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.

Background

IL-9 is a growth factor for T- and mast-cells that is secreted by human Th2 cells. We recently reported that IL-4+TGF-β directs mouse CD4+CD25CD62L+ T cells to commit to inflammatory IL-9 producing CD4+ T cells.

Methodology/Principal Findings

Here we show that human inducible regulatory T cells (iTregs) also express IL-9. IL-4+TGF-β induced higher levels of IL-9 expression in plate bound-anti-CD3 mAb (pbCD3)/soluble-anti-CD28 mAb (sCD28) activated human resting memory CD4+CD25CD45RO+ T cells as compared to naïve CD4+CD25CD45RA+ T cells. In addition, as compared to pbCD3/sCD28 plus TGF-β stimulation, IL-4+TGF-β stimulated memory CD4+CD25CD45RO+ T cells expressed reduced FOXP3 protein. As analyzed by pre-amplification boosted single-cell real-time PCR, human CD4+IL-9+ T cells expressed GATA3 and RORC, but not IL-10, IL-13, IFNγ or IL-17A/F. Attempts to optimize IL-9 production by pbCD3/sCD28 and IL-4+TGF-β stimulated resting memory CD4+ T cells demonstrated that the addition of IL-1β, IL-12, and IL-21 further enhance IL-9 production.

Conclusions/Significance

Taken together these data show both the differences and similarities between mouse and human CD4+IL9+ T cells and reaffirm the powerful influence of inflammatory cytokines to shape the response of activated CD4+ T cells to antigen.  相似文献   

2.
CD4+ T-cell help enables antiviral CD8+ T cells to differentiate into fully competent memory cells and sustains CD8+ T-cell-mediated immunity during persistent virus infection. We recently reported that mice of C57BL/6 and C3H strains differ in their dependence on CD28 and CD40L costimulation for long-term control of infection by polyoma virus, a persistent mouse pathogen. In this study, we asked whether mice of these inbred strains also vary in their requirement for CD4+ T-cell help for generating and maintaining polyoma virus-specific CD8+ T cells. CD4+ T-cell-depleted C57BL/6 mice mounted a robust antiviral CD8+ T-cell response during acute infection, whereas unhelped CD8+ T-cell effectors in C3H mice were functionally impaired during acute infection and failed to expand upon antigenic challenge during persistent infection. Using (C57BL/6 × C3H)F1 mice, we found that the dispensability for CD4+ T-cell help for the H-2b-restricted polyoma virus-specific CD8+ T-cell response during acute infection extends to the H-2k-restricted antiviral CD8+ T cells. Our findings demonstrate that dependence on CD4+ T-cell help for antiviral CD8+ T-cell effector differentiation can vary among allogeneic strains of inbred mice.  相似文献   

3.
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   

4.
Adaptive CD4+ and CD8+ T-cell responses have been associated with control of human immunodeficiency virus/simian immunodeficiency virus (HIV/SIV) replication. Here, we have designed a study with Indian rhesus macaques to more directly assess the role of CD8 SIV-specific responses in control of viral replication. Macaques were immunized with a DNA prime-modified vaccinia virus Ankara (MVA)-SIV boost regimen under normal conditions or under conditions of antibody-induced CD4+ T-cell deficiency. Depletion of CD4+ cells was performed in the immunized macaques at the peak of SIV-specific CD4+ T-cell responses following the DNA prime dose. A group of naïve macaques was also treated with the anti-CD4 depleting antibody as a control, and an additional group of macaques immunized under normal conditions was depleted of CD8+ T cells prior to challenge exposure to SIVmac251. Analysis of the quality and quantity of vaccine-induced CD8+ T cells demonstrated that SIV-specific CD8+ T cells generated under conditions of CD4+ T-cell deficiency expressed low levels of Bcl-2 and interleukin-2 (IL-2), and plasma virus levels increased over time. Depletion of CD8+ T cells prior to challenge exposure abrogated vaccine-induced protection as previously shown. These data support the notion that adaptive CD4+ T cells are critical for the generation of effective CD8+ T-cell responses to SIV that, in turn, contribute to protection from AIDS. Importantly, they also suggest that long-term protection from disease will be afforded only by T-cell vaccines for HIV that provide a balanced induction of CD4+ and CD8+ T-cell responses and protect against early depletion of CD4+ T cells postinfection.  相似文献   

5.
Lentiviral vectors deliver antigens to dendritic cells (DCs) in vivo, but they do not trigger DC maturation. We therefore expressed a viral protein that constitutively activates NF-κB, vFLIP from Kaposi's sarcoma-associated herpesvirus (KSHV), in a lentivector to mature DCs. vFLIP activated NF-κB in mouse bone marrow-derived DCs in vitro and matured these DCs to a similar extent as lipopolysaccharide; costimulatory markers CD80, CD86, CD40, and ICAM-1 were upregulated and tumor necrosis factor alpha and interleukin-12 secreted. The vFLIP-expressing lentivector also matured DCs in vivo. When we coexpressed vFLIP in a lentivector with ovalbumin (Ova), we found an increased immune response to Ova; up to 10 times more Ova-specific CD8+ T cells secreting gamma interferon were detected in the spleens of vFLIP_Ova-immunized mice than in the spleens of mice immunized with GFP_Ova. Furthermore, this increased CD8+ T-cell response correlated with improved tumor-free survival in a tumor therapy model. A single immunization with vFLIP_Ova also reduced the parasite load when mice were challenged with OVA-Leishmania donovani. In conclusion, vFLIP from KSHV is a DC activator, maturing DCs in vitro and in vivo. This demonstrates that NF-κB activation is sufficient to induce many aspects of DC maturation and that expression of a constitutive NF-κB activator can improve the efficacy of a vaccine vector.  相似文献   

6.
The antiviral role of CD4+ T cells in virus-induced pathologies of the central nervous system (CNS) has not been explored extensively. Control of neurotropic mouse hepatitis virus (JHMV) requires the collaboration of CD4+ and CD8+ T cells, with CD8+ T cells providing direct perforin and gamma interferon (IFN-γ)-mediated antiviral activity. To distinguish bystander from direct antiviral contributions of CD4+ T cells in virus clearance and pathology, memory CD4+ T cells purified from wild type (wt), perforin-deficient (PKO), and IFN-γ-deficient (GKO) immune donors were transferred to immunodeficient SCID mice prior to CNS challenge. All three donor CD4+ T-cell populations controlled CNS virus replication at 8 days postinfection, indicating IFN-γ- and perforin-independent antiviral function. Recipients of GKO CD4+ T cells succumbed more rapidly to fatal disease than untreated control infected mice. In contrast, wt and PKO donor CD4+ T cells cleared infectious virus to undetectable levels and protected from fatal disease. Recipients of all CD4+ T-cell populations exhibited demyelination. However, it was more severe in wt CD4+ T-cell recipients. These data support a role of CD4+ T cells in virus clearance and demyelination. Despite substantial IFN-γ-independent antiviral activity, IFN-γ was crucial in providing protection from death. IFN-γ reduced neutrophil accumulation and directed macrophages to white matter but did not ameliorate myelin loss.  相似文献   

7.
Members of the Bcl-2 family have critical roles in regulating tissue homeostasis by modulating apoptosis. Anti-apoptotic molecules physically interact and restrain pro-apoptotic family members preventing the induction of cell death. However, the specificity of the functional interactions between pro- and anti-apoptotic Bcl-2 family members remains unclear. The pro-apoptotic Bcl-2 family member Bcl-2 interacting mediator of death (Bim) has a critical role in promoting the death of activated, effector T cells following viral infections. Although Bcl-2 is an important Bim antagonist in effector T cells, and Bcl-xL is not required for effector T-cell survival, the roles of other anti-apoptotic Bcl-2 family members remain unclear. Here, we investigated the role of myeloid cell leukemia sequence 1 (Mcl-1) in regulating effector T-cell responses in vivo. We found, at the peak of the response to lymphocytic choriomeningitis virus (LCMV) infection, that Mcl-1 expression was increased in activated CD4+ and CD8+ T cells. Retroviral overexpression of Mcl-1-protected activated T cells from death, whereas deletion of Mcl-1 during the course of infection led to a massive loss of LCMV-specific CD4+ and CD8+ T cells. Interestingly, the co-deletion of Bim failed to prevent the loss of Mcl-1-deficient T cells. Furthermore, lck-driven overexpression of a Bcl-xL transgene only partially rescued Mcl-1-deficient effector T cells suggesting a lack of redundancy between the family members. In contrast, additional loss of Bax and Bak completely rescued Mcl-1-deficient effector T-cell number and function, without enhancing T-cell proliferation. These data suggest that Mcl-1 is critical for promoting effector T-cell responses, but does so by combating pro-apoptotic molecules beyond Bim.  相似文献   

8.
Poxvirus vectors have proven to be highly effective for boosting immune responses in diverse vaccine settings. Recent reports reveal marked differences in the gene expression of human dendritic cells infected with two leading poxvirus-based human immunodeficiency virus (HIV) vaccine candidates, New York vaccinia virus (NYVAC) and modified vaccinia virus Ankara (MVA). To understand how complex genomic changes in these two vaccine vectors translate into antigen-specific systemic immune responses, we undertook a head-to-head vaccine immunogenicity and efficacy study in the pathogenic HIV type 1 (HIV-1) model of AIDS in Indian rhesus macaques. Differences in the immune responses in outbred animals were not distinguished by enzyme-linked immunospot assays, but differences were distinguished by multiparameter fluorescence-activated cell sorter analysis, revealing a difference between the number of animals with both CD4+ and CD8+ T-cell responses to vaccine inserts (MVA) and those that elicit a dominant CD4+ T-cell response (NYVAC). Remarkably, vector-induced differences in CD4+/CD8+ T-cell immune responses persisted for more than a year after challenge and even accompanied antigenic modulation throughout the control of chronic infection. Importantly, strong preexposure HIV-1/simian immunodeficiency virus-specific CD4+ T-cell responses did not prove deleterious with respect to accelerated disease progression. In contrast, in this setting, animals with strong vaccine-induced polyfunctional CD4+ T-cell responses showed efficacies similar to those with stronger CD8+ T-cell responses.  相似文献   

9.
A productive CD8+ T-cell response to a viral infection requires rapid division and proliferation of virus-specific CD8+ T cells. Tetramer-based enrichment assays have recently given estimates of the numbers of peptide-major histocompatibility complex-specific CD8+ T cells in naïve mice, but precursor frequencies for entire viruses have been examined only by using in vitro limiting-dilution assays (LDAs). To examine CD8+ T-cell precursor frequencies for whole viruses, we developed an in vivo LDA and found frequencies of naïve CD8+ T-cell precursors of 1 in 1,444 for vaccinia virus (VV) (∼13,850 VV-specific CD8+ T cells per mouse) and 1 in 2,958 for lymphocytic choriomeningitis virus (LCMV) (∼6,761 LCMV-specific CD8+ T cells per mouse) in C57BL/6J mice. In mice immune to VV, the number of VV-specific precursors, not surprisingly, dramatically increased to 1 in 13 (∼1,538,462 VV-specific CD8+ T cells per mouse), consistent with estimates of VV-specific memory T cells. In contrast, precursor numbers for LCMV did not increase in VV-immune mice (1 in 4,562, with ∼4,384 LCMV-specific CD8+ T cells per VV-immune mouse). Using H-2Db-restricted LCMV GP33-specific P14-transgenic T cells, we found that, after donor T-cell take was accounted for, approximately every T cell transferred underwent a full proliferative expansion in response to LCMV infection. This high efficiency was also seen with memory populations, suggesting that most antigen-specific T cells will proliferate extensively at a limiting dilution in response to infections. These results show that frequencies of naïve and memory CD8+ T cell precursors for whole viruses can be remarkably high.The immune response to a viral infection often involves the rapid proliferation of CD8+ effector T cells that recognize virus-infected targets expressing 8- to 11-amino-acid-long peptides on class I major histocompatibility complex (MHC) molecules. This recognition is mediated by membrane-bound T-cell receptors (TCRs) that are generated through largely random DNA recombination events of the many TCRα and -β genes, encoding polypeptide chains that heterodimerize to form the recognition structure of T cells. The recombination of the segments also involves addition or deletion of nucleotides during the joining process, causing even greater diversity, and these processes allow for a very broad range of T-cell specificities, with a calculated theoretical diversity of ∼1015 TCRs in the mouse (7). By use of PCR, CDR3 spectratyping, and sequencing techniques, it was estimated that there are approximately 2 × 106 distinct TCR specificities in a mouse spleen (1, 5). This is far below the theoretical level of T-cell diversity, but considering estimates of T-cell degeneracy that propose that a single TCR can recognize up to 106 peptide-MHC (pMHC) complexes (17, 36), it is likely that the functional diversity is much greater than the number of individual TCRs.It has been of interest to calculate the number of T cells that would either recognize or respond to a pathogen or to a specific pMHC complex. Early estimates of numbers of CD8+ T cells that are specific to a single virus, i.e., precursor frequencies, took advantage of an in vitro limiting-dilution assay (LDA) and calculated CD8+ T-cell virus-specific precursor frequencies to be on the order of 1 in 100,000 in naïve mice and predicted that these cells needed to undergo about 15 divisions to reach the higher precursor frequencies found at day 8 postinfection (29, 30). The efficiency of such assays, however, is relatively poor. Later studies estimated the number of pMHC-specific CD8+ T cells in a naïve mouse by CDR3 sequencing. H-2Kd-restricted T cells specific to HLA residues 170 to 179 (HLA 170-179) were sorted by tetramer from human tumor-immunized mice, and their Vβ CDR3 regions were sequenced. After a plateau suggesting that the majority of the different TCRs had been sequenced was reached, exhaustive sequencing was then used to identify the frequencies of these sequences in naïve mice. These studies found that there were about 600 CD8+ T cells specific for that pMHC complex in naïve mice (4). A second strategy used an in vivo competition assay with H-2Db-restricted lymphocytic choriomeningitis virus (LCMV) GP33-specific P14-transgenic T cells to estimate the number of GP33-specific CD8 T cells in naïve mice and calculated the number to be between 100 to 200 cells per mouse (2).Others estimated numbers of pMHC-specific T cells by sequencing the CDR3β regions of antigen-specific T cells that had expanded during an acute infection. By calculating a measure of CDR3 diversity and then assuming a logarithmic distribution of diversity, they extrapolated the number of T-cell clones that responded to an acute infection. With this technique, 300 to 500 H-2Db-restricted mouse hepatitis virus (MHV)-encoded S510 clonotypes were calculated to be in the central nervous systems of acutely infected mice, with ∼100 to 900 clonotypes calculated to be in chronically infected mice (24). Later studies used a gamma interferon (IFN-γ) capture assay instead of tetramer sorting and estimated 1,100 to 1,500 H-2Db-restricted S510-specific clonotypes and 600 to 900 clonotypes of the subdominant H-2Kb-restricted MHV S598 peptide-specific T cells in the spleens of acutely infected mice (25). Those studies also estimated that there were 1,000 to 1,200 different H-2Db-restricted GP33-specific clonotypes that could respond to an LCMV infection.More-recent studies have taken advantage of magnetic tetramer binding enrichment and double tetramer staining of cells from the spleen and lymph nodes of naïve mice to determine pMHC precursor frequencies, with the assumption that most CD8+ T cells in a naïve mouse reside in lymphoid organs and will react with tetramers. This technique was first described by Moon et al. for CD4+ T cells, and it detected ∼190 I-Ab 2W1S 52-68-specific T cells, ∼20 I-Ab Salmonella enterica serovar Typhimurium FLiC 427-441-specific T cells, and ∼16 I-Ab chicken ovalbumin (OVA) 323-339-specific T cells per mouse (19). This same technique was then used to determine numbers of pMHC-specific CD8+ T cells for epitopes derived from a variety of viruses and found 15 to 1,070 pMHC-specific CD8+ T cells per mouse, depending on the specificity of the pMHC tetramer (10, 15, 23). Determinations of CD8+ T-cell precursor frequencies in humans are currently not experimentally attainable, but exhaustive sequencing of an HLA-A2.1-restricted influenza A virus (IAV) M1 58-66-specific T-cell response has suggested that there are at least 141 different clonotypes that can grow out in response to an in vitro stimulation with peptide, providing a minimum number of T cells that can respond to this pMHC complex in humans (22).Most of the assays estimate the number of T cells specific to single peptides in individual mice. These assays, therefore, do not determine the numbers of CD8+ T cells that can proliferate in response to an entire virus, especially if the virus is known to have many epitopes or if epitopes for the virus have not been described. By examining the average number of pMHC-specific CD8+ T cells in a naïve mouse and comparing this to the number of pMHC-specific CD8+ T cells that are in a mouse at the peak of the T-cell response, it can be calculated that CD8+ T cells divide approximately 12 to 14 times after virus infection (23). Considering that the progeny of one precursor after only 12 divisions can result in just over 4,000 cells, and since recent experiments using H-2Kb-restricted chicken OVA 257-264-specific OT-1-transgenic T cells have confirmed that the progeny from a single cell can be detected in a mouse after infection (31), an in vivo LDA was set up to take advantage of the extensive division and proliferation of virus-specific CD8+ T cells in order to determine virus-specific CD8+ T-cell precursor frequencies.Here, we show that by transferring limiting amounts of carboxyfluorescein succinimidyl ester (CFSE)-labeled Thy1.1+ Ly5.2+ heterogeneous CD8+ T cells into Thy1.2+ Ly5.1+ hosts, we are able to calculate CD8+ T-cell precursor frequencies for whole viruses. Our calculations are based on finding the number of donor CD8+ T cells that results in low-level-CFSE (CFSElo) (i.e., proliferated) donor CD8 T cells in 50% of the hosts. Using probit or Reed and Muench 50% endpoint calculations (3, 26), we are able to calculate CD8+ T-cell precursor frequencies. We show here that frequencies of naïve CD8+ T-cell precursors for whole viruses are quite high and that our in vivo LDA calculates whole-virus precursor frequencies in line with determinations using other methods with naïve and immune mice.  相似文献   

10.
Activation of CD4+ T cells helps establish and sustain other immune responses. We have previously shown that responses against a broad set of nine CD4+ T-cell epitopes were present in the setting of lymphocytic choriomeningitis virus (LCMV) Armstrong infection in the context of H-2d. This is quite disparate to the H-2b setting, where only two epitopes have been identified. We were interested in determining whether a broad set of responses was unique to H-2d or whether additional CD4+ T-cell epitopes could be identified in the setting of the H-2b background. To pursue this question, we infected C57BL/6 mice with LCMV Armstrong and determined the repertoire of CD4+ T-cell responses using overlapping 15-mer peptides corresponding to the LCMV Armstrong sequence. We confirmed positive responses by intracellular cytokine staining and major histocompatibility complex (MHC)-peptide binding assays. A broad repertoire of responses was identified, consisting of six epitopes. These epitopes originate from the nucleoprotein (NP) and glycoprotein (GP). Out of the six newly identified CD4+ epitopes, four of them also stimulate CD8+ T cells in a statistically significant manner. Furthermore, we assessed these CD4+ T-cell responses during the memory phase of LCMV Armstrong infection and after infection with a chronic strain of LCMV and determined that a subset of the responses could be detected under these different conditions. This is the first example of a broad repertoire of shared epitopes between CD4+ and CD8+ T cells in the context of viral infection. These findings demonstrate that immunodominance is a complex phenomenon in the context of helper responses.  相似文献   

11.
Simian immunodeficiency virus (SIV) infection of rhesus macaques (RMs) provides a reliable model to study the relationship between lentivirus replication, cellular immune responses, and CD4+ T-cell dynamics. Here we investigated, using SIVmac251-infected RMs of a Chinese genetic background (which experience a slower disease progression than Indian RMs), the dynamics of CD4+ CCR5+ T cells, as this subset of memory/activated CD4+ T cells is both a preferential target of virus replication and a marker of immune activation. As expected, we observed that the number of circulating CD4+ CCR5+ T cells decreases transiently at the time of peak viremia. However, at 60 days postinfection, i.e., when set-point viremia is established, the level of CD4+ CCR5+ T cells was increased compared to the baseline level. Interestingly, this increase correlated with faster disease progression, higher plasma viremia, and early loss of CD4+ T-cell function, as measured by CD4+ T-cell count, the fraction of memory CD4+ T cells, and the recall response to purified protein derivative. Taken together, these data show a key difference between the dynamics of the CD4+ CCR5+ T-cell pool (and its relationship with disease progression) in Chinese RMs and those described in previous reports for Indian SIVmac251-infected RMs. As the SIV-associated changes in the CD4+ CCR5+ T-cell pool reflect the opposing forces of SIV replication (which reduces this cellular pool) and immune activation (which increases it), our data suggest that in SIV-infected Chinese RMs the impact of immune activation is more prominent than that of virus replication in determining the size of the pool of CD4+ CCR5+ T cells in the periphery. As progression of HIV infection in humans also is associated with a relative expansion of the level of CD4+ CCR5+ T cells, we propose that SIV infection of Chinese RMs is a very valuable and important animal model for understanding the pathogenesis of human immunodeficiency virus infection.  相似文献   

12.
Previous studies determined that the CD8+ T-cell response elicited by recombinant adenovirus exhibited a protracted contraction phase that was associated with long-term presentation of antigen. To gain further insight into this process, a doxycycline-regulated adenovirus was constructed to enable controlled extinction of transgene expression in vivo. We investigated the impact of premature termination of transgene expression at various time points (day 3 to day 60) following immunization. When transgene expression was terminated before the maximum response had been attained, overall expansion was attenuated, yielding a small memory population. When transgene expression was terminated between day 13 and day 30, the memory population was not sustained, demonstrating that the early memory population was antigen dependent. Extinction of transgene expression at day 60 had no obvious impact on memory maintenance, indicating that maintenance of the memory population may ultimately become independent of transgene expression. Premature termination of antigen expression had significant but modest effects on the phenotype and cytokine profile of the memory population. These results offer new insights into the mechanisms of memory CD8+ T-cell maintenance following immunization with a recombinant adenovirus.Recombinant human adenovirus 5 (rHuAd5) vector vaccines have garnered considerable attention as platforms for eliciting CD8+ T-cell immunity due to their strong immunogenicity in numerous studies, including primate studies and preliminary human trials (30, 32, 53). While these vectors may not represent the optimal serotype for use in humans, due to the high prevalence of preexisting immunity, the robust immunogenicity of rHuAd5 in preclinical models merits further investigation, since the biological information derived from these studies will offer important insights that can be extended to other vaccine platforms.CD8+ T cells play an important role in host defense against tumors and viral infections. During the primary phase of the CD8+ T-cell response, the activated precursors undergo a rapid and dramatic expansion in cell number, followed by a period of contraction where 80 to 90% of the antigen-specific population dies off, leaving the remaining cells to constitute the memory population (44). CD8+ T cells mature over the course of the primary response and acquire the ability to produce gamma interferon (IFN-γ), tumor necrosis factor alpha (TNF-α), and, to a lesser degree, interleukin 2 (IL-2). Memory T cells can be divided into central memory and effector memory T cells based on phenotype and anatomical location (44). These phenotypic differences have also been linked to functional differences; however, these relationships remain controversial (2, 16, 20, 46, 55).Various reports have revealed some unexpected qualities of the CD8+ T-cell response generated by intramuscular immunization with rHuAd5. The rHuAd5-induced CD8+ T-cell response exhibited a protracted contraction phase, and the memory population was composed primarily of effector and effector-memory cells (23, 38, 39, 41, 51). The phenotype of the rHuAd5-elicited CD8+ T-cell population was more consistent with the CD8+ T-cell population observed in persistent infections, such as polyomavirus (25), murine herpesvirus-68 (35), and murine cytomegalovirus (MCMV) (1) infections, than with that observed in acute infections, such as lymphocytic choriomeningitis virus (LCMV) (44), vaccinia virus (15), and influenza virus (24) infections. Further investigation demonstrated that, as in a persistent infection, antigen presentation persisted for a prolonged period following intramuscular immunization with rHuAd5, and transgene expression could persist at low levels for more than 1 year following infection (41, 51). These data suggest that the sustained effector phenotype may arise from prolonged, low-level transgene expression from the rHuAd5 vector, although this connection was not formally proven. It is difficult to fully appreciate the implications of these observations at this time, since chronic exposure to antigen is often associated with CD8+ T-cell dysfunction, yet rHuAd5 vectors have been used successfully to elicit protective immunity in many models of pathogen infection and tumor challenge (5, 54). Nevertheless, other reports have provided evidence that rHuAd5 vectors can, indeed, lead to dysfunctional CD8+ T-cell immunity (27, 36). Therefore, further investigation is necessary in order to properly assess the implications of the prolonged antigen expression following rHuAd5 immunization in terms of sustaining a functional memory CD8+ T-cell response.In the current report, we sought to determine the relationship between transgene expression and CD8+ T-cell maintenance and memory. To this end, we constructed an Ad vector with a doxycycline (DOX)-regulated expression cassette that would permit attenuation of gene expression at various times postinfection. Using this reagent, we addressed two key questions. (i) How does the duration of antigen expression affect the magnitude of primary CD8+ T-cell expansion? (ii) Is antigen expression required beyond the peak expansion to maintain the memory CD8+ T-cell population?  相似文献   

13.
Virus-specific CD8+ T cells play a central role in the control of viral infections, including human immunodeficiency virus type 1 (HIV-1) infection. However, despite the presence of strong and broad HIV-specific CD8+ T-cell responses in chronic HIV-1 infection, these cells progressively lose critical effector functions and fail to clear the infection. Mounting evidence suggests that the upregulation of several inhibitory regulatory receptors on the surface of CD8+ T cells during HIV-1 infection may contribute directly to the impairment of T-cell function. Here, we investigated the role of killer immunoglobulin receptors (KIR), which are expressed on NK cells and on CD8+ T cells, in regulating CD8+ T-cell function in HIV-1 infection. KIR expression was progressively upregulated on CD8+ T cells during HIV-1 infection and correlated with the level of viral replication. Expression of KIR was associated with a profound inhibition of cytokine secretion, degranulation, proliferation, and activation by CD8+ T cells following stimulation with T-cell receptor (TCR)-dependent stimuli. In contrast, KIR+ CD8+ T cells responded potently to TCR-independent stimulation, demonstrating that these cells are functionally competent. KIR-associated suppression of CD8+ T-cell function was independent of ligand engagement, suggesting that these regulatory receptors may constitutively repress TCR activation. This ligand-independent repression of TCR activation of KIR+ CD8+ T cells may represent a significant barrier to therapeutic interventions aimed at improving the quality of the HIV-specific CD8+ T-cell response in infected individuals.  相似文献   

14.
The early immune response fails to prevent the establishment of chronic human immunodeficiency virus (HIV) infection but may influence viremia during primary infection, thereby possibly affecting long-term disease progression. CD25+ FoxP3+ regulatory T cells may contribute to HIV/simian immunodeficiency virus (SIV) pathogenesis by suppressing efficient antiviral responses during primary infection, favoring high levels of viral replication and the establishment of chronic infection. In contrast, they may decrease immune activation during chronic infection. CD4+ regulatory T cells have been studied in the most detail, but CD8+ CD25+ FoxP3+ T cells also have regulatory properties. We monitored the dynamics of CD25+ FoxP3+ T cells during primary and chronic SIVmac251 infection in cynomolgus macaques. The number of peripheral CD4+ CD25+ FoxP3+ T cells paralleled that of memory CD4+ T cells, with a rapid decline during primary infection followed by a rebound to levels just below baseline and gradual depletion during the course of infection. No change in the proportion of CD25+ FoxP3+ T cells was observed in peripheral lymph nodes. A small number of CD4+ CD25+ FoxP3+ T cells at set point was associated with a high plasma viral load. In contrast, peripheral CD8+ CD25+ FoxP3+ T cells were induced a few days after peak plasma viral load during primary infection. The number of these cells was positively correlated with viral load and negatively correlated with CD4+ T-cell activation, SIV antigen-specific proliferative responses during primary infection, and plasma viral load at set point, with large numbers of CD8+ CD25+ FoxP3+ T cells being indicative of a poor prognosis.  相似文献   

15.
16.
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   

17.
CD8+ T cells (TCD8+) play a crucial role in immunity to viruses. Antiviral TCD8+ are initially activated by recognition of major histocompatibility complex (MHC) class I-peptide complexes on the surface of professional antigen-presenting cells (pAPC). Migration of pAPC from the site of infection to secondary lymphoid organs is likely required during a natural infection. Migrating pAPC can be directly infected with virus or may internalize antigen derived from virus-infected cells. The use of experimental virus infections to assess the requirement for pAPC migration in initiation of TCD8+ responses has proven difficult to interpret because injected virus can readily drain to secondary lymphoid organs without the need for cell-mediated transport. To overcome this ambiguity, we examined the generation of antigen-specific TCD8+ after immunization with recombinant adenoviruses that express antigen driven by skin-specific or ubiquitous promoters. We show that the induction of TCD8+ in response to tissue-targeted antigen is less efficient than the response to ubiquitously expressed antigen and that the resulting TCD8+ fail to clear all target cells pulsed with the antigenic peptide. This failure to prime a fully functional TCD8+ response results from a reduced period of priming to peripherally expressed antigen versus ubiquitously expressed antigen and correlated with a brief burst of pAPC migration from the skin, a requirement for induction of the response to peripheral antigen. These results indicate that a reduced duration of pAPC migration after virus infection likely reduces the amplitude of the TCD8+ response, allowing persistence of the peripheral virus.The induction of effector CD8+ T cells (TCD8+) is a vital step in the eradication or control of many viral infections. The induction of antiviral TCD8+ requires the presentation of virally derived peptides in complex with major histocompatibility complex (MHC) class I on the surface of specialized professional antigen-presenting cells (pAPC), most commonly a subset of dendritic cells (DC) that bear the CD8α chain (1, 29). The CD8α+ DC reside only in secondary lymphoid organs and not in the tissues, implying that cell-mediated transport or drainage of virus particles to a lymph node is required for initiation of a TCD8+ response. Partial inhibition of DC migration from the skin can impair the initiation of a TCD8+ response (2). After influenza infection in the lungs, there is a burst of DC migration, followed by a refractory period in which no DC migration occurs (19). The functional consequences of this refractory period of DC migration have not been explored.A number of viruses, particularly human papillomaviruses, infect the skin and are ignored by the immune response for extended periods of time (31). We sought to explore the possibility that, after a low-level peripheral virus infection of the skin, changes in DC migration may limit the availability of antigen in the draining lymph node and thus the induction of a TCD8+ response. There are a number of confounding factors that make the study of DC migration in the initiation of an antiviral TCD8+ response difficult. Virus particles may directly drain to the lymph node within seconds (11, 13, 25). In addition, many viruses will alter DC functions, including migration, after infection of the DC itself. This may occur via specific viral modulation of DC function (16) or via nonspecific shut down of host protein synthesis (26), both of which will affect migration. Thus, it is often not possible to distinguish between the effects of virus infection upon DC migration, drainage of virus directly to the lymph node, and the natural response that follows migration of DC responding to a peripheral virus infection.There is currently no mouse model of a peripheral virus infection that is confined to the skin, as no natural mouse papillomavirus has ever been isolated. Therefore, to address these issues, we have made use of another small DNA virus, namely, an adenovirus vector that is replication deficient (rAd). These vectors express influenza virus nucleoprotein (NP) under the control of a ubiquitous (cytomegalovirus [CMV] immediate-early) or tissue-targeted promoter (K14, targeted to keratinocytes, the site of papillomavirus replication). Antigen driven by the K14 promoter is expressed only in skin cells, so only uninfected DC can present antigen in this system, removing the need to account for modulation of the function of virus-infected DC.We demonstrate that when antigen is expressed in only keratinocytes in the skin, the efficiency of TCD8+ induction is reduced and the time period for which antigen is available to prime effector cells is reduced dramatically. DC-mediated transport is required for antigen to reach the lymph node where a TCD8+ response is initiated. The reduced time period of antigen presentation is the result of a transient blockade in DC migration from the site of infection. The blockade in DC migration reduced the delivery of viral antigen to the lymph node needed to induce a TCD8+ response. The resulting TCD8+ response to peripheral viral antigen is not capable of clearing all target cells presenting a viral peptide, thus allowing the persistence of peripheral virus-infected cells. These results provide a potential mechanism for the long-term evasion of the immune response by papillomaviruses following natural infection and also have important implications for tissue targeted gene therapy vectors.  相似文献   

18.
Dendritic cells located at the body surfaces, e.g. skin, respiratory and gastrointestinal tract, play an essential role in the induction of adaptive immune responses to pathogens and inert antigens present at these surfaces. In the respiratory tract, multiple subsets of dendritic cells (RDC) have been identified in both the normal and inflamed lungs. While the importance of RDC in antigen transport from the inflamed or infected respiratory tract to the lymph nodes draining this site is well recognized, the contribution of individual RDC subsets to this process and the precise role of migrant RDC within the lymph nodes in antigen presentation to T cells is not clear. In this report, we demonstrate that two distinct subsets of migrant RDC - exhibiting the CD103+ and CD11bhi phenotype, respectively - are the primary DC presenting antigen to naïve CD4+ and CD8+ T lymphocytes in the draining nodes in response to respiratory influenza virus infection. Furthermore, the migrant CD103+ RDC subset preferentially drives efficient proliferation and differentiation of naive CD8+ T cells responding to infection into effector cells, and only the CD103+ RDC subset can present to naïve CD8+ T cells non-infectious viral vaccine introduced into the respiratory tract. These results identify CD103+ and CD11bhi RDC as critical regulators of the adaptive immune response to respiratory tract infection and potential targets in the design of mucosal vaccines.  相似文献   

19.
A large number of dendritic cell (DC) subsets have now been identified based on the expression of a distinct array of surface markers as well as differences in functional capabilities. More recently, the concept of unique subsets has been extended to the lung, although the functional capabilities of these subsets are only beginning to be explored. Of particular interest are respiratory DCs that express CD103. These cells line the airway and act as sentinels for pathogens that enter the lung, migrating to the draining lymph node, where they add to the already complex array of DC subsets present at this site. Here we assessed the contributions of these individual populations to the generation of a CD8+ T-cell response following respiratory infection with poxvirus. We found that CD103+ DCs were the most effective antigen-presenting cells (APC) for naive CD8+ T-cell activation. Surprisingly, we found no evidence that lymph node-resident or parenchymal DCs could prime virus-specific cells. The increased efficacy of CD103+ DCs was associated with the increased presence of viral antigen as well as high levels of maturation markers. Within the CD103+ DCs, we observed a population that expressed CD8α. Interestingly, cells bearing CD8α were less competent for T-cell activation than their CD8α counterparts. These data show that lung-migrating CD103+ DCs are the major contributors to CD8+ T-cell activation following poxvirus infection. However, the functional capabilities of cells within this population differ with the expression of CD8, suggesting that CD103+ cells may be divided further into distinct subsets.In order for the body to mount an adaptive immune response to a pathogen, T cells circulating through lymph nodes (LN) must be alerted to the presence of infection in the periphery. This occurs as a result of presentation of pathogen-derived epitopes on professional antigen-presenting cells (APC), primarily dendritic cells (DC). The DC that reside in the tissue continually sample the local environment for the presence of foreign/pathogenic antigens. In a noninfected tissue, DC exist in an immature state, i.e., they are highly phagocytic and have low levels of expression of costimulatory molecules (3). Following an encounter with infection-associated signals, e.g., pathogen-associated molecular patterns (PAMPs) and/or inflammatory cytokines, DC undergo maturation (3). This process results in upregulation of chemokine receptors, which promotes trafficking to the lymph node, as well as increased expression of costimulatory molecules and cytokines, which are necessary accessory signals for the activation of naive T cells (2, 3).Unlike many other tissues, the lung is constantly assaulted with foreign antigens, both environmental and infectious. This includes a large number of viruses which spread via aerosolized droplets. As such, it is critical to understand how the immune system detects these infections and subsequently elicits an efficacious adaptive CD8+ T-cell response. While an important role for DC as the activators of naive T cells is clear, the contribution of distinct DC subsets in this process is less understood. Multiple DC subsets are present within the lung draining lymph nodes, and as such, all are potential regulators of T-cell activation (for a review, see references 14 and 32). These subsets are either resident in the lymph node or present at this site as a result of migration from the periphery, in this case, the lung. These DC subsets are defined by the array of molecules expressed at their surface. Among the subsets resident within the lymph nodes are those which express CD8α or CD4 or are double negative (express neither CD4 nor CD8α) (32). These subsets appear to be segregated in their capabilities to elicit T-cell responses. For example, previous studies have suggested that CD8α+ DC are the predominant DC subset involved in priming CD8+ T cells (4), while CD8α CD4+ DC are more important in the regulation of CD4+ T cells (31). Further, CD8α+ DC are efficient at cross-presentation, a property shown to be critical in the generation of CD8+ T-cell responses in a number of infectious models (24, 33).In addition to LN-resident populations, lung-resident DC that have migrated to the lymph node following infection make up a significant portion of LN DC. CD103 (an αE integrin)-expressing DC reside at the airway mucosa and surrounding pulmonary vessels (35). In contrast, CD103 CD11b+ DC are restricted to the lung parenchyma. Given the relatively recent identification of these distinct lung-resident DC populations, there is limited information available regarding their role in T-cell activation following infection. However, they have been assessed in several models, including influenza virus, respiratory syncytial virus (RSV), and Bordetella pertussis (1, 5, 15, 19, 23, 26, 37). At present, the relative contributions of migrating versus resident DC populations remain controversial. Earlier studies reported a role for LN-resident CD8α+ DC in priming naive CD8+ T cells in addition to lung-migrating DC (5). More recently, however, studies have suggested that activating potential is restricted primarily to lung-migrating DC (1, 23). The underlying cause of these discrepancies is currently unknown but may reflect differences in the markers used to identify the DC subsets or in the individual infection models. Regardless, our understanding of the role of these subsets remains incomplete.We have analyzed the migration and maturation of DC following respiratory infection with the orthopoxvirus vaccinia virus (VV). These studies revealed that airway-resident CD103+ DC were the most efficient activators of virus-specific CD8+ T cells. Further studies determined that this was the result of both increased access to viral antigen and increased maturation within this subset. In our analyses, we found no evidence to support a role for LN-resident CD8α+ DC or lung-migrating CD11b+ DC in T-cell activation. Further, we found that CD103+ DC were heterogeneous with regard to their functional capabilities. Interestingly, this correlated with the expression of CD8α. While more-recent studies have found CD8α expression on CD103+ DC (30), none have looked at the functional capabilities of these cells separately from those of CD8α CD103+ DC. Our findings are the first to suggest that CD8α expression within the CD103+ population may identify a distinct subset that differs in its functional capabilities.  相似文献   

20.
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