Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Curr Top Microbiol Immunol ; 311: 117-53, 2006.
Article in English | MEDLINE | ID: mdl-17048707

ABSTRACT

T cell responses to viral infections can mediate either protective immunity or damaging immunopathology. Viral infections induce the proliferation of T cells specific for viral antigens and cause a loss in the number of T cells with other specificities. In immunologically naive hosts, viruses will induce T cell responses that, dependent on the MHC, recognize a distinct hierarchy of virus-encoded T cell epitopes. This hierarchy can change if the host has previously encountered another pathogen that elicited a memory pool ofT cells specific to a cross-reactive epitope. This heterologous immunity can deviate the normal immune response and result in either beneficial or harmful effects on the host. Each host has a unique T cell repertoire caused by the random DNA rearrangement that created it, so the specific T cells that create the epitope hierarchy differ between individuals. This "private specificity" seems of little significance in the T cell response of a naive host to infection, but it is of profound importance under conditions of heterologous immunity, where a small subset of a cross-reactive memory pool may expand and dominate a response. Examples are given of how the private specificities of immune responses under conditions of heterologous immunity influence the pathogenesis of murine and human viral infections.


Subject(s)
Immunologic Memory , T-Lymphocytes/immunology , Virus Diseases/immunology , Animals , Epitopes, T-Lymphocyte/immunology , Humans , Immunity, Active , Immunity, Cellular , Immunity, Innate/immunology , Mice , Species Specificity
2.
Nat Immunol ; 2(11): 1067-76, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11668342

ABSTRACT

A potent role for memory CD8+ T cells in heterologous immunity was shown with a respiratory mucosal model of viral infection. Memory CD8+ T cells generated after lymphocytic choriomeningitis virus (LCMV) infection were functionally activated in vivo to produce interferon-gamma (IFN-gamma) during acute infection with vaccinia virus (VV). Some of these antigen-specific memory cells selectively expanded in number, which resulted in modulation of the original LCMV-specific T cell repertoire. In addition, there was an organ-selective compartmental redistribution of these LCMV-specific T cells during VV infection. The presence of these LCMV-specific memory T cells correlated with enhanced VV clearance, decreased mortality and marked changes in lung immunopathology. Thus, the participation of pre-existing memory T cells specific to unrelated agents can alter the dynamics of mucosal immunity and disease course in response to a pathogen.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Lung/pathology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Nasal Mucosa/immunology , Respiratory Tract Infections/immunology , T-Lymphocyte Subsets/immunology , Vaccinia virus/immunology , Vaccinia/immunology , Administration, Intranasal , Animals , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/classification , Cells, Cultured/immunology , Cells, Cultured/metabolism , Cytokines/biosynthesis , Humans , Immunization , Interferon-gamma/biosynthesis , Lung/immunology , Lymphocyte Activation , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/isolation & purification , Male , Mice , Mice, Inbred C57BL , Organ Specificity , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , T-Lymphocytes, Cytotoxic/immunology , Vaccinia/pathology , Vaccinia/virology , Vaccinia virus/isolation & purification
3.
J Immunol ; 166(11): 6784-94, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11359837

ABSTRACT

The existence of gammadelta T cells has been known for over 15 years, but their significance in innate immunity to virus infections has not been determined. We show here that gammadelta T cells are well suited to provide a rapid response to virus infection and demonstrate their role in innate resistance to vaccinia virus (VV) infection in both normal C57BL/6 and beta TCR knockout (KO) mice. VV-infected mice deficient in gammadelta T cells had significantly higher VV titers early postinfection (PI) and increased mortality when compared with control mice. There was a rapid and profound VV-induced increase in IFN-gamma-producing gammadelta T cells in the peritoneal cavity and spleen of VV-infected mice beginning as early as day 2 PI. This rapid response occurred in the absence of priming, as there was constitutively a significant frequency of VV-specific gammadelta T cells in the spleen in uninfected beta TCR KO mice, as demonstrated by limiting dilution assay. Also, like NK cells, another mediator of innate immunity to viruses, gammadelta T cells in uninfected beta TCR KO mice expressed constitutive cytolytic activity. This cytotoxicity was enhanced and included a broader range of targets after VV infection. VV-infected beta TCR KO mice cleared most of the virus by day 8 PI, the peak of the gammadelta T cell response, but thereafter the gammadelta T cell number declined and the virus recrudesced. Thus, gammadelta T cells can be mediators of innate immunity to viruses, having a significant impact on virus replication early in infection in the presence or absence of the adaptive immune response.


Subject(s)
Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Vaccinia virus/immunology , Vaccinia/immunology , Vaccinia/virology , Animals , Cell Movement/genetics , Cell Movement/immunology , Cytotoxicity Tests, Immunologic , Cytotoxicity, Immunologic/genetics , Epitopes, T-Lymphocyte/analysis , Genes, T-Cell Receptor beta , Genetic Predisposition to Disease , Immunity, Cellular/genetics , Immunity, Innate/genetics , Kinetics , Lymphocyte Activation/genetics , Lymphocyte Count , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Peritoneal Cavity/cytology , Receptors, Antigen, T-Cell, gamma-delta/deficiency , Receptors, Antigen, T-Cell, gamma-delta/genetics , Survival Rate , T-Lymphocyte Subsets/virology , T-Lymphocytes, Cytotoxic/immunology , Vaccinia/genetics , Vaccinia/mortality , Virus Replication/genetics , Virus Replication/immunology
4.
J Immunol ; 166(3): 1554-61, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11160196

ABSTRACT

Infection of mice with a series of heterologous viruses causes a reduction of memory CD8(+) T cells specific to viruses from earlier infections, but the fate of the virus-specific memory CD4(+) T cell pool following multiple virus infections has been unknown. We have previously reported that the virus-specific CD4(+) Th precursor (Thp) frequency remains stable into long-term immunity following lymphocytic choriomeningitis virus (LCMV) infection. In this study, we questioned whether heterologous virus infections or injection with soluble protein CD4 Ags would impact this stable LCMV-specific CD4(+) Thp memory pool. Limiting dilution analyses for IL-2-producing cells and intracellular cytokine staining for IFN-gamma revealed that the LCMV-specific CD4(+) Thp frequency remains relatively stable following multiple heterologous virus infections or protein Ag immunizations, even under conditions that dramatically reduce the LCMV-specific CD8(+) CTL precursor frequency. These data indicate that the CD4(+) and CD8(+) memory T cell pools are regulated independently and that the loss in CD8(+) T cell memory following heterologous virus infections is not a consequence of a parallel loss in the memory CD4(+) T cell population.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Immunologic Memory , Lymphocytic choriomeningitis virus/immunology , T-Lymphocyte Subsets/immunology , Acute Disease , Amino Acid Sequence , Animals , Antigens/administration & dosage , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cell Death/immunology , Hemocyanins/administration & dosage , Hemocyanins/immunology , Hemorrhagic Fever, American/immunology , Herpesviridae Infections/immunology , Immunophenotyping , Injections, Intraperitoneal , Interferon-gamma/biosynthesis , Intracellular Fluid/chemistry , Intracellular Fluid/immunology , Intracellular Fluid/virology , Lymphocyte Activation , Lymphocyte Count , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Staining and Labeling , Stem Cells/immunology , Stem Cells/virology , T-Lymphocyte Subsets/virology , Vaccinia/immunology
5.
Microbes Infect ; 2(9): 1025-39, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10967283

ABSTRACT

CD8 T cells exist in a dynamic network whose repertoire remains static in the absence of infection but changes in the presence of foreign antigens. Individuals each have unique T-cell repertoires that continually evolve in the presence of antigen and of cross-reactive heterologous antigens, and homeostatic forces drive deletions in T-cell memory pools to accommodate the entry of new memory cells into a finite immune system.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epstein-Barr Virus Infections/immunology , Lymphocytic Choriomeningitis/immunology , Animals , Antigens, Viral , Epstein-Barr Virus Infections/virology , Flow Cytometry , Herpesvirus 4, Human/immunology , Interferon-gamma/analysis , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/immunology , Spleen/immunology , T-Lymphocyte Subsets/immunology , Virus Latency
7.
Immunity ; 11(6): 733-42, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10626895

ABSTRACT

Using a variety of techniques, including limiting dilution assays (LDA), intracellular IFNgamma assays, and Db-IgG1 MHC dimer staining to measure viral peptide-specific T cell number and function, we show here that heterologous virus infections quantitatively delete and qualitatively alter the memory pool of T cells specific to a previously encountered virus. We also show that a prior history of a virus infection can alter the hierarchy of the immunodominant peptide response to a second virus and that virus infections selectively reactivate memory T cells with distinct specificities to earlier viruses. These results are consistent with a model for the immune system that accommodates memory T cell populations for multiple pathogens over the course of a lifetime.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Immunodominant Epitopes/immunology , Immunologic Memory/immunology , Virus Diseases/immunology , Animals , Antigens, Viral/immunology , Cell Line, Transformed , Dimerization , H-2 Antigens/immunology , Histocompatibility Antigen H-2D , Immunoglobulin G/immunology , Interferon-gamma/analysis , Lymphocytic choriomeningitis virus/immunology , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muromegalovirus/immunology , Pichinde virus/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Staining and Labeling , Vaccinia virus/immunology
8.
J Exp Med ; 188(9): 1705-15, 1998 Nov 02.
Article in English | MEDLINE | ID: mdl-9802982

ABSTRACT

A basic principle of immunology is that prior immunity results in complete protection against a homologous agent. In this study, we show that memory T cells specific to unrelated viruses may alter the host's primary immune response to a second virus. Studies with a panel of heterologous viruses, including lymphocytic choriomeningitis (LCMV), Pichinde (PV), vaccinia (VV), and murine cytomegalo (MCMV) viruses showed that prior immunity with one of these viruses in many cases enhanced clearance of a second unrelated virus early in infection. Such protective immunity was common, but it depended on the virus sequence and was not necessarily reciprocal. Cell transfer studies showed that both CD4 and CD8 T cell populations from LCMV-immune mice were required to transfer protective immunity to naive hosts challenged with PV or VV. In the case of LCMV-immune versus naive mice challenged with VV, there was an enhanced early recruitment of memory phenotype interferon (IFN) gamma-secreting CD4(+) and CD8(+) cells into the peritoneal cavity and increased IFN-gamma levels in this initial site of virus replication. Studies with IFN-gamma receptor knockout mice confirmed a role for IFN-gamma in mediating the protective effect by LCMV-immune T cell populations when mice were challenged with VV but not PV. In some virus sequences memory cell populations, although clearing the challenge virus more rapidly, elicited enhanced IFN-gamma-dependent immunopathogenesis in the form of acute fatty necrosis. These results indicate that how a host responds to an infectious agent is a function of its history of previous infections and their influence on the memory T cell pool.


Subject(s)
Immunologic Memory , T-Lymphocytes/immunology , Viruses/immunology , Adipose Tissue/immunology , Adipose Tissue/pathology , Animals , Female , Immunization , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/immunology , Muromegalovirus/pathogenicity , Necrosis , Phenotype , Pichinde virus/immunology , Pichinde virus/pathogenicity , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/immunology , Vaccinia virus/pathogenicity , Virus Diseases/immunology , Virus Diseases/pathology , Virus Diseases/prevention & control , Viruses/pathogenicity
9.
J Immunol ; 158(11): 5366-73, 1997 Jun 01.
Article in English | MEDLINE | ID: mdl-9164957

ABSTRACT

Generally, it has been assumed that memory T cells are dormant and inactive cells in the absence of their specific Ag. Recent work has challenged this assumption by showing that a portion of the CD8+ memory T cell pool is in cycle. In this study, we demonstrate that a significant number of blast-size memory CD8+ T cells in mice, long after lymphocytic choriomeningitis virus (LCMV) infection, mediate cytolysis against highly sensitive targets without any in vivo or in vitro restimulation and expansion with Ag. Peptide-coated RMA-S targets were sufficiently sensitive to detect low but significant cytolytic activity in bulk 51Cr release assays in nonstimulated LCMV-specific splenic memory CTL populations. Most of the directly cytotoxic activity was against the GP33 epitope, and this persisted throughout the lifetime of the mouse following infection. The cytotoxic activity was not inhibited by cyclosporin A, indicating that these cells were already in an active state and not dependent on further stimulation in vitro. It was formally shown that the cytotoxic activity was mediated by the CD8+ CTL by sorting for the blast-size CD8+ population and by blocking target cell lysis with anti-CD8 Ab. Thus, at any time after the original infection some portion of the memory CD8+ T cell pool is cycling, and it remains cytolytically active long after resolution of the original infection. These CTL may provide a rapidly acting defense mechanism against reinfection.


Subject(s)
Immunologic Memory , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Viral/immunology , Mice , Mice, Inbred C57BL
10.
Immunol Rev ; 159: 79-93, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9416504

ABSTRACT

Both alpha beta and gamma delta T-cell populations and natural killer (NK) cells include cytotoxic, interferon (IFN)-gamma-producing lymphocytes that actively respond to viral infections. We show here that all three populations can provide "natural resistance" to viruses very early in infection and describe how the T-cell populations are modulated to provide this function. gamma delta T cells were shown to play a role in controlling vaccinia virus (VV) infections, as VV grew to much higher titers in gamma delta T-cell knockout mice than in normal mice 3-4 days post-infection. Our studies of the alpha beta T-cell responses to viruses revealed an interactive network of T cells that is modulated substantially during systemic infections. There is an induction phase associated with a massive virus-specific CD8 T-cell response, an apoptosis phase during which the T cells become sensitized to activation-induced cell death (AICD), a silencing phase, during which the T-cell number and activation state is reduced, and, finally, a memory phase associated with the very stable preservation of virus-specific memory cytotoxic T-lymphocyte precursors (pCTL). Infection of mice immune to one virus with a heterologous virus leads to a selective expansion of memory CTL cross-reacting between the two viruses, but, after homeostasis is again established, there is a quantitative reduction and qualitative alteration of memory to the first virus. Our results suggest that memory alpha beta T cells cross-reactive between heterologous viruses mediate both immunopathology and protective immunity at early stages of the second virus infection. Thus, memory alpha beta T cells can, like gamma delta T cells and NK cells, provide natural immunity to viral infections.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Virus Diseases/immunology , Animals , Humans , Immunity, Innate
11.
J Exp Med ; 183(6): 2489-99, 1996 Jun 01.
Article in English | MEDLINE | ID: mdl-8676069

ABSTRACT

Experimental analyses of the acute cytotoxic T lymphocyte (CTL) response to viruses have focused on studying these infections in immunologically naive hosts. In the natural environment, however, viral CTL responses occur in hosts that are already immune to other infectious agents. To address which factors contribute to the maintenance and waning of immunological memory, the following study examined the frequencies of virus-specific CTL precursor cells (pCTL) not only using the usual experimental paradigm where mice undergo acute infections with a single virus, and in mice immune to a single virus, but also in immune mice after challenge with various heterologous viruses. As determined by limiting dilution assays, the pCTL frequency (p/f) per CD8+ T cell specific for lymphocytic choriomeningitis virus (LCMV), Pichinde virus (PV), or vaccinia virus (VV) increased during the acute infections, peaking at days 7-8 with frequencies as high as 1/27-1/74. Acute viral infections such as these elicit major expansions in the CD8+ T cell number, which has been reported to undergo apoptosis and decline after most of the viral antigen has been cleared. Although the decline in the total number of virus-specific pCTL after their peak in the acute infection was substantial, for all three viruses the virus-specific p/f per CD8+ T cell decreased only two- to fourfold and remained at these high levels with little fluctuation for well over a year. The ratios of the three immunodominant peptide-specific to total LCMV-specific clones remained unchanged between days 7 and 8 of acute infection and long-term memory, suggesting that the apoptotic events did not discriminate on the basis of T cell receptor specificity, but instead nonspecifically eliminated a large proportion of the activated T cells. However, when one to five heterologous viruses (LCMV, PV, VV, murine cytomegalovirus, and vesicular stomatitis virus) were sequentially introduced into this otherwise stable memory pool, the stability of the memory pool was disrupted. With each successive infection, after the immune system had returned to homeostasis, the memory p/f specific to viruses from earlier infections declined. Reductions in memory p/f were observed in all tested immunological compartments (spleen, peripheral blood, lymph nodes, and peritoneal cavity), and on average in the spleen revealed a 3 +/- 0.4-fold decrease in p/f after one additional viral infection and an 8.4 +/- 3-fold decrease after two additional viral infections. Thus, subsequent challenges with heterologous antigens, which themselves induce memory CTL, may contribute to the waning of CTL memory pool to earlier viruses as the immune system accommodates ever-increasing numbers of new memory cells within a limited lymphoid population. This demonstrates that virus infections do not occur in immunological isolation, and that CD8+ T cell responses are continually being modulated by other infectious agents.


Subject(s)
Immunologic Memory , Lymphocytic choriomeningitis virus/immunology , Pichinde virus/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Vaccinia virus/immunology , Vesicular stomatitis Indiana virus/immunology , Virus Diseases/immunology , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cricetinae , Lymph Nodes/immunology , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Spleen/immunology , Viral Proteins/chemistry , Viral Proteins/immunology
12.
J Cell Biochem ; 59(2): 135-42, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8904307

ABSTRACT

Apoptosis is an important mechanism enabling the selection of the non-self-reactive T cell repertoire and for maintaining homeostasis in the immune system after it has expanded to combat infections. Highly activated, proliferating T cells become susceptible to apoptosis driven by a number of stimuli, and T cells activated during a viral infection become susceptible to "activation induced cell death" after repeated stimulation through the T cell receptor (TcR). This is a major mechanism for the immune deficiencies observed during many viral infections. During infections with a high antigen load this can lead to a selective deletion of virus-specific cytotoxic T lymphocytes (CTL) and to the establishment of persistent infection. More commonly, the CTL control the infection first, and high levels of apoptosis in the expanded lymphocyte population occur after antigen and growth factors become limiting. This cell death does not seem to depend on TcR specificity, as the residual population contains a remarkably stable population of memory CTL precursors that approximate the frequency per CD8 cell of that seen during the peak of the acute infection. Subsequent infections with heterologous viruses result in an expansion and then an apoptotic elimination of T cells, with the consequence being a reduction in precursor CTL specific for the first virus. Thus, apoptosis shapes the quality and quantity of T cell memory.


Subject(s)
Apoptosis/physiology , Immune Tolerance , Immunologic Memory , T-Lymphocytes/immunology , Virus Diseases/immunology , Animals , Cellular Senescence , Humans , Lymphocyte Activation , T-Lymphocytes, Cytotoxic
13.
J Immunol ; 154(8): 4010-7, 1995 Apr 15.
Article in English | MEDLINE | ID: mdl-7706740

ABSTRACT

Anti-viral CTL were induced in vitro using a particle bombardment device or "gene-gun" to deliver plasmid DNA encoding the nucleoprotein of the lymphocytic choriomeningitis virus (LCMV). Using this plasmid we were able to study T cell-mediated immunity in the absence of a neutralizing Ab response. Upon a single DNA immunization, a nearly 2 log10 reduction in splenic viral titers was observed 3 days after LCMV infection. After two or three immunizations a greater than 3 log10 inhibition of viral titers in the spleen was observed, with most animals having no detectable virus. C57BL/6 mice immunized with DNA encoding the nucleoprotein gene were also challenged with LCMV intracranially. Upon intracranial challenge, vaccinated animals displayed either protection or enhanced immunopathology leading to accelerated kinetics of death. Using limiting dilution analysis it was possible to detect LCMV-specific CTL precursors in both the spleen and lymph nodes of vaccinated animals. C57BL/6 mice inoculated with DNA demonstrated an anamnestic CTL response detectable at day 4 after LCMV challenge. Thus DNA vaccines are capable of inducing an anti-viral T cell response that can inhibit viral replication and mediate either protective immunity or immunopathology. Vaccination with DNA may therefore provide a useful alternative to current viral or subunit vaccines once the efficacy of immunization with DNA is optimized.


Subject(s)
Cytotoxicity, Immunologic , DNA, Viral/administration & dosage , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Vaccines/genetics , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Gene Transfer Techniques , Immunity, Cellular , Immunologic Memory , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/prevention & control , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nucleoproteins/chemistry , Nucleoproteins/immunology , Peptides/chemistry , Peptides/immunology
14.
Curr Opin Immunol ; 6(4): 553-9, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7524536

ABSTRACT

Recent studies have defined an immunological network by which the acute cytotoxic T-lymphocyte response to viral infection modulates or is modulated by the antigen load and by crossreactive memory T cells. Down regulation of the acute CTL response can be associated with either antigen-dependent or antigen-independent apoptosis, and the host enters a state of immune deficiency as these T cells become sensitized to apoptotic mechanisms.


Subject(s)
Apoptosis , Epitopes/immunology , T-Lymphocytes, Cytotoxic/immunology , Virus Diseases/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Humans , Immunologic Memory , Isoantigens/immunology
15.
J Exp Med ; 179(6): 1933-43, 1994 Jun 01.
Article in English | MEDLINE | ID: mdl-8195718

ABSTRACT

Analyses of the relationships between different viruses and viral proteins have focused on homologies between linear amino acid sequences, but cross-reactivities at the level of T cell recognition may not be dependent on a conserved linear sequence of several amino acids. The CTL response to Pichinde virus (PV) and vaccinia virus (VV) in C57BL/6 mice previously immunized with lymphocytic choriomeningitis virus (LCMV) included the reactivation of memory cytotoxic T lymphocyte (CTL) specific to LCMV. Limiting dilution assays (LDA) demonstrated that at least part of this reactivation of memory cells in LCMV-immune mice related to cross-reactivity at the clonal level, even though acute infections with these viruses in nonimmune mice elicited CTL responses that did not cross-react in conventional bulk CTL assays. Precursor CTL (pCTL) to LCMV were generated in splenic leukocytes from LCMV-immune mice acutely infected with PV or VV when stimulated in vitro with only the second virus but not with uninfected peritoneal exudate cells (PECs). Cytotoxicity mediated by LCMV-specific CTL clones activated by PV infection was greatly inhibited by anti-CD8 antibody, suggesting that these memory CTL clones recognizing LCMV-infected targets were of low affinity. LCMV-immune splenocytes stimulated in vitro with PV or VV demonstrated a low but significant precursor frequency (p/f) to the heterologous viruses, and splenocytes from PV- or VV-immune mice when stimulated in vitro against LCMV generated a low but significant p/f to LCMV. Short-term CTL clones cross-reactive between LCMV and PV were derived from splenic leukocytes from LCMV-immune mice acutely infected with PV. To distinguish whether the cross-reactivity was directed against a viral peptide or a virus-induced endogenous cellular neoantigen, we demonstrated that a pCTL frequency to PV about 1/4-1/7 that of the frequency to LCMV could be generated from LCMV-immune splenic leukocytes stimulated with the immunodominant LCMV NP peptide. A partially homologous PV peptide generated from the equivalent site to the LCMV NP peptide did not sensitize targets to lysis by either LCMV- or PV-specific CTLs, suggesting that the cross-reactivity in killing was not due to evolutionarily conserved equivalent sequences. Experiments also indicated that prior immunity to one virus could modulate future primary immune responses to a second virus. Elevated pCTL frequencies to PV were seen after acute PV infection of LCMV-immune mice, and elevated pCTL frequencies to LCMV were seen after acute LCMV infection of PV- and VV-immune mice.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Immunologic Memory , Lymphocytic choriomeningitis virus/immunology , Pichinde virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/immunology , Viral Vaccines/immunology , Animals , Base Sequence , CD8 Antigens/immunology , Cell Line, Transformed , Cross Reactions , Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oligopeptides/chemical synthesis , Oligopeptides/immunology , Time Factors
16.
Scand J Immunol ; 36(1): 107-17, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1352063

ABSTRACT

The human tumour cell lines RPMI 8226 and Daudi are potent inducers of V gamma 9-expressing T cells. The inducing element of RPMI 8226 has not been defined but evidence suggests that a member of the GroEL heat shock protein (HSP) family (HSP 58) may have a role in the induction by Daudi cells. The present study examined the reactivity patterns of gamma delta T-cell clones generated in response to RPMI 8226 and addressed the possible role of HSP 58 in this process. RPMI 8226 induced a population of V gamma 9 TCR+ cells which were heterogeneous in terms of their cell surface markers, patterns of proliferation and cytotoxic responses. All clones expressed CD3, CD2, CD18 and CD29. They demonstrated variability in expression of CD56, CD8 and HLA-DR. RPMI 8226 stimulated proliferation in purified bulk gamma delta cultures and clones. Daudi was also capable of inducing these cells to proliferate while mycobacterial products were not effective. The clones demonstrated a limited non-MHC-restricted cytotoxicity pattern with some evidence of clonal heterogeneity. Although both Daudi and RPMI 8226 were sensitive to lysis by the clones, cold inhibition experiments indicated differential activity towards these targets. Anti-HSP 58 was inhibitory to gamma delta T-cell induction by RPMI 8226, Daudi and mycobacterial products. However, the anti-HSP 58 antibody appears to bind to the surface of at least six different tumour cell lines with no correlation to their ability to induce gamma delta T cells and the anti-HSP 58 inhibited non-gamma delta responses.


Subject(s)
Bacterial Proteins/immunology , Heat-Shock Proteins/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Tumor Cells, Cultured/immunology , Chaperonin 60 , Clone Cells , Cytotoxicity, Immunologic , Humans , In Vitro Techniques , Interleukin-2/pharmacology , Lymphocyte Activation , Recombinant Proteins
17.
Ann Neurol ; 30(6): 794-800, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1838679

ABSTRACT

T cells are postulated to contribute to the injury of the oligodendrocyte-myelin complex underlying the demyelinating disease multiple sclerosis (MS). The apparent lack of class I or II major histocompatibility complex (MHC) expression in situ on human oligodendrocytes and the consistent failure to identify a universal myelin antigen in MS suggest that the immune damage might be mediated by effector T cells that are capable of reacting in an antigen-nonspecific and possibly MHC-unrestricted manner, such as T cells expressing the gamma-delta T-cell receptor. Since gamma-delta T cells are reported to be present in MS plaques and an increased number are found in the cerebrospinal fluid of patients with MS, we directly examined whether gamma-delta T cells are capable of inducing injury to human oligodendrocytes. We found, using a 6-hour 51Cr release assay, that oligodendrocytes cultured from surgically resected human brain specimens were effectively lysed in a dose-dependent manner by human gamma-delta T cells (28 +/- 5% mean specific lysis, n = 6, at an effector-target ratio of 20:1). Although heat shock protein HSP72, a putative gamma-delta T-cell recognition molecule, could be induced in vitro in our oligodendrocytes, an antibody to HSP72 did not inhibit gamma-delta T cell-mediated lysis of oligodendrocytes. These results suggest that gamma-delta T cells gaining entry into the central nervous system may be deleterious to oligodendrocytes and thus may contribute to the pathogenesis of MS.


Subject(s)
Autoimmune Diseases/pathology , Brain/pathology , Multiple Sclerosis/pathology , Oligodendroglia/immunology , Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocyte Subsets/immunology , Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , Cytotoxicity, Immunologic , Dose-Response Relationship, Immunologic , Epilepsy/pathology , Heat-Shock Proteins/physiology , Humans , Models, Biological , Multiple Sclerosis/etiology , Multiple Sclerosis/immunology , T-Lymphocyte Subsets/pathology
18.
Can J Infect Dis ; 1(2): 35-40, 1990.
Article in English | MEDLINE | ID: mdl-22553437

ABSTRACT

Thirty-seven adult patients with acute urinary tract infections (UTI) were randomized to receive either a seven day (lower UTI) or a 14 day (upper UTI) course of norfloxacin 400 mg orally twice daily, or nalidixic acid 1 g orally four times per day. Mean age, underlying disease and infecting organisms were similar in the two groups. Nine patients in the norfloxacin group and seven in the nalidixic acid group had presumptive evidence of upper UTI. Overall, 12 patients had antibody-coated bacteria-positive infections. The infecting organisms were: Escherichia coli (27), coagulase-negative staphylococci (four), Citrobacter freundii (three), Klebsiella pneumoniae (three), and Proteus mirabilis, Proteus vulgaris, Pseudomonas aeruginosa, Enterobacter agglomerans, Streptococcus agalactiae, Enterococcus faecalis (one of each). All of the organisms were susceptible to norfloxacin, while 81% were susceptible to nalidixic acid. The effects on the periurethral and anal canal flora were similar in both groups. Five patients in each group experienced adverse clinical effects. The cure rates for norfloxacin and nalidixic acid were 79 and 83%, respectively. There were two failures, two relapses and four reinfections in the norfloxacin group. In the nalidixic acid group, there were two failures, one relapse and four reinfections. One of the failure patients in the nalidixic acid group developed resistance to the drug, and two of the four reinfections were due to organisms resistant to nalidixic acid. In this patient population it was concluded that nalidixic acid may be as effective as norfloxacin in the treatment of acute, symptomatic UTI.

20.
Can J Physiol Pharmacol ; 58(2): 220-2, 1980 Feb.
Article in English | MEDLINE | ID: mdl-6769574

ABSTRACT

Porcine follicular fluid (PFF) contains a factor (inhibin or folliculostatin) which is reported to selectively inhibit the secretion of follicle-stimulating hormone (FSH) from the anterior pituitary gland. Chronic treatment of hemicastrate immature rats with PFF is able to partially inhibit the FSH-mediated hypertrophy of the remaining testis. However, the pituitaries from PFF-treated rats are paradoxically very sensitive to stimulation with gonadotrophin-releasing hormone (GnRH) and secrete significantly more FSH than control glands. Furthermore, this increased sensitivity results in a large increase in luteinizing hormone (LH) secretion. These observations suggest that under certain circumstances PFF is not selective for FSH and that it surprisingly stimulates rather than inhibits gonadotrophin secretion.


Subject(s)
Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropin-Releasing Hormone/pharmacology , Ovarian Follicle/physiology , Pituitary Gland, Anterior/drug effects , Proteins/therapeutic use , Testis/physiology , Animals , Female , Follicle Stimulating Hormone/metabolism , Gonadotropin-Releasing Hormone/therapeutic use , Inhibins , Luteinizing Hormone/metabolism , Male , Pituitary Gland, Anterior/metabolism , Rats , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...