of results obtained using this model may be limited. Here, we explored the MHC class II-restricted TCD4+ repertoire induced by mousepox (ECTV) infection and the functional profile of the responding epitope-specific TCD4+, comparing these results to those induced by VACV infection under matched conditions. Despite a high degree of homology between the two viruses, we observed distinct specificity and functional profiles of TCD4+ responses at both acute and memory time points, with VACV-specific TCD4+ memory being notably compromised. These data offer insight into the impact of epitope-extrinsic factors on the resulting TCD4+ responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Ectromelia virus/immunology , Poxviridae Infections/immunology , Poxviridae Infections/virology , Vaccinia virus/immunology , Animals , Cytokines/biosynthesis , Disease Models, Animal , Epitopes/immunology , Female , Immunologic Memory , Mice, Inbred C57BL , T-Lymphocyte Subsets/immunology
8.
J Virol ; 88(16): 9472-5, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24899206

ABSTRACT

We assessed several routes of immunization with vaccinia virus (VACV) in protecting mice against ectromelia virus (ECTV). By a wide margin, skin scarification provided the greatest protection. Humoral immunity and resident-memory T cells notwithstanding, several approaches revealed that circulating, memory CD8(+) T cells primed via scarification were functionally superior and conferred enhanced virus control. Immunization via the epithelial route warrants further investigation, as it may also provide enhanced defense against other infectious agents.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Epithelium/immunology , Vaccinia virus/immunology , Animals , Immunity, Humoral/immunology , Immunization/methods , Immunologic Memory/immunology , Mice , Mice, Inbred BALB C , Vaccination/methods
9.
Proc Natl Acad Sci U S A ; 109(25): 9983-8, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22665800

ABSTRACT

CD4(+) T cells are generally regarded as helpers and regulators of the immune response. Although cytolytic CD4(+) T cells have been described, whether those generated during the course of a viral infection play a role in virus control remains unknown. Here we show that during acute infection with ectromelia virus, the mouse homolog of the human virus of smallpox, large numbers of CD4(+) T cells in the draining lymph node and liver of resistant mice have a cytotoxic phenotype. We also show that these cells kill targets in vivo in a perforin-dependent manner and that mice with specific deficiency of perforin in CD4(+) T cells have impaired virus control. Thus, perforin-dependent CD4(+) T-cell killing of infected cells is an important mechanism of antiviral defense.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Ectromelia virus/immunology , Perforin/immunology , Animals , Major Histocompatibility Complex/immunology , Mice
10.
J Virol ; 86(13): 7298-309, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22532670

ABSTRACT

Vaccinia virus (VACV) stimulates long-term immunity against highly pathogenic orthopoxvirus infection of humans (smallpox) and mice (mousepox [ectromelia virus {ECTV}]) despite the lack of a natural host-pathogen relationship with either of these species. Previous research revealed that VACV is able to induce polyfunctional CD8(+) T-cell responses after immunization of humans. However, the degree to which the functional profile of T cells induced by VACV is similar to that generated during natural poxvirus infection remains unknown. In this study, we monitored virus-specific T-cell responses following the dermal infection of C57BL/6 mice with ECTV or VACV. Using polychromatic flow cytometry, we measured levels of degranulation, cytokine expression (gamma interferon [IFN-γ], tumor necrosis factor alpha [TNF-α], and interleukin-2 [IL-2]), and the cytolytic mediator granzyme B. We observed that the functional capacities of T cells induced by VACV and ECTV were of a similar quality in spite of the markedly different replication abilities and pathogenic outcomes of these viruses. In general, a significant fraction (≥50%) of all T-cell responses were positive for at least three functions both during acute infection and into the memory phase. In vivo killing assays revealed that CD8(+) T cells specific for both viruses were equally cytolytic (∼80% target cell lysis after 4 h), consistent with the similar levels of granzyme B and degranulation detected among these cells. Collectively, these data provide a mechanism to explain the ability of VACV to induce protective T-cell responses against pathogenic poxviruses in their natural hosts and provide further support for the use of VACV as a vaccine platform able to induce polyfunctional T cells.


Subject(s)
Ectromelia virus/immunology , T-Lymphocytes/immunology , Vaccinia virus/immunology , Animals , Cell Degranulation , Cytokines/biosynthesis , Cytotoxicity Tests, Immunologic , Disease Models, Animal , Ectromelia virus/pathogenicity , Ectromelia virus/physiology , Ectromelia, Infectious/immunology , Female , Flow Cytometry , Granzymes/biosynthesis , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Vaccinia/immunology , Vaccinia virus/pathogenicity , Vaccinia virus/physiology
11.
Curr Opin HIV AIDS ; 6(3): 169-73, 2011 May.
Article in English | MEDLINE | ID: mdl-21399496

ABSTRACT

PURPOSE OF REVIEW: Over the past 2 years, a clearer picture has emerged regarding the properties of HIV-specific CD8+ T cells associated with immunologic control of HIV replication. These properties represent a potential mechanism by which rare patients might control HIV replication in the absence of antiretroviral therapy. This review addresses the background and recent findings that have lead to our current understanding of these mechanism(s). RECENT FINDINGS: Patients with immunologic control of HIV are not distinguished by targeted specificities, or greater numbers or breadth of their HIV-specific CD8+ T-cell response. For this reason, recent work has focused greater attention on qualitative features of this response. The qualitative features most closely associated with immunologic control of HIV are related to the granule-exocytosis-mediated elimination of HIV-infected CD4 T cells. The ability of HIV-specific CD8+ T cells to increase their contents of proteins known to mediate cytotoxicity, such as granzyme B and perforin, appears to be a critical means by which HIV-specific cytotoxic capacity is regulated. SUMMARY: Investigation from multiple groups has now focused upon HIV-specific CD8+ T-cell granule-exocytosis-mediated cytotoxicity as a correlate of immunologic control of HIV. In the near future, a more detailed understanding of the qualities associated with immunologic control may provide critical insights regarding the necessary features of a response that should be stimulated by immunotherapies or T-cell-based vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV Long-Term Survivors , HIV-1/immunology , T-Lymphocytes, Cytotoxic/immunology , Humans , Viremia/immunology , Viremia/prevention & control
12.
Blood ; 117(14): 3799-808, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21289310

ABSTRACT

Recent data suggest that CD8+ T-cell effector activity is an important component in the control of HIV replication in elite controllers (ECs). One critical element of CD8+ T-cell effector function and differentiation is the T-box transcription factor T-bet. In the present study, we assessed T-bet expression, together with the effector proteins perforin, granzyme A (Grz A), granzyme B (Grz B), and granulysin, in HIV-specific CD8+ T cells from ECs (n = 20), chronically infected progressors (CPs; n = 18), and highly active antiretroviral therapy (HAART)-suppressed individuals (n = 19). Compared with the other cohort groups, HIV-specific CD8+ T cells among ECs demonstrated a superior ability to express perforin and Grz B, but with no detectable difference in the levels of Grz A or granulysin. We also observed higher levels of T-bet in HIV-specific CD8+ T cells from ECs, with an ensuing positive correlation between T-bet and levels of both perforin and Grz B. Moreover, HIV-specific CD8+ T cells in ECs up-regulated T-bet to a greater extent than CPs after in vitro expansion, with concomitant up-regulation of perforin and Grz B. These results suggest that T-bet may play an important role in driving effector function, and its modulation may lead to enhanced effector activity against HIV.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , HIV Infections/immunology , HIV/immunology , T-Box Domain Proteins/metabolism , Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active , CD8-Positive T-Lymphocytes/pathology , Cohort Studies , Cross-Sectional Studies , Disease Progression , Granzymes/metabolism , HIV Infections/drug therapy , HIV Infections/metabolism , HIV Infections/pathology , Humans , Immune Tolerance/drug effects , Immune Tolerance/immunology , Immune Tolerance/physiology , Perforin/metabolism , T-Box Domain Proteins/physiology , T-Cell Antigen Receptor Specificity , Treatment Outcome
13.
PLoS Pathog ; 6(5): e1000917, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20523897

ABSTRACT

Many immune correlates of CD8(+) T-cell-mediated control of HIV replication, including polyfunctionality, proliferative ability, and inhibitory receptor expression, have been discovered. However, no functional correlates using ex vivo cells have been identified with the known ability to cause the direct elimination of HIV-infected cells. We have recently discovered the ability of human CD8(+) T-cells to rapidly upregulate perforin--an essential molecule for cell-mediated cytotoxicity--following antigen-specific stimulation. Here, we examined perforin expression capability in a large cross-sectional cohort of chronically HIV-infected individuals with varying levels of viral load: elite controllers (n = 35), viremic controllers (n = 29), chronic progressors (n = 27), and viremic nonprogressors (n = 6). Using polychromatic flow cytometry and standard intracellular cytokine staining assays, we measured perforin upregulation, cytokine production, and degranulation following stimulation with overlapping peptide pools encompassing all proteins of HIV. We observed that HIV-specific CD8(+) T-cells from elite controllers consistently display an enhanced ability to express perforin directly ex vivo compared to all other groups. This ability is not restricted to protective HLA-B haplotypes, does not require proliferation or the addition of exogenous factors, is not restored by HAART, and primarily originates from effector CD8(+) T-cells with otherwise limited functional capability. Notably, we found an inverse relationship between HIV-specific perforin expression and viral load. Thus, the capability of HIV-specific CD8(+) T-cells to rapidly express perforin defines a novel correlate of control in HIV infection.


Subject(s)
CD8-Positive T-Lymphocytes , HIV Infections/immunology , HIV Infections/metabolism , Pore Forming Cytotoxic Proteins/metabolism , AIDS Vaccines/immunology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Alleles , Antiretroviral Therapy, Highly Active , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cohort Studies , Cross-Sectional Studies , Cytokines/metabolism , Disease Progression , Flow Cytometry , HIV Infections/drug therapy , HLA-B Antigens/genetics , Haplotypes , Humans , Perforin , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/virology , Viral Load/immunology , Virus Replication/immunology
14.
PLoS Pathog ; 6(3): e1000798, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20221423

ABSTRACT

The prevailing paradigm of T lymphocyte control of viral replication is that the protective capacity of virus-specific CD8(+) T cells is directly proportional to the number of functions they can perform, with IL-2 production capacity considered critical. Having recently defined rapid perforin upregulation as a novel effector function of antigen-specific CD8(+) T cells, here we sought to determine whether new perforin production is a component of polyfunctional CD8(+) T cell responses that contributes to the control of several human viral infections: cytomegalovirus (CMV), Epstein-Barr virus (EBV), influenza (flu), and adenovirus (Ad). We stimulated normal human donor PBMC with synthetic peptides whose amino acid sequences correspond to defined CTL epitopes in the aforementioned viruses, and then used polychromatic flow cytometry to measure the functional capacity and the phenotype of the responding CD8(+) T cells. While EBV and flu-specific CD8(+) T cells rarely upregulate perforin, CMV-specific cells often do and Ad stimulates an exceptionally strong perforin response. The differential propensity of CD8(+) T cells to produce either IL-2 or perforin is in part related to levels of CD28 and the transcription factor T-bet, as CD8(+) T cells that rapidly upregulate perforin harbor high levels of T-bet and those producing IL-2 express high amounts of CD28. Thus, "polyfunctional" profiling of antigen-specific CD8(+) T cells must not be limited to simply the number of functions the cell can perform, or one particular memory phenotype, but should actually define which combinations of memory markers and functions are relevant in each pathogenic context.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Interleukin-2/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Virus Diseases/immunology , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cytomegalovirus Infections/immunology , Epstein-Barr Virus Infections/immunology , Humans , Immunologic Memory/immunology , Immunophenotyping , Influenza, Human/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-2/immunology , Lymphocyte Activation/immunology , Perforin , Pore Forming Cytotoxic Proteins/immunology , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , Up-Regulation/immunology
15.
J Immunol ; 182(9): 5560-9, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380804

ABSTRACT

CTL are endowed with the ability to eliminate pathogens through perforin-mediated cytotoxic activity. The mechanism for perforin-mediated Ag-specific killing has been solely attributed to cytotoxic granule exocytosis from activated CD8(+) T cells. In this study, we redefine this mechanism, demonstrating that virus-specific CD8(+) T cells rapidly up-regulate perforin in response to stimulation temporally with IFN-gamma and CD107a expression. Following Ag-specific activation, newly synthesized perforin rapidly appears at the immunological synapse, both in association with and independent of cytotoxic granules, where it functions to promote cytotoxicity. Our work suggests a novel mechanism of CTL cytotoxicity and identifies a novel correlate of CD8(+) T cell-mediated immunity.


Subject(s)
Cytoplasmic Granules/immunology , Cytotoxicity Tests, Immunologic , Epitopes, T-Lymphocyte/immunology , Immunological Synapses/immunology , Perforin/biosynthesis , Perforin/physiology , T-Lymphocytes, Cytotoxic/immunology , Up-Regulation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Degranulation/immunology , Cell Line, Transformed , Cytoplasmic Granules/metabolism , Cytotoxicity Tests, Immunologic/methods , Humans , Lymphocyte Activation/immunology , Perforin/metabolism , Protein Transport/immunology , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/virology
16.
Cytometry A ; 73(11): 1050-7, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18615597

ABSTRACT

Perforin and granzymes work synergistically to induce apoptosis in target cells recognized by cytotoxic T lymphocytes. While perforin is readily detectable by flow cytometry in resting CD8 T cells, upregulation of perforin in activated cells is thought to require proliferation. However, perforin undergoes numerous conformational changes during its maturation, which may affect the ability of conventional antibodies to recognize newly synthesized perforin. Polychromatic flow cytometry was used to detect perforin and cytokine production following stimulation of ex vivo human CD8 T cells. Two different anti-perforin antibodies, clones B-D48 and deltaG9, were used to discriminate various forms of perforin after cellular activation. We provide evidence for the rapid upregulation of perforin protein, which may contribute to the ability of CD8 T cells to kill multiple targets over time. The deltaG9 clone recognizes the granule-associated conformation of perforin, while the B-D48 clone is able to detect perforin in multiple forms. Finally, we show there is variability in the ability of CD8 T cells to upregulate perforin. Human CD8 T cells are capable of new perforin production immediately following activation. This work defines a novel flow cytometric procedure that can be used to more completely assess the cytotoxic capacity of human CD8 T cells.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Flow Cytometry/methods , Perforin/genetics , Up-Regulation/genetics , Clone Cells , Granzymes/genetics , Granzymes/metabolism , Humans , Kinetics , Lymphocyte Activation , Perforin/chemistry , Protein Conformation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Staining and Labeling
SELECTION OF CITATIONS
SEARCH DETAIL
...