首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
2.
Herpes simplex virus (HSV) inhibits major histocompatibility complex (MHC) class I expression in infected cells and does so much more efficiently in human cells than in murine cells. Given this difference, if MHC class I-restricted T cells do not play an important role in protection of mice from HSV, an important role for these cells in humans would be unlikely. However, the contribution of MHC class I-restricted T cells to the control of HSV infection in mice remains unclear. Further, the mechanisms by which these cells may act to control infection, particularly in the nervous system, are not well understood, though a role for gamma interferon (IFN-γ) has been proposed. To address the roles of MHC class I and of IFN-γ, C57BL/6 mice deficient in MHC class I expression (β2 microglobulin knockout [β2KO] mice), in IFN-γ expression (IFN-γKO mice), or in both (IFN-γKO/β2KO mice) were infected with HSV by footpad inoculation. β2KO mice were markedly compromised in their ability to control infection, as indicated by increased lethality and higher concentrations of virus in the feet and spinal ganglia. In contrast, IFN-γ appeared to play at most a limited role in viral clearance. The results suggest that MHC class I-restricted T cells play an important role in protection of mice against neuroinvasive HSV infection and do so largely by mechanisms other than the production of IFN-γ.

Two gene products of herpes simplex virus (HSV) block presentation of viral proteins by class I major histocompatibility complex (MHC) molecules: the viral host shutoff protein (vhs), which is present in the viral particle, and the immediate-early protein ICP47 (1, 14, 41, 42). Through the sequential action of these proteins, antigen presentation by MHC class I is inhibited early in the viral replication cycle. ICP47 binds to human transporter associated with antigen-processing proteins (TAP), thereby inhibiting peptide loading on MHC class I and recognition by HSV-specific, MHC class I-restricted, CD8+ T cells (1, 14, 42, 43). This effect is greatest in nonhematopoietic cells in which the abundance of MHC class I and TAP are lower than in antigen-presenting cells (41). As a consequence, HSV is more likely to impair recognition of infected target cells in the tissues than to block the generation of antigen-specific CD8+ T cells. Consistent with this, recent studies indicate that HSV antigen-specific CD8+ cytotoxic-T-lymphocyte (CTL) precursors can be readily detected in the blood and cutaneous lesions of HSV-infected individuals (16, 31, 32). However, NK cells and HSV antigen-specific CD4+ T cells are detected earlier than antigen-specific CD8+ T cells in lesions of humans with recurrent HSV-2 disease (16). This finding has led to the proposal that gamma interferon (IFN-γ) produced by infiltrating NK and CD4+ T cells overrides the inhibitory effects of HSV on TAP function and MHC class I expression (22, 41), thereby allowing the eradication of virus by CD8+ T cells, whose numbers increase in lesions around the time of viral clearance (16, 31). In patients with AIDS, a lower frequency in the blood of HSV antigen-specific CD8+ CTL precursors is associated with more frequent and severe recurrences of genital disease (32). These correlative data suggest that CD8+ T cells may play an important role in the clearance of HSV in humans, at least from mucocutaneous lesions.ICP47 inhibits murine TAP poorly (1, 42), which may explain the greater ease with which anti-HSV CD8+ CTLs have been detected in mice than in humans (3, 8, 28, 34, 35). Despite the weak interaction of ICP47 with murine TAP, results of a recent study (12) suggested that ICP47 impairs CD8+ T-cell-dependent viral clearance from the nervous system: CD8+ T cells protected susceptible BALB/c or A/J mice from lethal, nervous system infection with an HSV mutant lacking ICP47 but did not appear to protect against infection with wild-type HSV or to contribute to clearance of either virus from the eye. These findings are consistent with data suggesting that CD8+ T cells limit persistence of HSV in the spinal ganglia and decrease spread to the central nervous system (35, 36). However, other studies have concluded that CD4+ T cells but not CD8+ T cells play the critical role in viral clearance and protection from lethal primary infection with wild-type HSV (20, 23, 24) or that either CD4+ or CD8+ T cells are sufficient for protection (26, 37). Since the effects of ICP47 are likely to be greater in humans than in mice, if MHC class I-restricted CD8+ T cells do not play an important role in protection of mice from lethal, neuroinvasive infection due to wild-type HSV, an important role in humans would be unlikely.The mechanisms by which T cells may limit the spread of infection in the nervous system are not clearly understood. Studies by Simmons and colleagues suggested that CD8+ T cells may lyse infected Schwann cells or satellite cells but that they probably do not lyse infected neurons (31, 32). They and others have proposed that CD8+ T cells protect neurons through the production of cytokines, in particular IFN-γ (35, 36). IFN-γ contributes to the clearance of HSV from mucocutaneous sites (4, 24, 25, 37, 44). However, the role of IFN-γ in protection from lethal, neuroinvasive infection is uncertain and may vary with the strain of mice, method used to inhibit IFN-γ function, and route of inoculation (4, 5, 24, 37, 44). IFN-γ is produced in the ganglia of mice with acute or latent HSV infection (5, 13, 19). Both CD4+ and CD8+ T cells (and NK cells) produce IFN-γ, but CD4+ T cells appear to be the predominant source of IFN-γ following intravaginal infection with HSV (24, 25). Thus, it is possible that the disparity in results regarding the relative importance of CD4+ and CD8+ T cells in protection from lethal, neuroinvasive HSV infection reflects their redundant roles in production of this cytokine or that IFN-γ and CD8+ T cells contribute independently to control of infection in the nervous system.To address in parallel the contributions of MHC class I-restricted T cells and of IFN-γ to protection of mice from HSV, MHC class I and CD8+ T-cell-deficient β2 microglobulin knockout (β2KO) mice, IFN-γ knockout (IFN-γKO) mice, and mice deficient in both MHC class I and IFN-γ expression (IFN-γKO/β2KO) were studied. The results indicated that loss of MHC class I expression in β2KO mice substantially increased their susceptibility to HSV, whereas the loss of IFN-γ expression had a much more limited effect. These findings indicate that MHC class I-restricted T cells play an important role in protection against neuroinvasive HSV infection in mice and that they do so largely by mechanisms other than the production of IFN-γ. Though MHC class I expression is more severely impaired in β2KO mice than in human cells infected with wild-type HSV, these findings support the notion that inhibition of MHC class I expression is an important factor in the virulence of this virus.  相似文献   

3.
Pseudorabies virus (PRV; suid herpesvirus 1) infection causes heavy economic losses in the pig industry. Therefore, vaccination with live attenuated viruses is practiced in many countries. This vaccination was demonstrated to induce extrathymic virus-specific memory CD4+CD8+ T lymphocytes. Due to their major histocompatibility complex (MHC) class II-restricted proliferation, it is generally believed that these T lymphocytes function as memory T-helper cells. To directly prove this hypothesis, 15-amino-acid, overlapping peptides of the viral glycoprotein gC were used for screening in proliferation assays with peripheral blood mononuclear cells of vaccinated d/d haplotype inbred pigs. In these experiments, two naturally processed T-cell epitopes (T1 and T2) which are MHC class II restricted were identified. It was shown that extrathymic CD4+CD8+ T cells are the T-lymphocyte subpopulation that responds to epitope T2. In addition, we were able to show that cytokine secretion can be induced in these T cells through recall with inactivated PRV and demonstrated that activated PRV-primed CD4+CD8+ T cells are able to induce PRV-specific immunoglobulin synthesis by PRV-primed, resting B cells. Taken together, these results demonstrate that the glycoprotein gC takes part in the priming of humoral anti-PRV memory responses. The experiments identified the first T-cell epitopes so far known to induce the generation of virus-specific CD4+CD8+ memory T lymphocytes and showed that CD4+CD8+ T cells are memory T-helper cells. Therefore, this study describes the generation of virus-specific CD4+CD8+ T cells, which is observed during vaccination, as a part of the potent humoral anti-PRV memory response induced by the vaccine.Pseudorabies virus (PRV), a member of the Alphaherpesvirinae, is the causative agent of Aujeszky’s disease. This disease is lethal to young pigs and causes important economic losses (52). Therefore, vaccination of pigs is practiced in many countries.Several humoral immune system effector mechanisms are involved in the protection of pigs from PRV infection. Virus-neutralizing antibodies, antibodies mediating antibody-dependent cell-mediated cytotoxicity, and antibodies mediating complement-mediated lysis of PRV-infected target cells have been demonstrated (22, 23, 53, 54). The main targets of this humoral immune response were shown to be the viral glycoproteins (3, 45), and passive immunization with monoclonal antibodies (MAbs) against gB, gC, and gD protects pigs from a lethal challenge (20, 49).The protection conferred through cell-mediated immunity is poorly understood. An increase in major histocompatibility complex (MHC)-unrestricted cell-mediated cytotoxicity against uninfected and PRV-infected cells has been detected after infection or vaccination of pigs with PRV (16, 53, 54), and specific cellular immune responses to PRV infections could be demonstrated by stimulation of proliferation and lymphokine secretion of porcine PRV-immune lymphocytes (10, 17, 42, 43, 51) as well as by the detection of PRV-specific cytotoxic lymphocytes (21, 56).There are some difficulties in defining more precisely the impact of cell-mediated immune effector mechanisms to protection from PRV-infection and their interplay with the observed humoral immune response. Considerably fewer porcine than human or mouse differentiation markers are available (34). In addition, the immune system of swine differs considerably from that of humans and mice. The pig has a substantial number of CD4CD8 T lymphocytes in the peripheral blood (4, 6, 12, 36, 39). In young animals, this subpopulation of T lymphocytes comprises up to 60% of the T lymphocytes and contains mainly γδ T lymphocytes. The pig is also the only species so far known to contain a substantial number of resting extrathymic CD4+CD8+ T lymphocytes (28, 36, 39). This T-lymphocyte population shows morphologically the phenotype of mature T lymphocytes (40) and increases with age to up to 60% of peripheral T lymphocytes (29, 35, 39, 55). Further, it was demonstrated that CD4+CD8+ T lymphocytes comprise memory T cells which proliferate upon stimulation with recall antigen (43, 55). Since the observed proliferative response was shown to be MHC class II-restricted, it was speculated that the porcine CD4+CD8+ T-cell subset contains memory T-helper lymphocytes (43). However, the ability of these T lymphocytes to secrete cytokines or to provide help to B cells has so far not been demonstrated.To gain a better understanding of immune effector mechanisms conferring protection from PRV infection, the function of these unusual extrathymic T-lymphocyte subsets has to be elucidated. In the present study, we identified two T-cell epitopes on glycoprotein gC which are primed during vaccination of d/d haplotype inbred pigs (41) against PRV and demonstrated that MHC class II-restricted, peripheral CD4+CD8+ memory T lymphocytes are the responding T lymphocytes. We were further able to show that PRV-specific, extrathymic CD4+CD8+ T lymphocytes are able to secrete cytokines and have the capacity to stimulate the secretion of PRV-specific immunoglobulins (Ig) by PRV-primed B cells. These results demonstrate that porcine CD4+CD8+ T lymphocytes can function as memory T-helper cells and can direct humoral anti-PRV memory responses.  相似文献   

4.
Most individuals infected with human immunodeficiency virus type 1 (HIV-1) initially harbor macrophage-tropic, non-syncytium-inducing (M-tropic, NSI) viruses that may evolve into T-cell-tropic, syncytium-inducing viruses (T-tropic, SI) after several years. The reasons for the more efficient transmission of M-tropic, NSI viruses and the slow evolution of T-tropic, SI viruses remain unclear, although they may be linked to expression of appropriate chemokine coreceptors for virus entry. We have examined plasma viral RNA levels and the extent of CD4+ T-cell depletion in SCID mice reconstituted with human peripheral blood leukocytes following infection with M-tropic, dual-tropic, or T-tropic HIV-1 isolates. The cell tropism was found to determine the course of viremia, with M-tropic viruses producing sustained high viral RNA levels and sparing some CD4+ T cells, dual-tropic viruses producing a transient and lower viral RNA spike and extremely rapid depletion of CD4+ T cells, and T-tropic viruses causing similarly lower viral RNA levels and rapid-intermediate rates of CD4+ T-cell depletion. A single amino acid change in the V3 region of gp120 was sufficient to cause one isolate to switch from M-tropic to dual-tropic and acquire the ability to rapidly deplete all CD4+ T cells.The envelope gene of human immunodeficiency virus type 1 (HIV-1) determines the cell tropism of the virus (11, 32, 47, 62), the use of chemokine receptors as cofactors for viral entry (4, 17), and the ability of the virus to induce syncytia in infected cells (55, 60). Cell tropism is closely linked to but probably not exclusively determined by the ability of different HIV-1 envelopes to bind CD4 and the CC or the CXC chemokine receptors and initiate viral fusion with the target cell. Macrophage-tropic (M-tropic) viruses infect primary cultures of macrophages and CD4+ T cells and use CCR5 as the preferred coreceptor (2, 5, 15, 23, 26, 31). T-cell-tropic (T-tropic) viruses can infect primary cultures of CD4+ T cells and established T-cell lines, but not primary macrophages. T-tropic viruses use CXCR4 as a coreceptor for viral entry (27). Dual-tropic viruses have both of these properties and can use either CCR5 or CXCR4 (and infrequently other chemokine receptors [25]) for viral entry (24, 37, 57). M-tropic viruses are most frequently transmitted during primary infection of humans and persist throughout the duration of the infection (63). Many, but not all, infected individuals show an evolution of virus cell tropism from M-tropic to dual-tropic and finally to T-tropic with increasing time after infection (21, 38, 57). Increases in replicative capacity of viruses from patients with long-term infection have also been noted (22), and the switch to the syncytium-inducing (SI) phenotype in T-tropic or dual-tropic isolates is associated with more rapid disease progression (10, 20, 60). Primary infection with dual-tropic or T-tropic HIV, although infrequent, often leads to rapid disease progression (16, 51). The viral and host factors that determine the higher transmission rate of M-tropic HIV-1 and the slow evolution of dual- or T-tropic variants remain to be elucidated (4).These observations suggest that infection with T-tropic, SI virus isolates in animal model systems with SCID mice grafted with human lymphoid cells or tissue should lead to a rapid course of disease (1, 8, 4446). While some studies in SCID mice grafted with fetal thymus and liver are in agreement with this concept (33, 34), our previous studies with the human peripheral blood leukocyte-SCID (hu-PBL-SCID) mouse model have shown that infection with M-tropic isolates (e.g., SF162) causes more rapid CD4+ T-cell depletion than infection with T-tropic, SI isolates (e.g., SF33), despite similar proviral copy numbers, and that this property mapped to envelope (28, 41, 43). However, the dual-tropic 89.6 isolate (19) caused extremely rapid CD4+ T-cell depletion in infected hu-PBL-SCID mice that was associated with an early and transient increase in HIV-1 plasma viral RNA (29). The relationship between cell tropism of the virus isolate and the pattern of disease in hu-PBL-SCID mice is thus uncertain. We have extended these studies by determining the kinetics of HIV-1 RNA levels in serial plasma samples of hu-PBL-SCID mice infected with primary patient isolates or laboratory stocks that differ in cell tropism and SI properties. The results showed significant differences in the kinetics of HIV-1 replication and CD4+ T-cell depletion that are determined by the cell tropism of the virus isolate.  相似文献   

5.
We have evaluated the potential of conferring protective immunity to herpes simplex virus type 2 (HSV-2) by selectively inducing an HSV-specific CD8+ cytotoxic T-lymphocyte (CTL) response directed against a single major histocompatibility complex class I-restricted CTL recognition epitope. We generated a recombinant vaccinia virus (rVV-ES-gB498-505) which expresses the H-2Kb-restricted, HSV-1/2-cross-reactive CTL recognition epitope, HSV glycoprotein B residues 498 to 505 (SSIEFARL) (gB498-505), fused to the adenovirus type 5 E3/19K endoplasmic reticulum insertion sequence (ES). Mucosal immunization of C57BL/6 mice with this recombinant vaccinia virus induced both a primary CTL response in the draining lymph nodes and a splenic memory CTL response directed against HSV gB498-505. To determine the ability of the gB498-505-specific memory CTL response to provide protection from HSV infection, immunized mice were challenged with a lethal dose of HSV-2 strain 186 by the intranasal (i.n.) route. Development of the gB498-505-specific CTL response conferred resistance in 60 to 75% of mice challenged with a lethal dose of HSV-2 and significantly reduced the levels of infectious virus in the brains and trigeminal ganglia of challenged mice. Finally, i.n. immunization of C57BL/6 mice with either a recombinant influenza virus or a recombinant vaccinia virus expressing HSV gB498-505 without the ES was also demonstrated to induce an HSV-specific CTL response and provide protection from HSV infection. This finding confirms that the induction of an HSV-specific CTL response directed against a single epitope is sufficient for conferring protective immunity to HSV. Our findings support the role of CD8+ T cells in the control of HSV infection of the central nervous system and suggest the potential importance of eliciting HSV-specific mucosal CD8+ CTL in HSV vaccine design.

Both humoral and cell-mediated components of the immune response are involved in controlling herpes simplex virus (HSV) infection (51, 61). Studies of humans and of mice have implicated a role for both CD8+ (6, 25, 32, 33, 47, 6567) and CD4+ (27, 3739, 52, 53) T-lymphocyte subsets in mediating protection against HSV infection. For example, CD8+ T cells have been shown to be important in limiting replication of HSV in the footpad (6) and colonization of the spinal dorsal root ganglia (6, 66). In contrast, other studies using a zosteriform model of infection have primarily indicated a role for CD4+ T cells in the clearance of HSV (3739). Both CD4+ and CD8+ (56, 72, 7476) HSV-specific T lymphocytes have been detected in humans seropositive for HSV. However, the contribution of each subset in the control of HSV infection has not been clearly defined. This illustrates the controversy regarding the relative roles of each subset in the resolution of HSV infection.To address the role of the CD8+ T-cell subset in providing acquired immunity to HSV infection, we examined the protection afforded by HSV-specific, CD8+ cytotoxic T lymphocytes (CTL) directed to a single CTL recognition epitope. In previous studies by others, immunization with single CTL epitopes has been effective in controlling viral pathogens including lymphocytic choriomeningitis virus (14, 54, 62, 73), murine cytomegalovirus (15), influenza virus (55), and Sendai virus (28). Although HSV-encoded CTL recognition epitopes have been identified by their ability to serve as targets for HSV-specific CTL (3, 8, 24, 64), the ability of CTL directed to these individual epitopes to confer protection against HSV infection has not been determined. We have designed two separate vaccination strategies which permit the exclusive induction of a single HSV epitope-specific, CD8+ T-lymphocyte response and have evaluated the ability of this response to confer protective immunity to HSV infection.Hanke et al. (24) broadly identified an immunodominant, H-2Kb-restricted epitope within HSV glycoprotein B (gB). The minimal amino acid sequence of this epitope, gB498-505 (SSIEFARL), was demonstrated by Bonneau et al. (8), using synthetic peptides and an epitope-specific CTL clone. The amino acid sequence, SSIEFARL, is identical in both HSV type 1 (HSV-1) (gB498-505) and HSV-2 (gB496-503) (11). CTL specific for gB498-505 are readily induced by immunization with synthetic peptide (8), a cell line expressing gB498-505 in the context of simian virus 40 (SV40) T antigen (5), and a recombinant viral vector expressing this epitope in the context of a cellular protein (19). In the present study, two recombinant vaccinia viruses (rVV-ES-gB498-505 and rVV-gB498-505) and a recombinant influenza virus (WSN/NA/gB) were generated to express a single HSV-encoded epitope, HSV-1 gB498-505, and were characterized for the ability to induce a potent, HSV-specific CTL response upon mucosal immunization. To determine the protection afforded by immunization with each of the individual recombinant viruses, we used a lethal model of HSV-2 encephalitis. Our findings suggest that the induction of a CTL response directed against a single HSV-specific CTL recognition epitope is sufficient to confer significant protective immunity to HSV infection.  相似文献   

6.
Following intracerebral infection with Theiler’s murine encephalomyelitis virus (TMEV), susceptible strains of mice (SJL and PLJ) develop virus persistence and demyelination similar to that found in human multiple sclerosis. Resistant strains of mice (C57BL/6) clear virus and do not develop demyelination. To resolve the controversy about the role of CD4+ and CD8+ T cells in the development of demyelination and neurologic deficits in diseases of the central nervous system, we analyzed TMEV infection in CD4- and CD8-deficient B6, PLJ, and SJL mice. Genetic deletion of either CD4 or CD8 from resistant B6 mice resulted in viral persistence and demyelination during the chronic stage of disease. Viral persistence and demyelination were detected in all strains of susceptible background. Although genetic deletion of CD8 had no effect on the extent of demyelination in susceptible strains, deletion of CD4 dramatically increased the degree of demyelination observed. Whereas strains with deletions of CD4 showed severe neurologic deficits, mice with deletions of CD8 showed minimal or no deficits despite demyelination. In all strains, deletion of CD4 but not CD8 resulted in a decreased delayed-type hypersensitivity response to viral antigen. We conclude that each T-cell subset makes a discrete and nonredundant contribution to protection from viral persistence and demyelination in resistant strains. In contrast, in susceptible strains, CD8+ T cells do not provide protection against chronic demyelinating disease. Furthermore, in persistent TMEV infection of the central nervous system, neurologic deficits appear to result either from the absence of a protective class II-restricted immune response or from the presence of a pathogenic class I-restricted response.Multiple sclerosis (MS) is the most common demyelinating disease of the central nervous system (CNS) in humans. MS lesions are characterized by foci of inflammation, myelin destruction, and formation of astrocytic scars known as plaques. The presence of CD4+ T cells, CD8+ T cells (11), and macrophages in lesions suggests that pathogenesis is immunologically mediated; however, the specific contribution of specific cell types remains unknown (12, 44, 45). Although the etiology of MS is unknown, virus infection is the only epidemiological factor consistently associated with clinical exacerbation (43), and beta interferon, a cytokine with multiple known antiviral properties (46), is the only therapeutic agent definitively shown to decrease exacerbation and limit disability in MS (46). Therefore, the study of viral models of demyelination is extremely relevant.Theiler’s murine encephalomyelitis virus (TMEV), a picornavirus, induces a pathological and clinical disease similar to MS (24). Intracerebral infection with the Daniel strain (DA) of TMEV induces transient, acute neuronal polioencephalitis followed by chronic white matter demyelination and neurologic deficits in mice with susceptible (H-2f,p,q,r,s,v) major histocompatibility complex (MHC) haplotypes (15, 32). Mice with resistant (H-2b,d,k) MHC haplotypes recover from the acute disease with no obvious long-term sequelae or demyelination. Although TMEV infection of severely immunodeficient SCID mice results in severe neuronal encephalitis and death within approximately 2 weeks, these mice do not develop demyelination in the spinal cord white matter (38). However, when the immune systems of SCID mice are reconstituted by the adoptive transfer of splenocytes from immunocompetent mice or splenocytes treated with antibodies to CD4 or CD8, infection with TMEV results in chronic demyelination (38). These data indicate that similar to human MS, myelin destruction in chronic TMEV infection is immunologically mediated and requires contributions from both CD4+ and CD8+ T cells.Various reports have implicated both MHC class I- and class II-restricted cells in the pathogenesis of TMEV infection. CD4+ T cells have been implicated by studies demonstrating that demyelination is decreased following treatment with antibodies to CD4 (47) or I-A (34), is increased by adoptive transfer of a CD4+ T-cell line specific for VP2 capsid protein (9), and, in some studies, correlates with the development of a CD4-mediated delayed-type hypersensitivity (DTH) response against virus antigen (5). Furthermore, β2-microglobulin-deficient mice, which are deficient in MHC class I, CD8+ T cells, and natural killer cells, develop demyelinating disease (6, 16, 28). In contrast, a role for CD8+ T cells has been suggested by studies demonstrating that susceptibility to demyelination maps genetically to MHC class I (H-2D) (1, 35), differential expression of MHC class I in the CNS correlates with disease susceptibility (1), and depletion of CD8+ T cells diminishes demyelination (41). Myelin destruction and neurologic deficits develop in TMEV-infected Aβ0 mice which are deficient in functional MHC class II and CD4+ T cells (20). Of interest, both class I and class II-deficient mice share the resistant (H-2b) haplotype. This suggests that although multiple studies have implicated CD4+ and CD8+ T cells in the pathogenesis of TMEV infection, each of these components of the immune response is independently required for maintenance of resistance to demyelination.In order to definitively establish the contribution of CD4+ and CD8+ T cells to demyelination and neurologic deficits, mice lacking surface expression of CD4 or CD8 were backcrossed onto genetically resistant C57BL/6 (H-2b) and susceptible SJL (H-2s) and PLJ (H-2u) strains. In this report, we confirm that both CD4+ and CD8+ T cells are required for protection from viral persistence and demyelination in resistant strains of mice. We also demonstrate that genetic deletion of CD8 does not significantly affect the degree of demyelination or survival in susceptible strains; however, genetic deletion of CD4 greatly increases the degree of demyelination and worsens clinical disease. Of interest, genetic deletion of CD8 greatly reduces neurologic deficits in animals with demyelination.  相似文献   

7.
8.
Although the ability of serum-neutralizing antibodies to protect against picornavirus infection is well established, the contribution of cell-mediated immunity to protection is uncertain. Using major histocompatibility complex class II-deficient (RHAβ−/−) mice, which are unable to mediate CD4+ T-lymphocyte-dependent humoral responses, we demonstrated antibody-independent protection against lethal encephalomyocarditis virus (EMCV) infection in the natural host. The majority of RHAβ−/− mice inoculated with 104 PFU of attenuated Mengo virus (vMC24) resolved infection and were resistant to lethal challenge with the highly virulent, serotypically identical cardiovirus, EMCV. Protection in these mice was in the absence of detectable serum-neutralizing antibodies. Depletion of CD8+ T lymphocytes prior to lethal EMCV challenge ablated protection in vMC24-immunized RHAβ−/− mice. The CD8+ T-lymphocyte-dependent protection observed in vivo may, in part, be the result of cytotoxic T-lymphocyte (CTL) activity, as CD8+ T splenocytes exhibited in vitro cytolysis of EMCV-infected targets. The existence of virus-specific CD8+ T-lymphocyte memory in these mice was demonstrated by increased expression of cell surface activation markers CD25, CD69, CD71, and CTLA-4 following antigen-specific reactivation in vitro. Although recall response in vMC24-immunized RHAβ−/− mice was intact and effectual shortly after immunization, protection abated over time, as only 3 of 10 vMC24-immunized RHAβ−/− mice survived when rechallenged 90 days later. The present study demonstrating CD8+ T-lymphocyte-dependent protection in the absence of serum-neutralizing antibodies, coupled with our previous results indicating that vMC24-specific CD4+ T lymphocytes confer protection against lethal EMCV in the absence of prophylactic antibodies, suggests the existence of nonhumoral protective mechanisms against picornavirus infections.Picornaviruses are a family of positive-strand RNA viruses that are responsible for a variety of devastating human and animal diseases. The family is divided into six genera, enteroviruses, hepatoviruses, parechoviruses, rhinoviruses, aphthoviruses, and cardioviruses, that include such members as poliovirus, human rhinovirus, foot-and-mouth disease virus, and encephalomyocarditis virus (EMCV) (42). Mice are highly susceptible and considered the natural host for cardioviruses such as Mengo virus and EMCV, (7, 35), infections with which result in acute neurotropic disease producing rapid and lethal meningoencephalomyelitis (16, 47). The ability to protect mice against cardiovirus-induced disease by the elicitation or passive transfer of neutralizing antibodies is well documented (2, 13, 26, 41). Current dogma asserts that prophylaxis against picornavirus infection is afforded by serum-neutralizing antibodies (23, 25, 28). Existing picornavirus vaccines (23, 25), in addition to current strategies using recombinant-attenuated and protein-subunit vaccines (27, 32), are designed to elicit a protective neutralizing antibody response to capsid determinants. Indeed, serum-neutralizing titers are used to evaluate host immune status to a particular picornavirus pathogen.Mengo virus and EMCV are members of a single cardiovirus serotype and are indistinguishable by immune sera (42). The dramatic attenuation of Mengo virus by a truncation in the 5′-noncoding-region poly(C) tract preserves complete integrity of all virally encoded proteins (10), allowing in vivo exposure of structural and nonstructural proteins that may elicit an immune response. Normal immunocompetent mice immunized with an attenuated strain of Mengo virus (vMC24) elicit high serum-neutralizing antibody titers and are protected from lethal EMCV challenge (9, 29). In addition to invoking a potent humoral response, vMC24 is also capable of eliciting a cell-mediated immune (CMI) response (29) as an immunodominant CD8+ cytotoxic T-lymphocyte (CTL) epitope has been recently identified in the VP2 capsid protein in vMC24-immunized C57BL/6 mice (11).Earlier investigations of CMI responses to cardioviruses in T-cell deficiency models vacillated between elucidating the immunopathologic role that these cells may contribute in disease and discerning the beneficial aspects that T cells may mediate in protection. T-cell subset depletion of BALB/c mice with anti-CD4 or anti-CD8 antibodies prior to EMCV infection ameliorated clinical disease and reduced the frequency of demyelination (44), suggesting a participatory role for T cells in pathology. Conversely, mice rendered CD4 deficient prior to infection with Theiler’s murine encephalomyelitis virus (TMEV), another murine cardiovirus, failed to produce neutralizing antibodies; consequently, they were unable to clear virus from the central nervous system (CNS) and died from overwhelming encephalitis (49). Similarly, infection of major histocompatibility complex (MHC) class I (β2-microglobulin)-deficient (β2m−/−) mice with TMEV indicates a requisite role for CD8+ T cells in viral clearance and suggests that CD8+ T cells are not major mediators in demyelination or disease (13, 39).More recently, researchers have begun to unveil the beneficial role that CD8+ T cells may have in resolving infection and immune protection. An early and abundant TMEV-specific CD8+ T-cell response is critical in determining the balance between viral persistence or resolution of infection (6, 22, 30). Using vMC24-immunized C57BL/6 mice, Escriou et al identified an immunodominant CD8+ CTL epitope (11) that is cross-reactive to the same VP2 epitope of TMEV (5), although VP2 epitope-immunized C57BL/6 mice were not fully protected from subsequent lethal Mengo virus challenge.The present study is a direct extension of our earlier observation (29) that vMC24-specific CD4+ T cells are capable of adoptively transferring immune protection against lethal EMCV challenge in the absence of prophylactic levels of serum-neutralizing antibodies. Using MHC class II-deficient mice that lack CD4+ T cells and are incapable of T-cell-dependent humoral responses (15), we obtained evidence demonstrating CD8+ T cell-dependent protection against lethal EMCV infection in the absence of serum-neutralizing antibodies.  相似文献   

9.
The murine gammaherpesvirus MHV-68 multiplies in the respiratory epithelium after intranasal inoculation, then spreads to infect B cells in lymphoid germinal centers. Exposing B cells to MHV-68 in vitro caused an increase in cell size, up-regulation of the CD69 activation marker, and immunoglobulin M (IgM) production. The infectious process in vivo was also associated with increased CD69 expression on B cells in the draining lymph nodes and spleen, together with a rise in total serum Ig. However, whereas the in vitro effect on B cells was entirely T-cell independent, evidence of in vivo B-cell activation was minimal in CD4+ T-cell-deficient (I-Ab−/−) or CD4+ T-cell-depleted mice. Furthermore, the Ig present at high levels in serum was predominantly of the IgG class. Surprisingly, the titer of influenza virus-specific serum IgG in previously immunized mice fell following MHV-68 infection, suggesting that there was relatively little activation of memory B cells. Thus, CD4+ T cells seemed both to amplify a direct viral activation of B cells in lymphoid tissue and to promote new Ig class switching despite a lack of obvious cognate antigen.Herpesvirus (HV) infections are often associated with non-antigen-specific B-cell activation (13, 14, 16, 21, 22). Although no definite role has been established for this process in viral pathogenesis, it is of particular interest in gammaherpesvirus (γ-HV) infections, since chronic B-cell stimulation may contribute to the oncogenesis (9, 15) associated with Epstein-Barr virus (EBV) and human herpesvirus 8 (HHV-8) infections. Infection with EBV activates B cells expressing the immunoglobulin (Ig) V4-34 gene (4), which is also overrepresented in certain lymphomas (6, 25). EBV-activated V4-34-expressing B cells can undergo somatic mutation and isotype switching, indicating a participation in normal germinal-center interactions (5). The latent membrane protein 1 (LMP-1) of EBV, which has intracellular signaling substrates similar to those of CD40 (12), and LMP-2A, which can trigger lymphocyte activation (2), may both contribute to this process. However, analysis of lymphocyte interactions in vivo has not been possible with the human γ-HVs.The murine γ-HV-68 (MHV-68) is a natural γ-HV of small rodents that is related to EBV (8) and to HHV-8 (33). After intranasal (i.n.) infection of conventional mice, the virus spreads from the lung to the lymphoid tissue (29) and then persists in B lymphocytes (28) and in epithelial cells (27). This persistent infection is associated with an infectious mononucleosis-like illness (7, 20) characterized by a CD4-dependent splenomegaly and an increase in viral load (31). In BALB/c mice, MHV-68 causes an acute and apparently non-antigen-specific rise in total serum IgG (26). The virus-specific serum antibody response is, in contrast, relatively slow in onset and does not reach plateau levels until 2 to 3 months after infection (26). MHV-68-infected C57BL/6J (B6) mice have more IgG+ cells and fewer IgM+ cells in the spleen (18) than uninfected controls, but to what extent this represents normal immunity is unclear.There is evidence (3) of MHV-68 infection in splenic germinal centers, and both the non-antigen-specific B-cell activation and the CD4-dependent increase in viral load may reflect an exploitation by the virus of normal germinal-center function. The present analysis defines the need, or lack thereof, for CD4+ T-cell help to drive B-cell activation following in vitro or in vivo exposure to MHV-68.  相似文献   

10.
The cytolytic T-lymphocyte (CTL) response to respiratory infection with equine herpesvirus 1 (EHV-1) in CBA (H-2k) mice was investigated. Intranasal (i.n.) inoculation of mice with the attenuated EHV-1 strain KyA resulted in the generation of a primary virus-specific CTL response in the draining mediastinal lymph nodes 5 days following infection. EHV-1-specific CTL could be restimulated from the spleen up to 26 weeks after the resolution of infection, indicating that a long-lived memory CTL population was generated. Depletion of CD8+ T cells by treatment with antibody and complement prior to assay eliminated CTL activity from both primary and memory populations, indicating that cytolytic activity in this model was mediated by class I major histocompatibility complex-restricted, CD8+ T cells. A single i.n. inoculation with KyA induced protective immunity against infection with the pathogenic EHV-1 strain, RacL11. The adoptive transfer of splenocytes from KyA-immune donors into sublethally irradiated recipients resulted in a greater than 250-fold reduction in RacL11 in the lung. The elimination of both CD4+ and CD8+ T cells from the transferred cells abrogated clearance of RacL11, while the selective depletion of either subpopulation alone had little effect. These results suggested that both lymphocyte subpopulations contribute to viral clearance, with either subpopulation alone being sufficient.Equine herpesvirus 1 (EHV-1) is a prevalent respiratory pathogen of horses worldwide (8, 17, 38). Infection of horses with EHV-1 results in fever, respiratory distress, abortagenic disease, and severe neurological sequelae (3, 13, 27, 35, 36). The highly contagious respiratory transmission of EHV-1 has resulted in disastrous outbreaks of disease in domestic horse populations and has had a significant economic impact on the equine industry. EHV-1 infection of the horse results in the generation of a short-lived humoral response but does not confer long-term protection, as disease often occurs following natural infection (10, 22). Although both live and inactivated vaccines are currently available for EHV-1, only relatively short-lived protection has been observed (11, 12, 24). Furthermore, it is not clear which immune functions are responsible for conferring the short-lived protection following vaccination. In addition to specific antibody responses, peripheral blood leukocytes from vaccinated horses produce gamma interferon in culture (19). EHV-1-specific, CD8+ class I major histocompatibility complex (MHC)-restricted cytotoxic T lymphocytes (CTL) have been identified in peripheral blood mononuclear cells, reaching maximal levels 2 to 3 weeks postinfection (p.i.) (4, 20). However, the effectiveness of the current vaccines in stimulating EHV-1-specific CTL and their role in protective immunity in vivo are currently not known.Horses inoculated with the attenuated EHV-1 strain Kentucky A (KyA) exhibited a reduction in clinical signs following challenge with a pathogenic EHV-1 strain (33, 34). Although the attenuated strain induced a protective response, in terms of a reduced duration of viral shedding and viremia, the ability of this strain to induce an EHV-1-specific antibody response was weaker than that of the virulent strain. This finding suggested that immune functions other than the generation of specific antibodies might be critical in the resolution of infection. To generate a more effective EHV-1 vaccine, a better understanding of the precise immune functions associated with protection and resolution from EHV-1 infection is essential.A murine model of respiratory EHV-1 infection which closely mimicked EHV-1 infection in the natural host was established in various strains of mice (5). Common features included replication in the respiratory mucosae, the development of pneumonitis, cell-associated viremia, and abortion (5, 6). Specific immune responses are important for modulating infection. In the mouse, the passive transfer of hyperimmune polyclonal rabbit EHV-1-specific antibodies into infected mice significantly reduced the viremia following challenge with live EHV-1 (5). Subsequent studies demonstrated that various EHV-1 glycoproteins, including gB, gC, gD and gH, were capable of inducing the generation of neutralizing antibodies (9, 23, 40, 49, 52, 56). Furthermore, inoculation of mice with gB, gC, and/or gD elicited a protective response against subsequent challenge with pathogenic EHV-1 (39, 49, 50, 52, 56). However, each of these EHV-1 gene products is capable of eliciting both B- and T-cell responses; thus, the role of distinct immune functions conferring protection is not clearly defined.In the most extensively studied BALB/c mouse model of EHV-1 infection, adoptive transfer experiments demonstrated that immune spleen cells isolated from mice primed with live, but not heat-killed, EHV-1 reduced viral levels in both the lungs and nasal turbinates of infected recipient mice (5). Although the specific cell population responsible for protection was not determined, the data suggested that cell-mediated immune functions may be critical in the resolution of EHV-1 infection. The adoptive transfer of defined T-cell subpopulations demonstrated that both CD4+ and CD8+ T cells play a role in controlling EHV-1 respiratory infection. The CD8+ T-cell subpopulation appeared to play a more dominant role, although the functions by which protection was mediated were not defined (7). If the immune response was elicited by immunization with recombinant baculovirus-derived EHV-1 glycoproteins, the response was altered so that CD4+ T cells were predominantly associated with protection (52). Thus, either T-cell subpopulation is likely to play an important role in the optimal response to infection.While adoptive transfer studies have implicated an important role for CD8+ T cells in the control of EHV-1 infection in the lungs of BALB/c mice (7), there has been no direct assessment of CD8+ T-cell effector functions in this model. This is due predominantly to the lack of suitable class I MHC-compatible, H-2d-expressing murine cells that are susceptible to infection with EHV-1. However, the attenuated KyA strain of EHV-1 has been propagated in our laboratory in suspension cultures of murine L-M fibroblasts that express the H-2k haplotype. Therefore, an alternative mouse model, using mice expressing the H-2k haplotype, was adopted principally to assess the activation of EHV-1-specific CTL responses. Initial studies by Awan et al. (5) demonstrated that CBA (H-2k) mice were susceptible to infection with EHV-1 strain Ab4. In the present study, CBA mice were susceptible to respiratory tract infection by both the nonpathogenic KyA and the pathogenic RacL11 strains of EHV-1. Furthermore, infection of CBA mice with the attenuated KyA strain generated a vigorous CD8+, class I MHC-restricted, EHV-1-specific primary CTL response in the draining mediastinal lymph nodes (MLN) and a long-term memory CTL response in the spleen. These studies provide the basis for a model system to analyze the potential importance of class I MHC-restricted CTL activity in controlling EHV-1 infection in vivo.  相似文献   

11.
DNA vaccination is an effective means of eliciting both humoral and cellular immunity, including cytotoxic T lymphocytes (CTL). Using an influenza virus model, we previously demonstrated that injection of DNA encoding influenza virus nucleoprotein (NP) induced major histocompatibility complex class I-restricted CTL and cross-strain protection from lethal virus challenge in mice (J. B. Ulmer et al., Science 259:1745–1749, 1993). In the present study, we have characterized in more detail the cellular immune responses induced by NP DNA, which included robust lymphoproliferation and Th1-type cytokine secretion (high levels of gamma interferon and interleukin-2 [IL-2], with little IL-4 or IL-10) in response to antigen-specific restimulation of splenocytes in vitro. These responses were mediated by CD4+ T cells, as shown by in vitro depletion of T-cell subsets. Taken together, these results indicate that immunization with NP DNA primes both cytolytic CD8+ T cells and cytokine-secreting CD4+ T cells. Further, we demonstrate by adoptive transfer and in vivo depletion of T-cell subsets that both of these types of T cells act as effectors in protective immunity against influenza virus challenge conferred by NP DNA.Cellular immune responses play an important role in protection from disease caused by infectious pathogens, such as viruses and certain bacteria (e.g., Mycobacterium tuberculosis). The specific T cells involved in conferring immunity can include both CD4+ and CD8+ T cells, often through the action of secreted cytokines and cytolytic activity, respectively. Certain types of vaccines, such as subunit proteins and whole or partially purified preparations of inactivated organisms, in general induce CD4+ T-cell responses but not CD8+ cytotoxic T lymphocytes (CTL). In contrast, live attenuated organisms and subunit proteins formulated with certain experimental adjuvants can induce both types of responses. Recently, a different approach consisting of direct immunization with plasmid DNA expression vectors (i.e., DNA vaccines) has shown promise as a viable means of inducing broad-spectrum T-cell responses. The effectiveness of DNA vaccines in animal models is likely due, at least in part, to expression of antigens in situ (35), leading to the induction of CTL (29), antibodies (3, 4, 10, 21, 22, 32), and cytokine-secreting lymphocyte responses (12, 36). During the past 5 years, many reports have been published on the immunogenicity of DNA vaccines encoding various antigens in several animal models, thereby illustrating the applicability of the technology to many pathogens (for a review, see reference 6). However, in only a few instances has the nature of the effector cells responsible for protective immunity been described (7, 16). In the present study, we have analyzed in detail the cellular immune responses induced by influenza virus nucleoprotein (NP) DNA and have established that both CD4+ T cells secreting Th1-type cytokines and CD8+ cytotoxic T cells play important effector roles in heterosubtypic protective immunity against lethal influenza virus challenge in mice.  相似文献   

12.
13.
We examined the phenotype and function of cells infiltrating the central nervous system (CNS) of mice persistently infected with Theiler’s murine encephalomyelitis virus (TMEV) for evidence that viral antigens are presented to T cells within the CNS. Expression of major histocompatibility complex (MHC) class II in the spinal cords of mice infected with TMEV was found predominantly on macrophages in demyelinating lesions. The distribution of I-As staining overlapped that of the macrophage marker sialoadhesin in frozen sections and coincided with that of another macrophage/microglial cell marker, F4/80, by flow cytometry. In contrast, astrocytes, identified by staining with glial fibrillary acidic protein, rarely expressed detectable MHC class II, although fibrillary gliosis associated with the CNS damage was clearly seen. The costimulatory molecules B7-1 and B7-2 were expressed on the surface of most MHC class II-positive cells in the CNS, at levels exceeding those found in the spleens of the infected mice. Immunohistochemistry revealed that B7-1 and B7-2 colocalized on large F4/80+ macrophages/microglia in the spinal cord lesions. In contrast, CD4+ T cells in the lesions expressed mainly B7-2, which was found primarily on blastoid CD4+ T cells located toward the periphery of the lesions. Most interestingly, plastic-adherent cells freshly isolated from the spinal cords of TMEV-infected mice were able to process and present TMEV and horse myoglobin to antigen-specific T-cell lines. Furthermore, these cells were able to activate a TMEV epitope-specific T-cell line in the absence of added antigen, providing conclusive evidence for the endogenous processing and presentation of virus epitopes within the CNS of persistently infected SJL/J mice.Theiler’s murine encephalomyelitis virus (TMEV) is a picornavirus that induces a lifelong persistent central nervous system (CNS) infection leading to a chronic CNS demyelinating disease when inoculated intracerebrally into susceptible strains of mice. Infected mice develop progressive symptoms of gait disturbance, spastic hind limb paralysis, and urinary incontinence (39), histologically related to perivascular and parenchymal mononuclear cell infiltration and demyelination of white matter tracts within the spinal cord (8, 9, 38). Several lines of evidence have demonstrated that demyelination is immunologically mediated. These include the ability of nonspecific immunosuppression with cyclophosphamide (37), antithymocyte serum (36), and anti-CD4 or anti-major histocompatibility complex (MHC) class II monoclonal antibodies (MAbs) (14, 16, 63) to inhibit or prevent disease and the ability of TMEV-specific tolerance to prevent induction of disease (28). In the highly susceptible SJL/J mouse strain, current evidence indicates that the myelin damage is initiated by TMEV-specific CD4+ T cells targeting virus antigen (16, 28, 45, 46, 54), while the chronic stage of the disease also involves CD4+ myelin epitope-specific T cells primed via epitope spreading (48). Thus, the immune response itself may be deleterious to CNS function, as exemplified in humans by multiple sclerosis (MS), for which TMEV infection serves as a model.The identity of the cells responsible for initiating and sustaining immune responses in the CNS remains controversial. The CNS lacks normal lymphatic circulation and tissue and is shielded from the systemic circulation by a specialized continuous vascular endothelium (6). There are specialized cells within the CNS with the potential to present antigens to T cells. In vitro, astrocytes (11, 59) and microglia (3, 13), particularly when treated with gamma interferon (IFN-γ), are capable of expressing MHC class II and presenting antigens to T cells. However, studies such as these have relied on the ability to isolate and continuously culture cells from neonatal or embryonic brain and have assumed that such cells are representative of the adult populations in vivo. Antigen presentation by neonatal cells in long-term culture may not faithfully reproduce the in vivo state in adult animals, as the ability of microglia directly isolated from adult rats to present myelin basic protein (MBP) to T-cell lines in vitro was found to differ from that of neonatally derived microglia (12). In addition, studies using allogeneic bone marrow chimeras between strains of mice or rats have generally supported the idea that cells of hematopoietic origin, i.e., microglia and macrophages, are the principal antigen-presenting cells (APCs) in the CNS active during the initiation of experimental autoimmune encephalomyelitis (EAE) (20, 22, 50). Although they are much more abundant than microglia, astrocytes are less potent when inducing EAE in chimeras (50).The role of antigen presentation in the CNS during TMEV-induced demyelination has not been addressed directly. We previously showed that a relatively large fraction of the CD4+, but not CD8+, T cells isolated from the spinal cords of TMEV-infected mice expressed high-affinity interleukin-2 (IL-2) receptor (IL-2R), a marker of recent T-cell activation. In addition, TMEV-specific CD4+ T cells could be demonstrated in the spinal cord infiltrates of TMEV-infected mice (54). This finding raises the possibility that T cells are locally activated within the target tissue and participate directly in the pathogenesis of disease. Macrophages (5, 41, 56), astrocytes (7, 56), and oligodendroglia (55, 56) in TMEV-infected mice contain virus and conceivably could present viral antigens to pathogenic CD4+ T cells within the CNS. Isolated microglia (34) and astrocytes (17) have been shown to support persistent viral infection in vitro, and astrocytes derived from neonatal mice have been shown to present TMEV to T cells in vitro (2). To examine whether CNS cells present viral antigens and participate in the pathogenesis of TMEV-induced demyelination, the expression of MHC class II and B7 costimulatory molecules was examined in detail. Based on our previous results showing that a large proportion of CD4+ T cells isolated from the CNS of TMEV-infected mice bear markers of recent activation, we also asked if mononuclear cells isolated from the CNS of TMEV-infected mice were capable of presenting viral antigens leading to the functional activation of Th1 lines in vitro.  相似文献   

14.
Mouse cytotoxic T lymphocytes (CTL) reactive with a H-2Db-presented 9-mer peptide of the human papillomavirus type 16 protein E749-57 (RAHYNIVTF) were generated from the spleen cells of wild-type C57BL/6 (B6) or B6 perforin-deficient (B6.P0) mice. CD8+ B6 CTL displayed peptide-specific perforin- and Fas-mediated lysis of E7-transfected mouse RMA lymphoma cells (RMA-E7), while CD8+ CTL from B6.P0 mice lysed RMA-E7 cells via Fas ligand (FasL) exclusively. Rapid and efficient lysis of syngeneic bystander B6 blasts or RMA cells by either B6 or B6.P0 Ag-activated CTL was mediated by a FasL-Fas mechanism. Fas-resistant bystanders were not lysed, nor were allogeneic Fas-sensitive C3H/HeJ (H-2k) or BALB/c (H-2d) bystander blasts. Interestingly, however, phorbol myristate acetate-ionomycin preactivation of B6.P0 effectors enabled lysis of allogeneic H-2k and H-2d bystanders even in the absence of antigenic stimulation. Lysis of syngeneic bystander cells was always FasL-Fas dependent and required effector-bystander contact and, in particular, an interaction between CTL LFA-1 and bystander ICAM-1. Thus, in the context of major histocompatibility complex class I molecule-peptide ligation of the T-cell receptors of CD8+ CTL, neighboring bystander cells that are syngeneic and Fas sensitive and express the adhesion molecule ICAM-1 are potential targets of CTL attack.With the dissection of two basic cytolytic mechanisms of cytotoxic T lymphocytes (CTL) (10, 14, 20, 34), it has become possible to delineate the important criteria that determine direct (Ag-restricted) and bystander cytotoxicity. CTL use complementary cytotoxic mechanisms, one based on the granule exocytosis of a calcium-dependent pore-forming protein, perforin (8, 26), and granzymes (35) and another that depends on a calcium-independent interaction of effector T-cell tumor necrosis factor or Fas ligand (TNF or FasL) and target cell TNF receptor (TNFR) or Fas (22, 33). The function of the granule exocytosis pathway appears to be largely in non-major histocompatibility complex (MHC)-restricted NK lysis of class I molecule-defective tumor cells and in direct CTL-mediated immunity against tumor cells (37) or virus-infected cells (11, 19, 39). By contrast, the FasL-Fas and TNF-TNFR interactions are important for the maintenance of T-cell homeostasis following exposure to foreign Ag (5, 42) and Th-1 FasL-mediated B-cell apoptosis (27, 28). Blockage of both TNF and FasL is required to abrogate T-cell death: TNF mediates the death of most CD8+ T cells, whereas FasL mediates the death of most CD4+ T cells (42). While FasL-dependent lysis appears to be the primary mechanism used by CD4+ Th-1 effectors, CD8+ CTL use FasL or TNF secondarily in the absence of perforin-mediated lysis (10, 14, 20).After T-cell activation, a functional role for FasL is not apparent for several days until the T cell becomes Fas sensitive and hence susceptible to autocrine T-cell suicide (1, 5, 38). However, by using alloreactive CTL cultures or clones, it has recently become apparent that in the presence of Ag-bearing target cells (i.e., upon T-cell receptor [TCR] activation) CTL can also lyse Ag-free bystander cells via a FasL-Fas interaction (13, 34). While the specificity of CTL toward Ag-bearing target cells has been considered a hallmark of an efficient immune response, CTL do not appear to spare Ag-free bystander cells during lysis of specific Ag-bearing target cells. In this study, we have generated CD8+ CTL from both wild-type and perforin-deficient (P0) mice reactive with a high-affinity H-2Db-binding peptide of human papillomavirus type 16 protein E7. These peptide-specific CTL have been employed to demonstrate the requirements for CD8+ CTL-mediated lysis of Ag-free bystander cells and in particular the different properties of CTL activated by antigen versus a nonspecific stimulus.  相似文献   

15.
Human immunodeficiency virus infection is characterized by a progressive decline in the number of peripheral blood CD4+ T lymphocytes, which finally leads to AIDS. This T-cell decline correlates with the degree of in vitro-induced lymphocyte apoptosis. However, such a correlation has not yet been described in feline AIDS, caused by feline immunodeficiency virus (FIV) infection. We therefore investigated the intensity of in vitro-induced apoptosis in peripheral blood lymphocytes from cats experimentally infected with a Swiss isolate of FIV for 1 year and for 6 years and from a number of long-term FIV-infected cats which were coinfected with feline leukemia virus. Purified peripheral blood lymphocytes were either cultured overnight under nonstimulating conditions or stimulated with phytohemagglutinin and interleukin-2 for 60 h. Under stimulating conditions, the isolates from the infected cats showed significantly higher relative counts of apoptotic cells than did those from noninfected controls (1-year-infected cats, P = 0.01; 6-year-infected cats, P = 0.006). The frequency of in vitro-induced apoptosis was inversely correlated with the CD4+ cell count (P = 0.002), bright CD8+ cell count (P = 0.009), and CD4/CD8 ratio (P = 0.01) and directly correlated with the percentage of bright major histocompatibility complex class II-positive peripheral blood lymphocytes (P = 0.004). However, we found no correlation between in vitro-induced apoptosis and the viral load in serum samples. Coinfection with feline leukemia virus enhanced the degree of in vitro-induced apoptosis compared with that in FIV monoinfected cats. We concluded that the degree of in vitro-induced apoptosis was closely related to FIV-mediated T-cell depletion and lymphocyte activation and could be used as an additional marker for disease progression in FIV infection.Feline immunodeficiency virus (FIV) infection is a naturally occurring infection, and disease progression in infected cats is associated with a decline in the number of CD4+ cells (2, 6, 22, 23, 36), a loss of bright CD8+ cells in the advanced stage of the disease (22), an increased number of activated T cells (39, 41), and a changed cytokine production, i.e., decreased production of interleukin-2 (IL-2) and concomitantly increased production of tumor necrosis factor alpha (TNF-α) (25, 26). The increased production of TNF-α has been reported to induce apoptosis in chronically FIV-infected cells (38). Apoptosis, a controlled mode of cell death (34), plays an important role in the regulation of immune responses (5, 14). As described for FIV (23), the hallmark of human immunodeficiency virus (HIV)-induced disease is the loss of T-helper cells (31, 43). Theoretically, cell loss can be caused by decreased production of cells, increased destruction, or a combination of the two mechanisms. Findings of an early infection of thymocytes followed by pathologic changes in the thymus support the model of decreased T-helper cell production triggered by HIV (13) and FIV (52). The destruction model is supported by findings of an increased number of peripheral blood T cells undergoing apoptosis upon HIV (20, 32) and FIV (11, 21, 33) infection. However, increased CD4+-T-cell turnover may not be the main cause of the observed T-helper cell decline in HIV-1 infection, as reviewed by others (44, 51). In addition, the degree of HIV-induced apoptosis correlates with the T-helper cell decline and disease progression (19, 40). However, such a relationship has not yet been described for FIV. It has been reported that cross-linking of CD4 molecules by HIV gp160 triggers apoptosis in noninfected CD4+ T cells (1). Investigation of this aspect in the feline system is especially interesting since FIV does not use the feline homologue of CD4 (50).The aim of the present study was to compare the degree of in vitro-induced lymphocyte apoptosis in FIV-infected cats with normal and decreased T-helper cell counts. We used two different culture conditions to trigger apoptosis in vitro: cells were either cultured overnight under nonstimulating conditions and in the absence of growth factors or cultured for 60 h in the presence of phytohemagglutinin, IL-2, and fetal calf serum. We additionally examined cats which were coinfected with the feline leukemia virus (FeLV). This coinfection is known to accelerate the progression toward feline AIDS (23) by an unknown mechanism (8).  相似文献   

16.
Most studies of human immunodeficiency virus type 1 (HIV-1)-specific cytotoxic T lymphocytes (CTL) have been confined to the evaluation of these effector cells in the peripheral blood. What has not been clear is the extent to which CTL activity in the blood actually reflects this effector cell function in the lymph nodes, the major sites of HIV-1 replication. To determine the concordance between CTL activity in lymph nodes and peripheral blood lymphocytes (PBL), CTL specific for simian immunodeficiency virus of macaques (SIVmac) have been characterized in lymph nodes of infected, genetically selected rhesus monkeys by using both Gag peptide-specific functional CTL assays and tetrameric peptide-major histocompatibility complex (MHC) class I molecule complex staining techniques. In studies of six chronically SIVmac-infected rhesus monkeys, Gag epitope-specific functional lytic activity and specific tetrameric peptide-MHC class I staining were readily demonstrated in lymph node T lymphocytes. Although the numbers of tetramer-binding cells in some animals differed from those documented in their PBL, the numbers of tetramer-binding cells from these two different compartments were not statistically different. Phenotypic characterization of the tetramer-binding CD8+ lymph node T lymphocytes of the infected monkeys demonstrated a high level of expression of the activation-associated adhesion molecules CD11a and CD49d, the Fas molecule CD95, and MHC class II-DR. These studies documented a low expression of the naive T-cell marker CD45RA and the adhesion molecule CD62L. This phenotypic profile of the tetramer-binding lymph node CD8+ T cells was similar to that of tetramer-binding CD8+ T cells from PBL. These observations suggest that characterization of AIDS virus-specific CTL activity by sampling of cells in the peripheral blood should provide a reasonable estimation of CTL in an individual’s secondary lymphoid tissue.CD8+ cytotoxic T lymphocytes (CTL) are important in containing the spread of human immunodeficiency virus type 1 (HIV-1) in infected individuals. Studies have shown that virus-specific CD8+ CTL can inhibit AIDS virus replication in autologous CD4+ T lymphocytes in vitro, probably by release of chemokines and cytokines, as well as by lysis of infected cells (35, 36). In vivo the containment of HIV-1 replication that occurs during the period of primary infection coincides temporally with the generation of virus-specific CTL (8, 17, 29). Finally, a potent CTL response is correlated with low virus load and a stable clinical status in individuals chronically infected with HIV-1 (25, 27).HIV-1 replication occurs predominantly in the lymph nodes of the infected individual (30). However, most studies of HIV-1-specific CTL have been confined to the evaluation of these effector cells in the peripheral blood. It is not clear to what extent CTL activity in the blood actually reflects this effector cell function at the major sites of HIV-1 replication. An extensive evaluation of CTL in lymph nodes of HIV-1-infected humans has not been undertaken, at least in part because of the numerous surgical procedures that would be required for such a study. The use of such procedures in clinically stable individuals might be difficult to rationalize.The simian immunodeficiency virus (SIV)-infected macaque provides an ideal animal model in which to examine AIDS virus-specific CTL in lymph nodes. SIVmac-infected rhesus monkeys develop a disease with remarkable similarities to HIV-1-induced disease in humans (19, 20). SIVmac-specific CTL are readily detected in infected monkeys by functional killing assays (21, 38). We have made use of a dominant CTL response to the SIVmac Gag epitope p11C, C-M in rhesus monkeys expressing the major histocompatibility complex (MHC) class I molecule Mamu-A*01 to explore the role of CTL in the immunopathogenesis of AIDS (1, 22). In the present study, CTL specific for SIVmac have been characterized in lymph nodes of infected, Mamu-A*01+ rhesus monkeys using both Gag peptide-specific functional CTL assays and tetrameric peptide-MHC class I molecule complex staining techniques (2, 6, 12, 18, 24, 27).  相似文献   

17.
18.
19.
20.
Human cytomegalovirus (HCMV) infection can be fatal to immunocompromised individuals. We have previously reported that gamma interferon and tumor necrosis factor alpha (TNF-α) synergistically inhibit HCMV replication in vitro. Ceramides have been described as second messengers induced by TNF-α. To investigate the mechanisms involved in the inhibition of HCMV by TNF-α, in the present study we have analyzed ceramide production by U373 MG astrocytoma cells and the effects of TNF-α versus ceramides on HCMV replication. Our results show that U373 MG cells did not produce ceramides upon incubation with TNF-α. Moreover, long-chain ceramides induced by treatment with exogenous bacterial sphingomyelinase inhibited HCMV replication in synergy with TNF-α. Surprisingly, short-chain permeant C6-ceramide increased viral replication. Our results show that the anti-HCMV activity of TNF-α is independent of ceramides. In addition, our results suggest that TNF-α and endogenous long-chain ceramides use separate pathways of cell signalling to inhibit HCMV replication, while permeant C6-ceramide appears to activate a third pathway leading to an opposite effect.Human cytomegalovirus (HCMV) infections are well controlled in the immunocompetent host. Cellular immune responses (CD4+ and CD8+ T cells and NK cells) which accompany both acute and latent infections (for a review, see reference 4) are thought to be the main components of this control. HCMV infection during immunosuppression such as in cancer, transplantations, or AIDS results in severe pathology (4). We have previously shown that tumor necrosis factor alpha (TNF-α), in synergy with gamma interferon (IFN-γ), inhibits the replication of HCMV (7). In mice, TNF-α is involved in the clearance of CMV infection (25). TNF-α is a cytokine with multiple effects which is produced by many cell types, including macrophages and CD8+ and CD4+ T lymphocytes (for a review, see reference 40), and is known to possess antiviral effects (20, 47). The molecular mechanisms involved in the signalling by TNF-α depend on the type of receptor, p55 (TNF-R1) or p75 (TNF-R2) (5), to which it binds. Some cells express only one type of TNF-α receptor; however, expression of these receptors is not always mutually exclusive (5). The cytotoxicity of TNF-α has been reported to be mediated by TNF-R1 (38), whose intracellular region carries a death domain which signals for programmed cell death (39). Signalling through TNF-R1 with specific antibodies can also protect Hep-G2 cells from vesicular stomatitis virus-mediated cytopathic effects (48). Ceramide production after TNF-α treatment has been widely reported (16, 19, 31) and has also been shown to depend on signalling through the TNF-R1 receptor (45). In these experiments concerning myeloid cells, TNF-α induced the activation of a sphingomyelinase, which cleaved sphingomyelin to release ceramide and phosphocholine. The production of ceramides can lead to cell apoptosis (11, 14, 23) or cell cycle arrest (13). Induction of apoptosis by TNF-α has been mimicked by exogenous sphingomyelinase and by synthetic, short-chain, permeant ceramides, which suggests that ceramides, as second messengers, are sufficient to induce the cytotoxic effects of TNF-α (11, 23). Acidic and neutral sphingomyelinases activated in different cell compartments may be responsible for the diverse effects of TNF-α (46), with the former being involved in signalling through NF-κB (34) and the latter being involved in signalling through a ceramide-activated protein kinase and phospholipase A2 (46).One of the characteristics of HCMV infection is the increase in the content of intracellular DNA, reported to be of viral (3, 8, 18) or cellular (12, 37) origin. Since TNF-α has been known to display antiproliferative properties and to block cells in the G1 phase (29), we initially tested its effects on the cell cycle of infected cells. Then, based on studies reporting that TNF-α induces ceramides in cells (16, 19, 31) and on a study showing the role of ceramide in cell cycle blockade (13), we originally postulated that ceramide was responsible for the antiviral effect of TNF-α. In the present study, we used astrocytoma cells (U373 MG) as a model for brain cells, which are important targets of HCMV in vivo (22). In contrast to fibroblasts, infected U373 MG cells release smaller quantities of virus particles even though all the cells were infected in our experiments. We believe that the U373 MG model is closer to HCMV infection in vivo. We show that ceramides are not produced by U373 MG cells upon incubation with even high concentrations of TNF-α. In addition, we demonstrate that exogenously added sphingomyelinase induces anti-HCMV effects whereas permeant C6-ceramide increases HCMV proliferation in U373 MG cells. This suggests that lipid second messengers can modulate HCMV infection and that TNF-α and ceramides use distinct signalling pathways in the control of HCMV infection. This is supported by our observation that the protein kinase JNK1 is activated exclusively by TNF-α in U373 MG cells and that TNF-α and exogenous sphingomyelinase act in synergy on HCMV infection.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号