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 15 毫秒
1.
Tumor-infiltrating dendritic cells are often ineffective at presenting tumor-derived antigen in vivo, a defect usually ascribed to the suppressive tumor environment. We investigated the effects of depleting CD4+CD25+ “natural” regulatory T cells (Treg) on the frequency, phenotype and function of total dendritic cell populations in B16.OVA tumors and in tumor-draining lymph nodes. Intraperitoneal injection of the anti-CD25 monoclonal antibody PC61 reduced Treg frequency in blood and tumors, but did not affect the frequency of tumor-infiltrating dendritic cells, or their expression of CD40, CD86 and MHCII. Tumor-infiltrating dendritic cells from PC61-treated or untreated mice induced the proliferation of allogeneic T cells in vitro, but could not induce proliferation of OVA-specific OTI and OTII T cells unless specific peptide antigen was added in culture. Some proliferation of naïve, OVA-specific OTI T cells, but not OTII T cells, was observed in the tumor-draining LN of mice carrying B16.OVA tumors, however, this was not improved by PC61 treatment. Experiments using RAG1−/− hosts adoptively transferred with OTI and CD25-depleted OTII cells also failed to show improved OTI and OTII T cell proliferation in vivo compared to C57BL/6 hosts. We conclude that the defective presentation of B16.OVA tumor antigen by tumor-infiltrating dendritic cells and in the tumor-draining lymph node is not due to the presence of “natural” CD4+CD25+ Treg.  相似文献   

2.
Stark FC  Gurnani K  Sad S  Krishnan L 《PloS one》2012,7(2):e32211
Central memory CD8+ T cells expressing the adhesion molecule CD62L (L-selectin) are potent mediators of anti-cancer immunity due to their ability to proliferate extensively upon antigen re-stimulation. The interaction of selectin with its ligands mediates leukocyte rolling along high endothelial venules. Mice deficient in α(1,3) Fucosyltransferase IV and VII (FtDKO) lack functional L, P and E selectin ligands. Thus, we addressed whether the lack of selectin ligand interactions alters tumor protection by CD8+ T cells in FtDKO mice. Listeria monocytogenes-OVA (LM-OVA) infection evoked potent OVA-specific CD8+ T cells that proliferated and contracted at similar kinetics and phenotype in FtDKO and wild-type mice. Additionally, OVA-specific CD8+ T cells in both mouse strains exhibited similar phenotypic differentiation, in vivo cytolytic activity and IFN-γ expression. However, FtDKO mice succumbed to B16-OVA tumors significantly earlier than wild-type mice. In contrast, FtDKO mice evoked strong recall memory CD8+ T cell responses and protection to systemic LM-OVA re-challenge. The diminished tumor protection in FtDKO mice was not related to defective antigen presentation by dendritic cells or reduced proliferation of antigen-specific CD8+ T cells. However, WT or FtDKO OVA-specific CD8+ T cells showed significantly reduced ability to traffic to lymph nodes upon adoptive transfer into naïve FtDKO recipients. Furthermore, FtDKO OVA-specific CD8+ T cells displayed poor ability to infiltrate tumors growing in WT mice. These results reveal that selectin ligand expression on host endothelium as well CD8+ T cells may be important for their efficient and continued extravasation into peripheral tumors.  相似文献   

3.

Background

In contrast to intestinal CD4+ regulatory T cells (Tregs), the generation and function of immunomodulatory intestinal CD8+ T cells is less well defined. To dissect the immunologic mechanisms of CD8+ T cell function in the mucosa, reactivity against hemagglutinin (HA) expressed in intestinal epithelial cells of mice bearing a MHC class-I-restricted T-cell-receptor specific for HA was studied.

Methodology and Principal Findings

HA-specific CD8+ T cells were isolated from gut-associated tissues and phenotypically and functionally characterized for the expression of Foxp3+ and their suppressive capacity. We demonstrate that intestinal HA expression led to peripheral induction of HA-specific CD8+Foxp3+ T cells. Antigen-experienced CD8+ T cells in this transgenic mouse model suppressed the proliferation of CD8+ and CD4+ T cells in vitro. Gene expression analysis of suppressive HA-specific CD8+ T cells revealed a specific up-regulation of CD103, Nrp1, Tnfrsf9 and Pdcd1, molecules also expressed on CD4+ Treg subsets. Finally, gut-associated dendritic cells were able to induce HA-specific CD8+Foxp3+ T cells.

Conclusion and Significance

We demonstrate that gut specific antigen presentation is sufficient to induce CD8+ Tregs in vivo which may maintain intestinal homeostasis by down-modulating effector functions of T cells.  相似文献   

4.
CD4+ helper T cells are critical orchestrators of immune responses to infection and vaccination. During primary responses, naïve CD8+ T cells may need “CD4 help” for optimal development of memory populations. The immunological factors attributed to CD4 help depend on the context of immunization and vary depending on the priming system. In response to immunization with radiation-attenuated Plasmodium yoelii sporozoites, CD8+ T cells in BALB/c mice fail to generate large numbers of effector cells without help from CD4+ T cells – a defect not observed in most systems. Given this unique early dependence on CD4 help, we evaluated the effects of CD4+ cells on the development of functional properties of CD8+ T cells and on their ability to abolish infection. First, we determined that this effect was not mediated by CD4+ non-T cells and did not involve CD1d-restricted NKT cells. We found that CD8+ T cells induced by sporozoites without CD4 help formed memory populations severely reduced in magnitude that could not limit parasite development in the liver. The inability of these “helpless” memory T cells to protect is not a result of defects in effector function, as their capacity to produce cytokines and undergo cytotoxic degranulation was indistinguishable from control memory T cells. These data indicate that CD4+ T help may not be necessary to develop the functional attributes of CD8+ T cells; however they are crucial to ensure the survival of effector and memory cells induced in primary responses.  相似文献   

5.
MHC class Ia-restricted CD8+ T cells are important mediators of the adaptive immune response against infections caused by intracellular microorganisms. Whereas antigen-specific effector CD8+ T cells can clear infection caused by intracellular pathogens, in some circumstances, the immune response is suboptimal and the microorganisms survive, causing host death or chronic infection. Here, we explored the cellular and molecular mechanisms that could explain why CD8+ T cell-mediated immunity during infection with the human protozoan parasite Trypanosoma cruzi is not optimal. For that purpose, we compared the CD8+ T-cell mediated immune responses in mice infected with T. cruzi or vaccinated with a recombinant adenovirus expressing an immunodominant parasite antigen. Several functional and phenotypic characteristics of specific CD8+ T cells overlapped. Among few exceptions was an accelerated expansion of the immune response in adenoviral vaccinated mice when compared to infected ones. Also, there was an upregulated expression of the apoptotic-signaling receptor CD95 on the surface of specific T cells from infected mice, which was not observed in the case of adenoviral-vaccinated mice. Most importantly, adenoviral vaccine provided at the time of infection significantly reduced the upregulation of CD95 expression and the proapoptotic phenotype of pathogen-specific CD8+ cells expanded during infection. In parallel, infected adenovirus-vaccinated mice had a stronger CD8 T-cell mediated immune response and survived an otherwise lethal infection. We concluded that a suboptimal CD8+ T-cell response is associated with an upregulation of CD95 expression and a proapoptotic phenotype. Both can be blocked by adenoviral vaccination.  相似文献   

6.

Background

Cooperation of CD4+ T helper cells with specific B cells is crucial for protective vaccination against pathogens by inducing long-lived neutralizing antibody responses. During infection with persistence-prone viruses, prolonged virus replication correlates with low neutralizing antibody responses. We recently described that a viral mutant of lymphocytic choriomeningitis virus (LCMV), which lacks a T helper epitope, counterintuitively induced an enhanced protective antibody response. Likewise, partial depletion of the CD4+ T cell compartment by using anti-CD4 antibodies enhanced protective antibodies.

Principal Findings

Here we have developed a protocol to selectively reduce the CD4+ T cell response against viral CD4+ T cell epitopes. We demonstrate that in vivo treatment with LCMV-derived MHC-II peptides induced non-responsiveness of specific CD4+ T cells without affecting CD4+ T cell reactivity towards other antigens. This was associated with accelerated virus-specific neutralizing IgG-antibody responses. In contrast to a complete absence of CD4+ T cell help, tolerisation did not impair CD8+ T cell responses.

Conclusions

This result reveals a novel “negative vaccination” strategy where specific CD4+ T cell unresponsiveness may be used to enhance the delayed protective antibody responses in chronic virus infections.  相似文献   

7.

Background

Defects in APC and regulatory cells are associated with diabetes development in NOD mice. We have shown previously that NOD APC are not effective at stimulating CD4+CD25+ regulatory cell function in vitro. We hypothesize that failure of NOD APC to properly activate CD4+CD25+ regulatory cells in vivo could compromise their ability to control pathogenic cells, and activation of NOD APC could restore this defect, thereby preventing disease.

Methodology/Principal Findings

To test these hypotheses, we used the well-documented ability of complete Freund''s adjuvant (CFA), an APC activator, to prevent disease in NOD mice. Phenotype and function of CD4+CD25+ regulatory cells from untreated and CFA-treated NOD mice were determined by FACS, and in vitro and in vivo assays. APC from these mice were also evaluated for their ability to activate regulatory cells in vitro. We have found that sick NOD CD4+CD25+ cells expressed Foxp3 at the same percentages, but decreased levels per cell, compared to young NOD or non-NOD controls. Treatment with CFA increased Foxp3 expression in NOD cells, and also increased the percentages of CD4+CD25+Foxp3+ cells infiltrating the pancreas compared to untreated NOD mice. Moreover, CD4+CD25+ cells from pancreatic LN of CFA-treated, but not untreated, NOD mice transferred protection from diabetes. Finally, APC isolated from CFA-treated mice increased Foxp3 and granzyme B expression as well as regulatory function by NOD CD4+CD25+ cells in vitro compared to APC from untreated NOD mice.

Conclusions/Significance

These data suggest that regulatory T cell function and ability to control pathogenic cells can be enhanced in NOD mice by activating NOD APC.  相似文献   

8.
To better understand the initiation of CD8+ T cell responses during infection, the primary response to the intracellular parasite Toxoplasma gondii was characterized using 2-photon microscopy combined with an experimental system that allowed visualization of dendritic cells (DCs) and parasite specific CD8+ T cells. Infection with T. gondii induced localization of both these populations to the sub-capsular/interfollicular region of the draining lymph node and DCs were required for the expansion of the T cells. Consistent with current models, in the presence of cognate antigen, the average velocity of CD8+ T cells decreased. Unexpectedly, infection also resulted in modulation of the behavior of non-parasite specific T cells. This TCR-independent process correlated with the re-modeling of the lymph node micro-architecture and changes in expression of CCL21 and CCL3. Infection also resulted in sustained interactions between the DCs and CD8+ T cells that were visualized only in the presence of cognate antigen and were limited to an early phase in the response. Infected DCs were rare within the lymph node during this time frame; however, DCs presenting the cognate antigen were detected. Together, these data provide novel insights into the earliest interaction between DCs and CD8+ T cells and suggest that cross presentation by bystander DCs rather than infected DCs is an important route of antigen presentation during toxoplasmosis.  相似文献   

9.
Many viruses encode proteins whose major function is to evade or disable the host T cell response. Nevertheless, most viruses are readily detected by host T cells, and induce relatively strong T cell responses. Herein, we employ transgenic CD4+ and CD8+ T cells as sensors to evaluate in vitro and in vivo antigen presentation by coxsackievirus B3 (CVB3), and we show that this virus almost completely inhibits antigen presentation via the MHC class I pathway, thereby evading CD8+ T cell immunity. In contrast, the presentation of CVB3-encoded MHC class II epitopes is relatively unencumbered, and CVB3 induces in vivo CD4+ T cell responses that are, by several criteria, phenotypically normal. The cells display an effector phenotype and mature into multi-functional CVB3-specific memory CD4+ T cells that expand dramatically following challenge infection and rapidly differentiate into secondary effector cells capable of secreting multiple cytokines. Our findings have implications for the efficiency of antigen cross-presentation during coxsackievirus infection.  相似文献   

10.
McComb S  Mulligan R  Sad S 《PloS one》2010,5(12):e15328

Background

CD8+ T cell responses develop rapidly during infection and are swiftly reduced during contraction, wherein >90% of primed CD8+ T cells are eliminated. The role of apoptotic mechanisms in controlling this rapid proliferation and contraction of CD8+ T cells remains unclear. Surprisingly, evidence has shown non-apoptotic activation of caspase-3 to occur during in vitro T-cell proliferation, but the relevance of these mechanisms to in vivo CD8+ T cell responses has yet to be examined.

Methods and Findings

We have evaluated the activity of caspase-3, a key downstream inducer of apoptosis, throughout the entirety of a CD8+ T cell response. We utilized two infection models that differ in the intensity, onset and duration of antigen-presentation and inflammation. Expression of cleaved caspase-3 in antigen specific CD8+ T cells was coupled to the timing and strength of antigen presentation in lymphoid organs. We also observed coordinated activation of additional canonical apoptotic markers, including phosphatidylserine exposure. Limiting dilution analysis directly showed that in the presence of IL7, very little cell death occurred in both caspase-3hi and caspase-3low CD8+ T cells. The expression of active caspase-3 peaked before effector phenotype (CD62Llow) CD8+ T cells emerged, and was undetectable in effector-phenotype cells. In addition, OVA-specific CD8+ cells remained active caspase-3low throughout the contraction phase.

Conclusions

Our results specifically implicate antigen and not inflammation in driving activation of apoptotic mechanisms without cell death in proliferating CD8+ T cells. Furthermore, the contraction of CD8+ T cell response following expansion is likely not mediated by the key downstream apoptosis inducer, caspase-3.  相似文献   

11.

Background

Pin1 has previously been described to regulate cells that participate in both innate and adaptive immunity. Thus far, however, no role for Pin1 has been described in modulating conventional dendritic cells, innate antigen presenting cells that potently activate naïve T cells, thereby bridging innate and adaptive immune responses.

Methodology/Principal Findings

When challenged with LPS, Pin1-null mice failed to accumulate spleen conventional dendritic cells (cDC). Analysis of steady-state spleen DC populations revealed that Pin1-null mice had fewer CD8+ cDC. This defect was recapitulated by culturing Pin1-null bone marrow with the DC-instructive cytokine Flt3 Ligand. Additionally, injection of Flt3 Ligand for 9 days failed to induce robust expansion of CD8+ cDC in Pin1-null mice. Upon infection with Listeria monocytogenes, Pin1-null mice were defective in stimulating proliferation of adoptively transferred WT CD8+ T cells, suggesting that decreases in Pin1 null CD8+ cDC may affect T cell responses to infection in vivo. Finally, upon analyzing expression of proteins involved in DC development, elevated expression of PU.1 was detected in Pin1-null cells, which resulted from an increase in PU.1 protein half-life.

Conclusions/Significance

We have identified a novel role for Pin1 as a modulator of CD8+ cDC development. Consistent with reduced numbers of CD8+ cDC in Pin1-null mice, we find that the absence of Pin1 impairs CD8+ T cell proliferation in response to infection with Listeria monocytogenes. These data suggest that, via regulation of CD8+ cDC production, Pin1 may serve as an important modulator of adaptive immunity.  相似文献   

12.
T cell-dependent autoimmune diseases are characterized by the expansion of T cell clones that recognize immunodominant epitopes on the target antigen. As a consequence, for a given autoimmune disorder, pathogenic T cell clones express T cell receptors with a limited number of variable regions that define antigenic specificity. Qa-1, a MHC class I-like molecule, presents peptides from the variable region of TCRs to Qa-1-restricted CD8+ T cells. The induction of Vß-specific CD8+ T cells has been harnessed in an immunotherapeutic strategy known as the “T cell vaccination” (TCV) that comprises the injection of activated and attenuated CD4+ T cell clones so as to induce protective CD8+ T cells. We hypothesized that Qa-1-restricted CD8+ regulatory T cells could also constitute a physiologic regulatory arm of lymphocyte responses upon expansion of endogenous CD4+ T cells, in the absence of deliberate exogenous T cell vaccination. We immunized mice with two types of antigenic challenges in order to sequentially expand antigen-specific endogenous CD4+ T cells with distinct antigenic specificities but characterized by a common Vß chain in their TCR. The first immunization was performed with a non-self antigen while the second challenge was performed with a myelin-derived peptide known to drive experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. We show that regulatory Vß-specific Qa-1-restricted CD8+ T cells induced during the first endogenous CD4+ T cell responses are able to control the expansion of subsequently mobilized pathogenic autoreactive CD4+ T cells. In conclusion, apart from the immunotherapeutic TCV, Qa-1-restricted specialized CD8+ regulatory T cells can also be induced during endogenous CD4+ T cell responses. At variance with other regulatory T cell subsets, the action of these Qa-1-restricted T cells seems to be restricted to the immediate re-activation of CD4+ T cells.  相似文献   

13.
CD8+ T cells can be primed by peptides derived from endogenous proteins (direct presentation), or exogenously acquired protein (cross-presentation). However, the relative ability of these two pathways to prime CD8+ T cells during a viral infection remains controversial. Cytomegaloviruses (CMVs) can infect professional antigen presenting cells (APCs), including dendritic cells, thus providing peptides for direct presentation. However, the viral immune evasion genes profoundly impair recognition of infected cells by CD8+ T cells. Nevertheless, CMV infection elicits a very strong CD8+ T cell response, prompting its recent use as a vaccine vector. We have shown previously that deleting the immune evasion genes from murine cytomegalovirus (MCMV) that target class I MHC presentation, has no impact on the size or breadth of the CD8+ T cell response elicited by infection, suggesting that the majority of MCMV-specific CD8+ T cells in vivo are not directly primed by infected professional APCs. Here we use a novel spread-defective mutant of MCMV, lacking the essential glycoprotein gL, to show that cross-presentation alone can account for the majority of MCMV-specific CD8+ T cell responses to the virus. Our data support the conclusion that cross-presentation is the primary mode of antigen presentation by which CD8+ T cells are primed during MCMV infection.  相似文献   

14.
Effective T cell responses can decisively influence the outcome of retroviral infection. However, what constitutes protective T cell responses or determines the ability of the host to mount such responses is incompletely understood. Here we studied the requirements for development and induction of CD4+ T cells that were essential for immunity to Friend virus (FV) infection of mice, according to their TCR avidity for an FV-derived epitope. We showed that a self peptide, encoded by an endogenous retrovirus, negatively selected a significant fraction of polyclonal FV-specific CD4+ T cells and diminished the response to FV infection. Surprisingly, however, CD4+ T cell-mediated antiviral activity was fully preserved. Detailed repertoire analysis revealed that clones with low avidity for FV-derived peptides were more cross-reactive with self peptides and were consequently preferentially deleted. Negative selection of low-avidity FV-reactive CD4+ T cells was responsible for the dominance of high-avidity clones in the response to FV infection, suggesting that protection against the primary infecting virus was mediated exclusively by high-avidity CD4+ T cells. Thus, although negative selection reduced the size and cross-reactivity of the available FV-reactive naïve CD4+ T cell repertoire, it increased the overall avidity of the repertoire that responded to infection. These findings demonstrate that self proteins expressed by replication-defective endogenous retroviruses can heavily influence the formation of the TCR repertoire reactive with exogenous retroviruses and determine the avidity of the response to retroviral infection. Given the overabundance of endogenous retroviruses in the human genome, these findings also suggest that endogenous retroviral proteins, presented by products of highly polymorphic HLA alleles, may shape the human TCR repertoire that reacts with exogenous retroviruses or other infecting pathogens, leading to interindividual heterogeneity.  相似文献   

15.
Allogeneic stem cell transplantation (SCT) is a potentially curative treatment for patients with hematologic malignancies. Its therapeutic effect is largely dependent on recognition of minor histocompatibility antigens (MiHA) by donor-derived CD8+ T cells. Therefore, monitoring of multiple MiHA-specific CD8+ T cell responses may prove to be valuable for evaluating the efficacy of allogeneic SCT. In this study, we investigated the use of the combinatorial encoding MHC multimer technique to simultaneously detect MiHA-specific CD8+ T cells in peripheral blood of SCT recipients. Feasibility of this approach was demonstrated by applying dual-color encoding MHC multimers for a set of 10 known MiHA. Interestingly, single staining using a fluorochrome- and Qdot-based five-color combination showed comparable results to dual-color staining for most MiHA-specific CD8+ T cell responses. In addition, we determined the potential value of combinatorial encoding MHC multimers in MiHA identification. Therefore, a set of 75 candidate MiHA peptides was predicted from polymorphic genes with a hematopoietic expression profile and further selected for high and intermediate binding affinity for HLA-A2. Screening of a large cohort of SCT recipients resulted in the detection of dual-color encoded CD8+ T cells following MHC multimer-based T cell enrichment and short ex vivo expansion. Interestingly, candidate MiHA-specific CD8+ T cell responses for LAG3 and TLR10 derived polymorphic peptides could be confirmed by genotyping of the respective SNPs. These findings demonstrate the potency of the combinatorial MHC multimer approach in the monitoring of CD8+ T cell responses to known and potential MiHA in limited amounts of peripheral blood from allogeneic SCT recipients.  相似文献   

16.

Background

The enzyme indoleamine 2,3-dioxygenase (IDO) contributes to immune tolerance in a variety of settings. In cancer IDO is expressed within the tumor itself as well as in antigen-presenting cells in tumor-draining lymph nodes, where it endorses the establishment of peripheral immune tolerance to tumor antigens. Recently, we described cytotoxic CD8+ T-cell reactivity towards IDO-derived peptides.

Methods and Findings

In the present study, we show that CD4+ helper T cells additionally spontaneously recognize IDO. Hence, we scrutinized the vicinity of the previously described HLA-A*0201-restricted IDO-epitope for CD4+ T-cell epitopes. We demonstrated the presence of naturally occurring IDO-specific CD4+ T cells in cancer patients and to a lesser extent in healthy donors by cytokine release ELISPOT. IDO-reactive CD4+ T cells released IFN-γ, TNF-α, as well as IL-17. We confirm HLA class II-restriction by the addition of HLA class II specific blocking antibodies. In addition, we detected a trend between class I- and class II-restricted IDO responses and detected an association between IDO-specific CD4+ T cells and CD8+ CMV-responses. Finally, we could detect IL-10 releasing IDO-reactive CD4+ T cells.

Conclusion

IDO is spontaneously recognized by HLA class II-restricted, CD4+ T cells in cancer patients and in healthy individuals. IDO-specific T cells may participate in immune-regulatory networks where the activation of pro-inflammatory IDO-specific CD4+ responses may well overcome or delay the immune suppressive actions of the IDO-protein, which are otherwise a consequence of the early expression of IDO in maturing antigen presenting cells. In contrast, IDO-specific regulatory T cells may enhance IDO-mediated immune suppression.  相似文献   

17.
Stimulating naïve CD8+ T cells with specific antigens and costimulatory signals is insufficient to induce optimal clonal expansion and effector functions. In this study, we show that the activation and differentiation of CD8+ T cells require IL-2 provided by activated CD4+ T cells at the initial priming stage within 0–2.5 hours after stimulation. This critical IL-2 signal from CD4+ cells is mediated through the IL-2Rβγ of CD8+ cells, which is independent of IL-2Rα. The activation of IL-2 signaling advances the restriction point of the cell cycle, and thereby expedites the entry of antigen-stimulated CD8+ T-cell into the S phase. Besides promoting cell proliferation, IL-2 stimulation increases the amount of IFNγ and granzyme B produced by CD8+ T cells. Furthermore, IL-2 at priming enhances the ability of P14 effector cells generated by antigen activation to eradicate B16.gp33 tumors in vivo. Therefore, our studies demonstrate that a full CD8+ T-cell response is elicited by a critical temporal function of IL-2 released from CD4+ T cells, providing mechanistic insights into the regulation of CD8+ T cell activation and differentiation.  相似文献   

18.
DNA immunization induces antibodies to the encoded protein, which indicates that the protein must gain access to the extracellular milieu, allowing it to interact with naïve B lymphocytes. It has been suggested that antigen release may be effected by cytotoxic-T-lymphocyte-mediated lysis of transfected antigen-expressing cells; this might be particularly important for the induction of responses to a noncytopathic, cytosolic protein. Here we show that the induction of antibody responses to one such DNA-encoded protein required neither perforin nor CD8+ T cells. In addition, there was no skewing of the immunoglobulin G isotypes in the absence of perforin.DNA immunization is unique in that antigenic proteins are synthesized within the transfected cell in the absence of any associated infectious agent, and the vaccine itself contains no soluble protein that could initiate humoral immune responses. Thus, if the immunizing plasmid expresses a cytoplasmic protein which is relatively noncytopathic and is unable to be effectively processed endogenously by the major histocompatibility complex (MHC) class II antigen presentation pathway, then it might be ineffective in inducing humoral immune responses. The nucleoprotein (NP) of lymphocytic choriomeningitis virus (LCMV) meets these criteria, yet intramuscular injection with a plasmid expressing the NP (pCMV-NP) has been shown by us and others to induce antibody responses in mice (13, 23, 24). Antibodies are also induced following DNA immunization with plasmids expressing other cytoplasmic antigens, such as the measles virus and influenza virus NPs (4, 18, 21). How might such antigens be released from the transfected cell? We hypothesized that the development of CD8+ antigen-specific cytotoxic T lymphocytes (CTL) could lead to the recognition and lysis of cells expressing plasmid-derived NP, resulting in the liberation of protein into the extracellular milieu, where it can then interact with B cells and antigen-presenting cells (APCs). CTL-mediated release of LCMV NP occurs during virus infection (12), and the possibility that it occurs following DNA immunization was strengthened by our observations of a profound myositis following intramuscular inoculation of pCMV-NP into LCMV-infected or -immune mice; the peak of the inflammatory infiltrate in acutely infected mice coincided with the development of anti-LCMV CTL, and destruction of muscle cells occurred (8, 22). Professional APCs, which are known to be a source of plasmid-expressed antigens (5) and which appear to be the cell type responsible for initiating immune responses following DNA immunization (6, 7), can also be recognized and lysed by CTL. Dendritic cells infected with human immunodeficiency virus are susceptible to lysis in vitro by CTL (11), and there is evidence to suggest that CD8+ T cells can limit the immune response by lysing APCs in vivo (13). Therefore, the possibility of lytic release of protein is not limited to NP-expressing myocytes but extends to most transfected somatic cells, including APCs. To assess what effect such lysis may have on the generation or maintenance of B-cell responses following DNA immunization, we analyzed humoral immune responses in DNA-immunized mice that lacked the cytolytic protein perforin (10, 20). Although antigen-specific CD8+-T-cell responses were induced in these mice by vaccination with pCMV-NP, they were unable to lyse NP-expressing cells in a perforin-dependent manner (data not shown).

Strong antibody responses are induced by DNA immunization of PKO mice.

To determine if a lack of perforin-mediated lysis by antigen-specific CTL resulted in an alteration in the temporal appearance or maintenance of antiviral serum antibodies, antibody levels were measured in PKO and C57BL/6 mice at 0, 2, 4, and 6 weeks after they received a single 50-μg intramuscular injection of pCMV-NP DNA. Serum immunoglobulin G (IgG) levels in individual mice were measured by enzyme-linked immunosorbent assay (ELISA), and the average for each group was calculated based upon the optical density measurement at a dilution of 1:200; the results are shown in Fig. Fig.1A.1A. Within 2 weeks of vaccination, anti-LCMV antibodies were demonstrable in both PKO and C57BL/6 mice, and the average levels in both groups were similar. The slight drop in antibody levels in the C57BL/6 mice at 4 weeks after DNA immunization is not statistically significant, but at 6 weeks, the difference is highly significant. In mice, the average half-life of an IgG molecule is approximately 6 to 10 days (17, 19). Therefore, the high level of antibodies present at 6 weeks postimmunization implies ongoing synthesis of NP-specific IgG in PKO mice, whereas the drop in antibody levels indicates decreased IgG synthesis in C57BL/6 mice. A representative analysis of the antibody response at 6 weeks postimmunization is presented in Fig. Fig.1B.1B. Anti-LCMV serum antibody levels were measured by ELISA in individual perforin-positive (C57BL/6, n = 6) or perforin-negative (PKO, n = 8) animals 6 weeks postvaccination. LCMV DNA-vaccinated C57BL/6 and PKO mice both produced anti-LCMV IgG. Together, these data clearly show that perforin-mediated release of plasmid-expressed LCMV NP is not required for the induction of humoral responses following intramuscular DNA injection. Open in a separate windowFIG. 1PKO mice mount strong antibody responses. (A) PKO and C57BL/6 mice (n = 4/group) were immunized with pCMV-NP and bled biweekly. Anti-LCMV serum antibodies were measured by ELISA. The mean optical density readings at 492 nm (OD492) are shown for serum dilutions of 1:200, with standard errors. (B) Representative analyses of antibody responses at 6 weeks postimmunization. Anti-LCMV antibody levels in groups of DNA-immunized PKO (n = 8) and C57BL/6 (n = 6) mice were measured by ELISA. For controls, sera were derived from LCMV-immune or nonimmune C57BL/6 mice.

Isotype responses are unaltered in DNA-immunized perforin-deficient mice.

IgG responses to the LCMV NP require CD4+-T-cell help, and unlike the LCMV glycoprotein, endogenously synthesized NP cannot gain access to the MHC class II antigen presentation pathway (15). Thus, the CD4+ T cells required for immunoglobulin isotype class switching must be primed by APCs which have acquired extracellular NP. To determine if the isotype pattern was skewed in the absence of perforin, titers of LCMV-specific IgG, IgG1, IgG2a, and IgG2b were determined by ELISA in individual PKO and C57BL/6 mice 6 weeks after DNA immunization (Fig. (Fig.2).2). At this time point, all of the pCMV-NP DNA-immunized mice contained virus-specific immunoglobulin in their serum. Anti-LCMV IgG endpoint titers among PKO mice ranged from 1:4,000 to 1:15,000, with a geometric mean titer (Fig. (Fig.2)2) of 1:6,825. In contrast, among the C57BL/6 vaccines, the total IgG titers ranged from 1:1,700 to 1:3,400, with a geometric mean of 1:2,455. Detectable IgG1 and IgG2a were present in the sera of all mice of both strains with the exception of a single PKO mouse which lacked demonstrable levels of anti-LCMV IgG1. Therefore, the vaccinated PKO mice showed no skewing of their isotype classes, suggesting that CD4+-T-cell responses are appropriately induced in these mice and providing further evidence that perforin is not required for the release of NP from transfected cells. Open in a separate windowFIG. 2No skewing of antibody isotypes in PKO mice. IgG isotype titers were measured in individual PKO and C57BL/6 mice at 6 weeks after immunization with pCMV-NP. Triangles, individual mice; circles, geometric mean titers for each group. The LCMV-immune sample represents pooled sera from virus-immune C57BL/6 mice.

CD8+ T cells play no part in controlling humoral responses to DNA-encoded antigens.

Our data show that perforin is not required to prime B-cell responses against a plasmid-expressed, cytoplasmic antigen. Perforin is expressed by CD8+ CTL, some CD4+ T cells, and natural killer (NK) cells; we can therefore conclude that perforin-mediated lysis by any of these cell populations is not required for DNA-mediated antibody induction. However, other CD8+-T-cell functions, such as the Fas pathway (16) or the secretion of cytotoxic cytokines, can mediate target cell death and thus could liberate intracellular antigens (9). Our observation that PKO mice had elevated antibody titers compared to those of C57BL/6 mice (Fig. (Fig.11 and and2)2) also raised the possibility that perforin-mediated lysis might actually depress humoral responses to DNA immunization by killing cells expressing foreign plasmid-encoded antigens. Consistent with this idea, CD8+-T-cell-mediated lysis of APCs has been shown to suppress immune responses (13), and perforin has been shown to play a role in regulating immunity (14). If this hypothesis is correct, the absence of perforin might result in elevated and/or prolonged responses to DNA-encoded immunogens. To determine whether CD8+ T cells might play a perforin-independent role in antigen release and antibody induction, CD8+ T cells were removed from C57BL/6 mice by immunodepletion prior to and throughout the course of a DNA immunization experiment. C57BL/6 mice were treated with either an anti-mouse CD8+-T-cell monoclonal antibody or saline daily for 3 days prior to immunization with pCMV-NP and weekly thereafter. Nondepleted PKO mice served as controls. Mice were bled biweekly, and the average IgG response for each group is shown in Fig. Fig.3A.3A. Antibody responses were easily detected in CD8-depleted mice, showing that CD8+-T-cell functions are not required to release soluble NP for the initiation of humoral immune responses. Furthermore, at all time points, the antibody responses in CD8-depleted and nondepleted C57BL/6 mice were indistinguishable; therefore, it appears to be unlikely that CD8+ T cells play a role in down-regulating DNA-induced antibody responses. To confirm that the CD8+ cells had been successfully ablated by the monoclonal antibody treatment, splenocytes were obtained from mice immediately before DNA immunization (data not shown) and from three mice (two depleted and one sham-depleted) at the conclusion of the experiment and were analyzed for CD8 expression by flow cytometry (Fig. (Fig.3B).3B). The administration of anti-CD8 monoclonal antibody maintained CD8+-T-cell populations at almost undetectable levels throughout the course of the experiment. Thus, we conclude that the development of fully functional CD8+ CTL following DNA immunization neither inhibits nor enhances humoral immune responses. Open in a separate windowFIG. 3Depletion of CD8 cells has no effect on antibody induction by plasmid DNA. (A) Anti-LCMV antibody titers were measured over time in C57BL/6 mice depleted of CD8+ T cells (C57+anti CD8), nondepleted C57BL/6 mice (C57+saline), and nondepleted PKO mice (PKO+saline). Each group consisted of three animals, and the average antibody endpoint titer for each group at each time point is shown. (B) To confirm successful depletion of CD8+ cells, splenocytes were harvested from two mice treated with anti-CD8 antibody and from one control mouse. These cells were evaluated for CD8 expression by flow cytometry.These data argue against the hypothesis that lytic-antigen-specific CD8+ CTL have a profound effect on the generation of humoral immunity to plasmid-encoded antigens. Perforin is normally expressed not only in CD8+ T cells but also in some CD4+ T cells and in NK cells. Our observations therefore are not restricted to perforin-mediated CD8+-T-cell lysis but also show that perforin-mediated lysis by CD4+ T cells or NK cells plays no essential role in antigen release. Furthermore, the ratios of IgG1 to IgG2a were similar regardless of the perforin status of the mouse (Fig. (Fig.2).2). Thus, not only is perforin expendable in IgG induction but also its absence has no effect on the antibody classes—and, presumably, the ratio of Th1 to Th2 cells—induced.In conclusion, it is clear that plasmids encoding proteins which are relatively noncytotoxic and incapable of endogenous MHC class II antigen presentation can elicit strong IgG responses in the absence of CD8+ T cells and perforin. Thus, the mechanism of antigen release and B-cell stimulation remains to be determined. It is important that we identify the underlying mechanisms to permit the rational optimization of DNA vaccines.  相似文献   

19.
Previous studies have suggested that coxsackievirus B (CVB) activates CD8+ T cells in vivo, but the extent of this activation and the antigen specificity of the CD8+ T cells remain uncertain. Furthermore, CVB-induced CD4+ T-cell responses have not been carefully investigated. Herein, we evaluate CD8+ and CD4+ T-cell responses both in a secondary lymphoid organ (spleen) and in peripheral tissues (heart and pancreas), using a recombinant CVB3 (rCVB3.6) that encodes well-characterized CD8+ and CD4+ T-cell epitopes. Despite reaching high levels in vivo, rCVB3.6 failed to trigger a marked expansion of CD8+ or CD4+ T cells, and T-cell activation was surprisingly limited. Furthermore, epitope-specific effector functions could not be detected using highly sensitive in vivo and ex vivo assays. Moreover, major histocompatibility complex (MHC) class I tetramer analysis indicated that our inability to detect CVB3-specific CD8+ T-cell responses could not be explained by the cells being dysfunctional. In contrast to naïve T cells, epitope-specific memory CD8+ and CD4+ T cells proliferated markedly, indicating that both of the rCVB3.6-encoded epitopes were presented by their respective MHC molecules in vivo. These data are consistent with the observation that several CVB3 proteins can limit the presentation of viral epitopes on the surface of infected cells and suggest that the level of MHC/peptide complex is sufficient to trigger memory but not naïve T cells. Finally, our findings have implications for the biological significance of cross-priming, a process thought by some to be important for the induction of antiviral CD8+ T-cell responses.Coxsackieviruses are members of the picornavirus family and enterovirus genus, which includes type A and B coxsackieviruses, polioviruses, echoviruses, and other unclassified enteroviruses. Although the majority of type B coxsackievirus (CVB) infections in humans are subclinical or cause relatively mild disease (including rash, myalgia, or upper respiratory complications), CVB are important human pathogens, and a substantial proportion of infections can lead to severe—even lethal—acute and chronic diseases. In particular, CVB is the most common infectious cause of myocarditis, which can lead to dilated cardiomyopathy and cardiac failure (38, 44, 45). CVB also targets cells of the central nervous system and the pancreas, frequently leading to aseptic meningitis and pancreatitis (7, 12, 33, 35, 40). Overall, CVB infection can cause considerable morbidity and mortality, particularly in newborns and in young or immunocompromised individuals (35, 52).The murine model of CVB3 infection is a valuable system for studying CVB pathogenesis and immunity, as mice infected with CVB develop diseases similar to those observed in humans (52, 53). Intraperitoneal inoculation of adult C57BL/6 mice with CVB3 results in systemic acute infection; viremia peaks on day 2 to 3 postinfection (p.i.), and infectious virus is cleared by 2 weeks p.i. (33, 34). Control of CVB3 infection depends on both cell-mediated and humoral components of the immune response. Agammaglobulinemic individuals are particularly susceptible to CVB3-associated encephalitis (15, 18), and mice lacking B cells develop a chronic infection and remain viremic for at least 2 months; viremia can be alleviated by the adoptive transfer of B cells from CVB3-immune wild-type mice (34). CD8+ T cells also play an important role in controlling virus replication. T cells are present in the inflammatory infiltrates associated with myocarditis and pancreatitis (17, 20, 41), and CD8+ T-cell depletion of CVB3-infected mice simultaneously increases viral titers and reduces myocarditis, suggesting that T-cell-mediated protection is associated with elevated immunopathology (17). This immunopathology can be uncoupled from antiviral efficacy; mice lacking perforin control cardiac infection just as well as wild-type mice but show markedly diminished myocarditis (14).Many—probably most—acute viral infections trigger extensive CD8+ T-cell activation and division; these responses can readily be detected directly ex vivo, without any need for extensive restimulation. The convincing evidence that CD8+ T cells can contribute to control of CVB3 in mice, together with the fact that CVB3 replicates to high titers in many mouse tissues, led us to surmise that CVB3—like most other viruses—would induce readily detectable CD8+ T-cell responses in mice. Indeed, early studies had identified cytolytic T-cell activity in CVB3-infected mice, although the precise antigen specificity of the cells was unknown (16, 21, 22). Subsequent elegant work showed that synthetic peptides representing CVB3 VP1 sequences could drive in vitro T-cell proliferation, but neither the phenotype of the proliferating T cells (CD4+ or CD8+) nor the precise epitope specificity was determined (19). Therefore, we undertook a preliminary analysis of epitope-specific CD8+ T-cell responses against CVB3; contrary to our expectations, we found that CVB3-induced epitope-specific CD8+ T-cell responses were difficult to detect (42). However, those studies were incomplete: they relied on ex vivo detection methods of rather limited sensitivity, and they were limited to cells from the spleen. Furthermore, those studies focused only on CD8+ T cells, and it is clear that regulation of antiviral CD8+ T cells differs from that of CD4+ T cells. Therefore, herein we have extended our previous analysis in five ways: first, we evaluate general T-cell activation in CVB3-infected mice; second, we use more sensitive in vivo approaches to detect epitope-specific T-cell responses; third, we investigate the possibility that the virus induces the expansion of dysfunctional T cells; fourth, we extend our analyses of CVB3 epitope-specific T-cell responses to major targets of infection, such as the heart, where CD8+ T cells are present in the virus-induced infiltrate; and, fifth, we investigate CD4+ T-cell responses induced by CVB3. Our studies employ a new recombinant CVB3 (rCVB3) that encodes both a CD8 and CD4 T-cell epitope derived from lymphocytic choriomeningitis virus (LCMV). Our data are not only relevant to understanding the T-cell responses induced by coxsackievirus in particular but also have broader implications for the mechanism(s) by which CD4+ and CD8+ T cells are induced by viruses in general.  相似文献   

20.
The role of CD4+ helper T cells in modulating the acquired immune response to herpes simplex virus type 1 (HSV-1) remains ill defined; in particular, it is unclear whether CD4+ T cells are needed for the generation of the protective HSV-1-specific CD8+-T-cell response. This study examined the contribution of CD4+ T cells in the generation of the primary CD8+-T-cell responses following acute infection with HSV-1. The results demonstrate that the CD8+-T-cell response generated in the draining lymph nodes of CD4+-T-cell-depleted C57BL/6 mice and B6-MHC-II−/− mice is quantitatively and qualitatively distinct from the CD8+ T cells generated in normal C57BL/6 mice. Phenotypic analyses show that virus-specific CD8+ T cells express comparable levels of the activation marker CD44 in mice lacking CD4+ T cells and normal mice. In contrast, CD8+ T cells generated in the absence of CD4+ T cells express the interleukin 2 receptor α-chain (CD25) at lower levels. Importantly, the CD8+ T cells in the CD4+-T-cell-deficient environment are functionally active with respect to the expression of cytolytic activity in vivo but exhibit a diminished capacity to produce gamma interferon and tumor necrosis factor alpha. Furthermore, the primary expansion of HSV-1-specific CD8+ T cells is diminished in the absence of CD4+-T-cell help. These results suggest that CD4+-T-cell help is essential for the generation of fully functional CD8+ T cells during the primary response to HSV-1 infection.Infection due to herpes simplex virus type 1 (HSV-1) results in a wide spectrum of clinical presentations depending on the host''s age, the host''s immune status, and the route of inoculation (47). HSV-1 typically causes mild and self-limited lesions on the orofacial areas or genital sites. However, the disease can be life-threatening, as in the case of neonatal and central nervous system infections (18). The host''s immune responses, particularly CD8+ T cells, play an important role in determining the outcome of HSV infections in both the natural human host (18, 19, 28) and experimental murine models (11, 43). Immunodepletion and adoptive transfer studies have demonstrated the role of CD8+ T cells in reducing viral replication, resolving cutaneous disease, and providing overall protection upon rechallenge (6, 25, 26). CD8+ T cells play a particularly important role in preventing infection of the peripheral nervous system (PNS) and the reactivation of latent virus from neurons in the sensory ganglia of infected mice (21, 24, 36). The mechanisms that CD8+ T cells employ include gamma interferon (IFN-γ) production and functions associated with cytolytic granule content at the sites of primary infection (23, 31, 38). In the PNS of infected mice, the mechanisms primarily involve IFN-γ secretion (16, 20, 29), particularly against infected neurons expressing surface Qa-1 (41). Histopathological evidence from HSV-1-infected human ganglion sections show a large CD8+-T-cell infiltrate and the presence of inflammatory cytokines, suggesting that the presence of activated, effector memory cells within the PNS is important for maintaining HSV-1 latency in the natural human host (10, 42).The generation of a robust CD8+-T-cell response is essential for the control of various infectious pathogens. Some studies suggest that a brief interaction with antigen-presenting cells (APCs) is sufficient for CD8+-T-cell activation and expansion into functional effectors (44). However, the magnitude and quality of the overall CD8+-T-cell response generated may be dependent on additional factors (49). Recent evidence suggests that CD4+ T cells facilitate the activation and development of CD8+-T-cell responses either directly through the provision of cytokines or indirectly by the conditioning of dendritic cells (DC) (8, 48, 51). Those studies suggested that the latter mechanism is the dominant pathway, wherein CD4+ T cells assist CD8+-T-cell priming via the engagement of CD40 ligand (CD154) on CD4+ T cells and CD40 expressed on DC (4, 30, 33). This interaction results in the activation and maturation of DC, making them competent to stimulate antigen-specific CD8+-T-cell responses (35, 37).The requirement for CD4+-T-cell help in the generation of primary and secondary CD8+-T-cell responses to antigen varies. Primary CD8+-T-cell responses to infectious pathogens, such as Listeria monocytogenes, lymphocytic choriomeningitis virus (LCMV), influenza virus, and vaccinia virus, can be mounted effectively independently of CD4+-T-cell help (3, 12, 22, 34). In contrast, primary CD8+-T-cell responses to nonmicrobial antigens display an absolute dependence on CD4+-T-cell help (4, 5, 30, 33, 46). This observed difference in the requirement for CD4+-T-cell help may ultimately be a product of the initial inflammatory stimulus generated following immunization (49). Microbial antigens trigger an inflammatory response that can lead to the direct activation and priming of APCs, such as DC, thereby bypassing the need for CD4+-T-cell help. Nonmicrobial antigens, however, trigger an attenuated inflammatory response that does not directly activate and prime DCs. In the absence of this inflammation, CD4+ T cells are thought to condition and license DC functions through CD154/CD40 interactions, which leads to the subsequent activation of antigen-specific CD8+-T-cell responses (5, 49). Even in the case of pathogens where primary CD8+-T-cell responses were independent of CD4+-T-cell help, the secondary responses to these pathogens were found to be defective in the absence of CD4+-T-cell help (3, 12, 34, 40).The requirement for CD4+-T-cell help in priming CD8+-T-cell responses against HSV-1 infection is not well defined. Earlier studies with HSV-1 suggested that CD4+ T cells play an important role in the generation of primary CD8+-T-cell responses, detected in vitro, to acute infection with HSV-1 (14), principally through the provision of interleukin 2 (IL-2) for optimal CD8+-T-cell differentiation and proliferation. Subsequent studies, utilizing an in vivo approach, indicated that CD4+ T cells were not required for CD8+-T-cell-mediated cytolytic function (23). CD4+ T cells are thought to provide help by conditioning DC in a cognate, antigen-specific manner, thereby making them competent to stimulate HSV-1-specific CD8+-T-cell responses (37). By contrast, findings from other studies show that CD4+-T-cell-depleted mice were able to fully recover from acute infection with HSV-1 (38). These studies imply that the absence of CD4+ T cells does not prevent priming of CD8+ T cells in vivo.Studies from this laboratory have identified two distinct HSV-1-specific CD8+-T-cell subpopulations generated during the primary response, based upon the ability to synthesize IFN-γ following antigenic stimulation in vitro (1). To better understand the need for CD4+-T-cell help, we examined the functional characteristics and phenotypes of these CD8+-T-cell populations generated during a primary response to acute infection with HSV-1 in mice lacking CD4+ T cells. Our findings show that primary CD8+-T-cell responses to HSV-1 are compromised in the absence of CD4+-T-cell help. Specifically, the HSV-1 gB-specific CD8+ T cells produced in the absence of CD4+ T cells were found to be active with regard to cytolysis in vivo but were functionally impaired in the production of IFN-γ and TNF-α compared with intact C57BL/6 mice. Virus-specific CD8+ T cells were also reduced in number in CD4-depleted mice and in B6 mice lacking major histocompatibility complex (MHC) class II expression (B6-MHC-II−/−) compared to wild-type (WT) mice. In addition, our data showed higher virus burdens in the infectious tissues obtained from mice lacking CD4+ T cells than in those from intact mice. Collectively, these findings demonstrate that CD4+-T-cell help is essential for the generation of primary CD8+-T-cell responses following acute cutaneous infection with HSV-1.  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