Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Infect Dis ; 215(3): 351-358, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27932620

ABSTRACT

Sanofi Pasteur has developed a chimeric yellow fever-dengue, live-attenuated, tetravalent dengue vaccine (CYD-TDV) that is currently approved for use in several countries. In clinical trials, CYD-TDV was efficacious at reducing laboratory-confirmed cases of dengue disease. Efficacy varied by dengue virus (DENV) serotype and prevaccination dengue immune status. We compared the properties of antibodies in naive and DENV-exposed individuals who received CYD-TDV. We depleted specific populations of DENV-reactive antibodies from immune serum samples to estimate the contribution of serotype-cross-reactive and type-specific antibodies to neutralization. Subjects with no preexisting immunity to DENV developed neutralizing antibodies to all 4 serotypes of DENV. Further analysis demonstrated that DENV4 was mainly neutralized by type-specific antibodies whereas DENV1, DENV2, and DENV3 were mainly neutralized by serotype cross-reactive antibodies. When subjects with preexisting immunity to DENV were vaccinated, they developed higher levels of neutralizing antibodies than naive subjects who were vaccinated. In preimmune subjects, CYD-TDV boosted cross-reactive neutralizing antibodies while maintaining type-specific neutralizing antibodies acquired before vaccination. Our results demonstrate that the quality of neutralizing antibodies induced by CYD-TDV varies depending on DENV serotype and previous immune status. We discuss the implications of these results for understanding vaccine efficacy.


Subject(s)
Antibodies, Viral/blood , Dengue Vaccines/immunology , Dengue/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line , Dengue/prevention & control , Flavivirus/immunology , Humans , Immunogenicity, Vaccine , Vaccines, Attenuated/immunology
2.
J Clin Invest ; 131(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34003796

ABSTRACT

The 4 serotypes of dengue virus (DENV1-4) are mosquito-borne flaviviruses that infect humans. Live attenuated tetravalent DENV vaccines are at different phases of clinical testing. DENV vaccine developers have relied on neutralizing antibodies (NAbs) as a correlate of protection. A leading tetravalent vaccine (Dengvaxia) stimulated NAbs to the 4 DENV serotypes, yet overall vaccine efficacy was low in children who were DENV seronegative at baseline before vaccination. We compared the properties of (a) NAbs induced by WT DENV1 or DENV3 infections, which are strongly correlated with protection from repeat infections, and (b) NAbs induced by Dengvaxia in individuals who subsequently experienced DENV1 or DENV3 breakthrough infections. WT infections induced NAbs that recognized epitopes unique (type specific) to each serotype, whereas the vaccine stimulated qualitatively different NAbs that recognized epitopes conserved (crossreactive) between serotypes. Our results indicate that, among children who were DENV-seronegative at baseline, unbalanced replication of the DENV type 4 vaccine component in the tetravalent vaccine stimulates Abs capable of crossneutralizing DENV1 and DENV3 in vitro, but not protecting in vivo. In DENV-seronegative individuals who are vaccinated, we propose that type-specific NAbs are a better correlate of protection than total levels of NAbs.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Dengue Vaccines/adverse effects , Dengue Vaccines/immunology , Dengue/immunology , Dengue/prevention & control , Adolescent , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Child , Child, Preschool , Cross Reactions , Dengue/virology , Dengue Virus/classification , Dengue Virus/immunology , Double-Blind Method , Female , Humans , Male , Serogroup , Treatment Failure , Vaccination
3.
J Virol ; 82(8): 3853-63, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18256152

ABSTRACT

Toll-like receptors (TLRs) are known predominantly for their role in activating the innate immune response. Recently, TLR signaling via MyD88 has been reported to play an important function in development of a B-cell response. Since B cells are a major latency reservoir for murine gammaherpesvirus 68 (MHV68), we investigated the role of TLR signaling in the establishment and maintenance of MHV68 latency in vivo. Mice deficient in MyD88 (MyD88(-/-)) or TLR3 (TLR3(-/-)) were infected with MHV68. Analysis of splenocytes recovered at day 16 postinfection from MyD88(-/-) mice compared to those from wild-type control mice revealed a lower frequency of (i) activated B cells, (ii) germinal-center B cells, and (iii) class-switched B cells. Accompanying this substantial defect in the B-cell response was an approximately 10-fold decrease in the establishment of splenic latency. In contrast, no defect in viral latency was observed in TLR3(-/-) mice. Analysis of MHV68-specific antibody responses also demonstrated a substantial decrease in the kinetics of the response in MyD88(-/-) mice. Analysis of wild-type x MyD88(-/-) mixed-bone-marrow chimeric mice demonstrated that there is a selective failure of MyD88(-/-) B cells to participate in germinal-center reactions as well as to become activated and undergo class switching. In addition, while MHV68 established latency efficiently in the MyD88-sufficient B cells, there was again a ca. 10-fold reduction in the frequency of MyD88(-/-) B cells harboring latent MHV68. This phenotype indicates that MyD88 is important for the establishment of MHV68 latency and is directly related to the role of MyD88 in the generation of a B-cell response. Furthermore, the generation of a B-cell response to MHV68 was intrinsic to B cells and was independent of the interleukin-1 receptor, a cytokine receptor that also signals through MyD88. These data provide evidence for a unique role for MyD88 in the establishment of MHV68 latency.


Subject(s)
Herpesviridae Infections/virology , Myeloid Differentiation Factor 88/immunology , Rhadinovirus/immunology , Rhadinovirus/physiology , Tumor Virus Infections/virology , Virus Latency , Animals , Antibodies, Viral/blood , B-Lymphocytes/immunology , B-Lymphocytes/virology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Herpesviridae Infections/immunology , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Spleen/immunology , Spleen/virology , Time Factors , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 3/immunology , Tumor Virus Infections/immunology , Viral Plaque Assay , Virus Activation/immunology
4.
PLoS Pathog ; 3(1): e11, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17257062

ABSTRACT

A critical determinant in chronic gammaherpesvirus infections is the ability of these viruses to establish latency in a lymphocyte reservoir. The nuclear factor (NF)-kappaB family of transcription factors represent key players in B-cell biology and are targeted by gammaherpesviruses to promote host cell survival, proliferation, and transformation. However, the role of NF-kappaB signaling in the establishment of latency in vivo has not been addressed. Here we report the generation and in vivo characterization of a recombinant murine gammaherpesvirus 68 (gammaHV68) that expresses a constitutively active form of the NF-kappaB inhibitor, IkappaBalphaM. Inhibition of NF-kappaB signaling upon infection with gammaHV68-IkappaBalphaM did not affect lytic replication in cell culture or in the lung following intranasal inoculation. However, there was a substantial decrease in the frequency of latently infected lymphocytes in the lung (90% reduction) and spleens (97% reduction) 16 d post intranasal inoculation. Importantly, the defect in establishment of latency in lung B cells could not be overcome by increasing the dose of virus 100-fold. The observed decrease in establishment of viral latency correlated with a loss of activated, CD69(hi) B cells in both the lungs and spleen at day 16 postinfection, which was not apparent by 6 wk postinfection. Constitutive expression of Bcl-2 in B cells did not rescue the defect in the establishment of latency observed with gammaHV68-IkappaBalphaM, indicating that NF-kappaB-mediated functions apart from Bcl-2-mediated B-cell survival are critical for the efficient establishment of gammaherpesvirus latency in vivo. In contrast to the results obtained following intranasal inoculation, infection of mice with gammaHV68-IkappaBalphaM by the intraperitoneal route had only a modest impact on splenic latency, suggesting that route of inoculation may alter requirements for establishment of virus latency in B cells. Finally, analyses of the pathogenesis of gammaHV68-IkappaBalphaM provides evidence that NF-kappaB signaling plays an important role during multiple stages of gammaHV68 infection in vivo and, as such, represents a key host regulatory pathway that is likely manipulated by the virus to establish latency in B cells.


Subject(s)
NF-kappa B/antagonists & inhibitors , Rhadinovirus/physiology , Virus Latency , Animals , Antigens, CD19/analysis , B-Lymphocytes/immunology , B-Lymphocytes/virology , Base Sequence , Female , I-kappa B Proteins/physiology , Lung/virology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Molecular Sequence Data , NF-KappaB Inhibitor alpha , NF-kappa B/physiology , NIH 3T3 Cells , Proto-Oncogene Proteins c-bcl-2/physiology , Signal Transduction , Spleen/virology , Viral Load , Virus Replication
5.
Curr Opin Immunol ; 14(4): 509-16, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12088687

ABSTRACT

An array of inhibitory and activating receptors initially identified on NK cells are also expressed by conventional CD8+ alphabeta T cells. New evidence strongly implicates these 'NK cell receptors' in modulating NK cell and virus-specific CD8+ T cell responses against a variety of viral infections. Precise regulation of NK cell and T cell responses by these receptors optimizes antiviral immunity while preventing immunological bystander pathology and autoimmunity.


Subject(s)
Killer Cells, Natural/immunology , Receptors, Immunologic/physiology , T-Lymphocytes, Cytotoxic/immunology , Virus Diseases/immunology , Animals , Humans , Immunity, Cellular , Mice , Models, Immunological , Receptors, KIR , Viruses/pathogenicity
6.
Open Forum Infect Dis ; 2(4): ofv172, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26719844

ABSTRACT

Background. Recent trials of recombinant, live-attenuated chimeric yellow fever-dengue tetravalent dengue vaccine (CYD-TDV) demonstrated efficacy against symptomatic, virologically confirmed dengue disease with higher point estimates of efficacy toward dengue virus (DENV)3 and DENV4 and moderate levels toward DENV1 and DENV2. It is interesting to note that serotype-specific efficacy did not correlate with absolute neutralizing antibody (nAb) geometric mean titer (GMT) values measured in a Vero-based plaque reduction neutralization test assay. The absence of Fcγ receptors on Vero cells may explain this observation. Methods. We performed parallel seroneutralization assays in Vero cells and CV-1 cells that express FcγRIIa (CV-1-Fc) to determine the neutralizing and enhancing capacity of serotype-specific DENV Abs present in CYD-TDV clinical trial sera. Results. Enhancement of DENV infection was observed in CV-1-Fc cells in naturally exposed nonvaccine sera, mostly for DENV3 and DENV4, at high dilutions. The CYD-TDV-vaccinated sera showed similar enhancement patterns. The CV-1-Fc nAb GMT values were 2- to 9-fold lower than Vero for all serotypes in both naturally infected individuals and CYD-TDV-vaccinated subjects with and without previous dengue immunity. The relative (CV-1-Fc/Vero) GMT decrease for anti-DENV1 and anti-DENV2 responses was not greater than for the other serotypes. Conclusions. In vitro neutralization assays utilizing FcγRIIa-expressing cells provide evidence that serotype-specific Ab enhancement may not be a primary factor in the serotype-specific efficacy differences exhibited in the CYD-TDV trials.

7.
J Immunol Methods ; 353(1-2): 8-19, 2010 Feb 28.
Article in English | MEDLINE | ID: mdl-19925804

ABSTRACT

Methods to prime human CD4(+) T cells in vitro would be of significant value for the pre-clinical evaluation of vaccine candidates and other immunotherapeutics. However, to date, there is no reliable method for the induction of primary human T cell responses in the laboratory. Here, we optimized a culture strategy incorporating highly purified lymphocytes and dendritic cells, in the absence of any exogenous growth factors, for the in vitro sensitization of naïve CD4(+) T cells against a variety of protein antigens. This fully autologous approach, which was superior to the more traditional PBMC assay for supporting the induction of primary human T helper cell responses in culture, elicited effector cells capable of producing a variety of Th cytokines, including IFNgamma, TNFalpha, IL-2, IL-5, IL-17 and IL-21, and memory cells that could be restimulated multiple times with a specific antigen. Through simple modifications to this culture method, we evaluated the role of dendritic cell maturation state and regulatory T cells on the sensitization of naïve T helper cells, which highlights its utility for addressing basic questions of human immunobiology. Finally, using the formulated yellow fever vaccine, YF-VAX (R), we provide a proof-of-concept demonstration of the utility of the system for evaluating the T cell immunogenicity of vaccine candidates in a pre-clinical setting.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Lymphocyte Activation , Antigen Presentation , Cell Separation , Cells, Cultured , Coculture Techniques , Humans , Immunologic Memory , Interferon-gamma/metabolism , Interleukins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/metabolism , Vaccines, Subunit/immunology , Yellow Fever Vaccine/immunology
8.
J Exp Med ; 205(3): 669-84, 2008 Mar 17.
Article in English | MEDLINE | ID: mdl-18332178

ABSTRACT

Little is known about herpesvirus modulation of T cell activation in latently infected individuals or the implications of such for chronic immune disorders. Murine gammaherpesvirus 68 (MHV68) elicits persistent activation of CD8(+) T cells bearing a Vbeta4(+) T cell receptor (TCR) by a completely unknown mechanism. We show that a novel MHV68 protein encoded by the M1 gene is responsible for Vbeta4(+) CD8(+) T cell stimulation in a manner reminiscent of a viral superantigen. During infection, M1 expression induces a Vbeta4(+) effector T cell response that resists functional exhaustion and appears to suppress virus reactivation from peritoneal cells by means of long-term interferon-gamma (IFNgamma) production. Mice lacking an IFNgamma receptor (IFNgammaR(-/-)) fail to control MHV68 replication, and Vbeta4(+) and CD8(+) T cell activation by M1 instead contributes to severe inflammation and multiorgan fibrotic disease. Thus, M1 manipulates the host CD8(+) T cell response in a manner that facilitates latent infection in an immunocompetent setting, but promotes disease during a dysregulated immune response. Identification of a viral pathogenecity determinant with superantigen-like activity for CD8(+) T cells broadens the known repertoire of viral immunomodulatory molecules, and its function illustrates the delicate balance achieved between persistent viruses and the host immune response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gammaherpesvirinae/immunology , Gammaherpesvirinae/pathogenicity , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Animals , Antigen Presentation , CD8-Positive T-Lymphocytes/classification , Chronic Disease , Gammaherpesvirinae/genetics , Immunologic Memory , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Phenotype , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/physiology , Interferon gamma Receptor
9.
J Exp Med ; 205(13): 3119-31, 2008 Dec 22.
Article in English | MEDLINE | ID: mdl-19047440

ABSTRACT

Correlates of immune-mediated protection to most viral and cancer vaccines are still unknown. This impedes the development of novel vaccines to incurable diseases such as HIV and cancer. In this study, we have used functional genomics and polychromatic flow cytometry to define the signature of the immune response to the yellow fever (YF) vaccine 17D (YF17D) in a cohort of 40 volunteers followed for up to 1 yr after vaccination. We show that immunization with YF17D leads to an integrated immune response that includes several effector arms of innate immunity, including complement, the inflammasome, and interferons, as well as adaptive immunity as shown by an early T cell response followed by a brisk and variable B cell response. Development of these responses is preceded, as demonstrated in three independent vaccination trials and in a novel in vitro system of primary immune responses (modular immune in vitro construct [MIMIC] system), by the coordinated up-regulation of transcripts for specific transcription factors, including STAT1, IRF7, and ETS2, which are upstream of the different effector arms of the immune response. These results clearly show that the immune response to a strong vaccine is preceded by coordinated induction of master transcription factors that lead to the development of a broad, polyfunctional, and persistent immune response that integrates all effector cells of the immune system.


Subject(s)
Gene Expression Regulation/immunology , Immune System Phenomena , Immunity, Innate/immunology , Vaccination , Yellow Fever Vaccine/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Proliferation , Flow Cytometry , Gene Expression Profiling , Gene Regulatory Networks , Humans , Interleukin-1beta/immunology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/physiology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transcription, Genetic
10.
J Virol ; 80(3): 1592-8, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16415035

ABSTRACT

The gammaherpesvirus immediate-early genes are critical regulators of virus replication and reactivation from latency. Rta, encoded by gene 50, serves as the major transactivator of the lytic program and is highly conserved among all the gammaherpesviruses, including Epstein-Barr virus, Kaposi's sarcoma-associated herpesvirus, and murine gammaherpesvirus 68 (gammaHV68). Introduction of a translation stop codon in gammaHV68 gene 50 (gene 50.stop gammaHV68) demonstrated that Rta is essential for virus replication in vitro. To investigate the role that virus replication plays in the establishment and maintenance of latency, we infected mice with gene 50.stop gammaHV68. Notably, the gene 50.stop virus established a long-term infection in lung B cells following intranasal infection of mice but was unable to establish latency in the spleen. This complete block in the establishment of latency in the spleen was also seen when lytic virus production was inhibited by treating mice infected with wild-type virus with the antiviral drug cidofovir, implicating virus replication and not an independent function of Rta in the establishment of splenic latency. Furthermore, we showed that gene 50.stop gammaHV68 was unable to prime the immune system and was unable to protect against a challenge with wild-type gammaHV68, despite its ability to chronically infect lung B cells. These data indicate gammaherpesviruses that are unable to undergo lytic replication in vivo may not be viable vaccine candidates despite the detection of cells harboring viral genome at late times postinfection.


Subject(s)
Rhadinovirus/genetics , Rhadinovirus/immunology , Animals , Antiviral Agents/pharmacology , Cidofovir , Codon, Terminator , Cytosine/analogs & derivatives , Cytosine/pharmacology , Genes, Viral , Herpesvirus Vaccines/genetics , Herpesvirus Vaccines/immunology , Humans , Lung/immunology , Lung/virology , Mice , Mice, Inbred C57BL , Mutation , Organophosphonates/pharmacology , Rhadinovirus/pathogenicity , Trans-Activators/genetics , Vaccination , Viral Proteins/genetics , Virus Latency/drug effects , Virus Latency/genetics
11.
J Virol ; 79(8): 5227-31, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15795307

ABSTRACT

Murine gammaherpesvirus 68 (gammaHV68) infection of mice results in the establishment of a chronic infection, which is largely maintained through latent infection of B lymphocytes. Acute virus replication is almost entirely cleared by 2 weeks postinfection. Spontaneous reactivation of gammaHV68 from latently infected splenocytes upon ex vivo culture can readily be detected at the early stages of infection (e.g., day 16). However, by 6 weeks postinfection, very little spontaneous reactivation is detected upon explant into tissue culture. Here we report that stimulation of latently infected splenic B cells harvested at late times postinfection with cross-linking surface immunoglobulin (Ig), in conjunction with anti-CD40 antibody treatment, triggers virus reactivation. As expected, this treatment resulted in B-cell activation, as assessed by upregulation of CD69 on B cells, and ultimately B-cell proliferation. Since anti-Ig/anti-CD40 stimulation resulted in splenic B-cell proliferation, we assessed whether this reactivation stimulus could overcome the previously characterized defect in virus reactivation of a v-cyclin null gammaHV68 mutant. This analysis demonstrated that anti-Ig/anti-CD40 stimulation could drive reactivation of the v-cyclin null mutant virus in latently infected splenocytes, but not to the levels observed with wild-type gammaHV68. Thus, there appears to be a role for the v-cyclin in B cells following anti-Ig/anti-CD40 stimulation independent of the induction of the cell cycle. Finally, to assess signals that are not mediated through the B-cell receptor, we demonstrate that addition of lipopolysaccharide to explanted splenocyte cultures also enhanced virus reactivation. These studies complement and extend previous analyses of Epstein-Barr virus and Kaposi's sarcoma-associated virus reactivation from latently infected cell lines by investigating reactivation of gammaHV68 from latently infected primary B cells recovered from infected hosts.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/virology , Gammaherpesvirinae/physiology , Virus Activation/physiology , Virus Latency/physiology , Animals , B-Lymphocytes/drug effects , CD40 Antigens/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Mice , Spleen/virology
12.
J Virol ; 79(15): 9480-91, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16014911

ABSTRACT

Murine gammaherpesvirus 68 (gammaHV68) provides a tractable small animal model with which to study the mechanisms involved in the establishment and maintenance of latency by gammaherpesviruses. Similar to the human gammaherpesvirus Epstein-Barr virus (EBV), gammaHV68 establishes and maintains latency in the memory B-cell compartment following intranasal infection. Here we have sought to determine whether, like EBV infection, gammaHV68 infection in vivo is associated with B-cell proliferation during the establishment of chronic infection. We show that gammaHV68 infection leads to significant splenic B-cell proliferation as late as day 42 postinfection. Notably, gammaHV68 latency was found predominantly in the proliferating B-cell population in the spleen on both days 16 and 42 postinfection. Furthermore, virus reactivation upon ex vivo culture was heavily biased toward the proliferating B-cell population. DNA methyltransferase 1 (Dnmt1) is a critical maintenance methyltransferase which, during DNA replication, maintains the DNA methylation patterns of the cellular genome, a process that is essential for the survival of proliferating cells. To assess whether the establishment of gammaHV68 latency requires B-cell proliferation, we characterized infections of conditional Dnmt1 knockout mice by utilizing a recombinant gammaHV68 that expresses Cre-recombinase (gammaHV68-Cre). In C57BL/6 mice, the gammaHV68-Cre virus exhibited normal acute virus replication in the lungs as well as normal establishment and reactivation from latency. Furthermore, the gammaHV68-Cre virus also replicated normally during the acute phase of infection in the lungs of Dnmt1 conditional mice. However, deletion of the Dnmt1 alleles from gammaHV68-infected cells in vivo led to a severe ablation of viral latency, as assessed on both days 16 and 42 postinfection. Thus, the studies provide direct evidence that the proliferation of latently infected B cells is critical for the establishment of chronic gammaHV68 infection.


Subject(s)
B-Lymphocytes/immunology , Gammaherpesvirinae/physiology , Herpesviridae Infections/immunology , Animals , Immunologic Memory , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/genetics , Time Factors , Virus Latency
13.
J Immunol ; 171(1): 17-21, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12816977

ABSTRACT

For viruses that establish persistent infection, continuous immunosurveillance by effector-competent antiviral CD8(+) T cells is likely essential for limiting viral replication. Although it is well documented that virus-specific memory CD8(+) T cells synthesize cytokines after short term in vitro stimulation, there is limited evidence that these T cells exhibit cytotoxicity, the dominant antiviral effector function. Here, we show that antiviral CD8(+) T cells in mice acutely infected by polyoma virus, a persistent mouse pathogen, specifically eliminate viral peptide-pulsed donor spleen cells within minutes after adoptive transfer and do so via a perforin-dependent mechanism. Antiviral memory CD8(+) T cells were similarly capable of rapidly mobilizing potent Ag-specific cytotoxic activity in vivo. These findings strongly support the concept that a cytotoxic effector-memory CD8(+) T cell population operates in vivo to control this persistent viral infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/immunology , Immunologic Memory , Polyomavirus/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , Acute Disease , Animals , Antiviral Agents/physiology , Cytotoxicity Tests, Immunologic , Immunophenotyping , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Polyomavirus Infections/immunology , Polyomavirus Infections/virology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/virology
14.
Nat Immunol ; 3(2): 189-95, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11812997

ABSTRACT

CD8(+) T lymphocytes mediate immunosurveillance against persistent virus infections and virus-induced neoplasia. Polyoma virus, a highly oncogenic natural mouse DNA virus, establishes persistent infection, but only a few mice are highly susceptible to tumors induced by the virus. Mature antiviral CD8(+) T cells expand in tumor-susceptible mice, but their cytotoxic effector activity is nonfunctional in vivo. Here we show that the natural killer cell inhibitory receptor, CD94-NKG2A, is up-regulated by antiviral CD8(+) T cells during acute polyoma infection and is responsible for down-regulating their antigen-specific cytotoxicity during both viral clearance and virus-induced oncogenesis.


Subject(s)
Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , Lectins, C-Type , Membrane Glycoproteins/metabolism , Polyomavirus Infections/immunology , Receptors, Immunologic/metabolism , Tumor Virus Infections/immunology , Animals , Cell Transformation, Viral , Cytotoxicity, Immunologic , Immunologic Memory , Immunologic Surveillance , Mice , Mice, Inbred C3H , Mice, Inbred CBA , NK Cell Lectin-Like Receptor Subfamily C , NK Cell Lectin-Like Receptor Subfamily D , Receptors, Natural Killer Cell
SELECTION OF CITATIONS
SEARCH DETAIL