首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
2.
Vγ9/Vδ2 T cells are a minor subset of T cells in human blood and differ from other T cells by their immediate responsiveness to microbes. We previously demonstrated that the primary target for Vγ9/Vδ2 T cells is (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMB-PP), an essential metabolite produced by a large range of pathogens. Here we wished to study the consequence of this unique responsiveness in microbial infection. The majority of peripheral Vγ9/Vδ2 T cells shares migration properties with circulating monocytes, which explains the presence of these two distinct blood cell types in the inflammatory infiltrate at sites of infection and suggests that they synergize in anti-microbial immune responses. Our present findings demonstrate a rapid and HMB-PP-dependent crosstalk between Vγ9/Vδ2 T cells and autologous monocytes that results in the immediate production of inflammatory mediators including the cytokines interleukin (IL)-6, interferon (IFN)-γ, tumor necrosis factor (TNF)-α, and oncostatin M (OSM); the chemokines CCL2, CXCL8, and CXCL10; and TNF-related apoptosis-inducing ligand (TRAIL). Moreover, under these co-culture conditions monocytes differentiate within 18 hours into inflammatory dendritic cells (DCs) with antigen-presenting functions. Addition of further microbial stimuli (lipopolysaccharide, peptidoglycan) induces CCR7 and enables these inflammatory DCs to trigger the generation of CD4+ effector αβ T cells expressing IFN-γ and/or IL-17. Importantly, our in vitro model replicates the responsiveness to microbes of effluent cells from peritoneal dialysis (PD) patients and translates directly to episodes of acute PD-associated bacterial peritonitis, where Vγ9/Vδ2 T cell numbers and soluble inflammatory mediators are elevated in patients infected with HMB-PP-producing pathogens. Collectively, these findings suggest a direct link between invading pathogens, microbe-responsive γδ T cells, and monocytes in the inflammatory infiltrate, which plays a crucial role in the early response and the generation of microbe-specific immunity.  相似文献   

3.
Type 1 diabetes (T1D) occurs through a breakdown of self-tolerance resulting in the autoimmune destruction of the insulin producing β-islets of the pancreas. A numerical and functional waning of CD4+Foxp3+ regulatory T (Treg) cells, prompted by a pancreatic IL-2 deficiency, accompanies Th1 autoimmunity and T1D progression in non-obese diabetic (NOD) mice. Recently, we identified a dominant subset of intra-islet Treg cells that expresses the ICOS costimulatory receptor and promotes self-tolerance delaying the onset of T1D. ICOS co-stimulation potently enhances IL-2 induced survival and proliferation, and suppressive activity of Treg cells in situ. Here, we propose an ICOS-dependent mechanism of Treg cell homing to the β-islets during pre-diabetes in the NOD model via upregulation of the CXCR3 chemokine receptor. The islet-specific ICOS+ Treg cell subset preferentially expresses CXCR3 in the pancreatic lymph nodes (pLN) in response to Teff cell-mediated pancreatic inflammation, an expression correlating with the onset and magnitude of IFN-γ production by Teff cells in pancreatic sites. We also reveal that intra-pancreatic APC populations and insulin-producing β, but not α nor δ, islet cells secrete the CXCR3 chemokines, CXCL9, 10 and 11, and selectively promote ICOS+CXCR3+ Treg cell chemotaxis in vitro. Strikingly, islet-derived Treg cells also produce these chemokines suggesting an auto-regulation of homing by this subset. Unlike ICOS- cells, ICOS+ Treg cells adopt a Th1-like Treg phenotype while maintaining their suppressive capacity, characterized by expression of T-bet and CXCR3 and production of IFN-γ in the draining pLNs. Finally, in vivo neutralization of IFN-γ blocked Treg cell CXCR3 upregulation evincing its role in regulating expression of this chemokine receptor by Treg cells. Thus, CXCR3-mediated trafficking of Treg cells could represent a mechanism of homeostatic immunoregulation during diabetogeneesis.  相似文献   

4.
The objective of this study was to functionally assess gamma/delta (γδ) T cells following pathogenic human immunodeficiency virus (HIV) infection of humans and nonpathogenic simian immunodeficiency virus (SIV) infection of sooty mangabeys. γδ T cells were obtained from peripheral blood samples from patients and sooty mangabeys that exhibited either a CD4-healthy (>200 CD4+ T cells/μl blood) or CD4-low (<200 CD4 cells/μl blood) phenotype. Cytokine flow cytometry was utilized to assess production of Th1 cytokines tumor necrosis factor alpha and gamma interferon following ex vivo stimulation with either phorbol myristate acetate/ionomycin or the Vδ2 γδ T-cell receptor agonist isopentenyl pyrophosphate. Sooty mangabeys were observed to have higher percentages of γδ T cells in their peripheral blood than humans did. Following stimulation, γδ T cells from SIV-positive (SIV+) mangabeys maintained or increased their ability to express the Th1 cytokines regardless of CD4+ T-cell levels. In contrast, HIV-positive (HIV+) patients exhibited a decreased percentage of γδ T cells expressing Th1 cytokines following stimulation. This dysfunction is primarily within the Vδ2+ γδ T-cell subset which incurred both a decreased overall level in the blood and a reduced Th1 cytokine production. Patients treated with highly active antiretroviral therapy exhibited a partial restoration in their γδ T-cell Th1 cytokine response that was intermediate between the responses of the uninfected and HIV+ patients. The SIV+ sooty mangabey natural hosts, which do not proceed to clinical AIDS, provide evidence that γδ T-cell dysfunction occurs in HIV+ patients and may contribute to HIV disease progression.  相似文献   

5.

Background

Double negative CD3+48 TCRαβ splenic cells (DNCD3) can suppress the immune responses to allo and xenografts, infectious agents, tumors, and some autoimmune disorders. However, little is known about their role in autoimmune diabetes, a disease characterized by the reduction of insulin production subsequent to destruction of pancreatic β-cells by a polyclonal population of self-reactive T-cells. Herein, we analyzed the function and phenotype of DNCD3 splenic cells in young NOD mice predisposed to several autoimmune disorders among which, the human-like autoimmune diabetes.

Methodology/Principal Findings

DNCD3 splenic cells from young NOD mice (1) provided long-lasting protection against diabetes transfer in NOD/Scid immunodeficient mice, (2) proliferated and differentiated in the spleen and pancreas of NOD/Scid mice and pre-diabetic NOD mice into IL-10-secreting TR-1 like cells in a Th2-like environment, and (3) their anti-diabetogenic phenotype is CD3+(CD4CD8)CD28+CD69+CD25low Foxp3 iCTLA-4TCRαβ+ with a predominant Vβ13 gene usage.

Conclusions/Significance

These findings delineate a new T regulatory component in autoimmune diabetes apart from that of NKT and CD4+CD25high Foxp3+T-regulatory cells. DNCD3 splenic cells could be potentially manipulated towards the development of autologous cell therapies in autoimmune diabetes.  相似文献   

6.
In mice implanted with an osmotic pump filled with the superantigen (SAG) staphylococcal enterotoxin A (SEA), the Vβ3+CD4+ T cells exhibited a high level of expansion whereas the Vβ11+CD4+ T cells exhibited a mild level of expansion. In contrast, in mice implanted with an osmotic pump filled with SE-like type P (SElP, 78.1% homologous with SEA), the Vβ11+CD4+ T cells exhibited a high level of expansion while the Vβ3+CD4+ T cells exhibited a low level of expansion, suggesting that the level of the SAG-induced response is determined by the affinities between the TCR Vβ molecules and SAG. Analyses using several hybrids of SEA and SElP showed that residue 206 of SEA determines the response levels of Vβ3+CD4+ and Vβ11+CD4+ T cells both in vitro and in vivo. Analyses using the above-mentioned hybrids showed that the binding affinities between SEA and the Vβ3/Vβ11 β chains and between SEA-MHC class II-molecule complex and Vβ3+/Vβ11+ CD4+ T cells determines the response levels of the SAG-reactive T cells both in vitro and in vivo.  相似文献   

7.
γδ T cells express several different toll-like receptor (TLR)s. The role of MyD88- dependent TLR signaling in TCR activation of murine γδ T cells is incompletely defined. Here, we report that Pam3CSK4 (PAM, TLR2 agonist) and CL097 (TLR7 agonist), but not lipopolysaccharide (TLR4 agonist), increased CD69 expression and Th1-type cytokine production upon anti-CD3 stimulation of γδ T cells from young adult mice (6-to 10-week-old). However, these agonists alone did not induce γδ T cell activation. Additionally, we noted that neither PAM nor CL097 synergized with anti-CD3 in inducing CD69 expression on γδ T cells of aged mice (21-to 22-month-old). Compared to young γδ T cells, PAM and CL097 increased Th-1 type cytokine production with a lower magnitude from anti-CD3- stimulated, aged γδ T cells. Vγ1+ and Vγ4+ cells are two subpopulations of splenic γδ T cells. PAM had similar effects in anti-CD3-activated control and Vγ4+ subset- depleted γδ T cells; whereas CL097 induced more IFN-γ production from Vγ4+ subset-depleted γδ T cells than from the control group. Finally, we studied the role of MyD88-dependent TLRs in γδ T cell activation during West Nile virus (WNV) infection. γδ T cell, in particular, Vγ1+ subset expansion was significantly reduced in both MyD88- and TLR7- deficient mice. Treatment with TLR7 agonist induced more Vγ1+ cell expansion in wild-type mice during WNV infection. In summary, these results suggest that MyD88-dependent TLRs provide co-stimulatory signals during TCR activation of γδ T cells and these have differential effects on distinct subsets.  相似文献   

8.

Background

IL-2 has been reported to be critical for peripheral Treg survival in mouse models. Here, we examined Treg maintenance in a series of paediatric liver transplant recipients who received basiliximab, a therapeutic anti-CD25 monoclonal antibody.

Methodology/Principal Findings

FoxP3+ CD4 T cells were analyzed by flow cytometry before liver grafting and more than 9 months later. We found that in vivo CD25 blockade did not lead to Treg depletion: the proportion of FoxP3+ cells among CD4 T cells and the level of FoxP3 expression were both unchanged. IL-2Rβ expression was enhanced in FoxP3+ cells both before and after basiliximab treatment, while the level of IL-2Rγ expression was similar in Tregs and non-Tregs. No significant change in the weak or absent expression of IL-7Rα and IL-15Rα expression on FoxP3+ cells was observed. Although the proportion of FoxP3+ cells among CD4 T cells did not vary, food allergies occurred more rapidly after liver grafting in patients who received basiliximab, raising questions as to Treg functionality in vivo in the absence of functional CD25.

Conclusions

CD25 appears non essential for human Treg peripheral maintenance in vivo. However, our results raise questions as to Treg functionality after therapeutic CD25 targeting.  相似文献   

9.

Background

Antigen-specific IFN-γ producing CD4+ T cells are the main mediators of protection against Mycobacterium tuberculosis infection both under natural conditions and following vaccination. However these cells are responsible for lung damage and poor vaccine efficacy when not tightly controlled. Discovering new tools to control nonprotective antigen-specific IFN-γ production without affecting protective IFN-γ is a challenge in tuberculosis research.

Methods and Findings

Immunization with DNA encoding Ag85B, a candidate vaccine antigen of Mycobacterium tuberculosis, elicited in mice a low but protective CD4+ T cell-mediated IFN-γ response, while in mice primed with DNA and boosted with Ag85B protein a massive increase in IFN-γ response was associated with loss of protection. Both protective and non-protective Ag85B-immunization generated antigen-specific CD8+ T cells which suppressed IFN-γ-secreting CD4+ T cells. However, ex vivo ligation of 4-1BB, a member of TNF-receptor super-family, reduced the massive, non-protective IFN-γ responses by CD4+ T cells in protein-boosted mice without affecting the low protective IFN-γ-secretion in mice immunized with DNA. This selective inhibition was due to the induction of 4-1BB exclusively on CD8+ T cells of DNA-primed and protein-boosted mice following Ag85B protein stimulation. The 4-1BB-mediated IFN-γ inhibition did not require soluble IL-10, TGF-β, XCL-1 and MIP-1β. In vivo Ag85B stimulation induced 4-1BB expression on CD8+ T cells and in vivo 4-1BB ligation reduced the activation, IFN-γ production and expansion of Ag85B-specific CD4+ T cells of DNA-primed and protein-boosted mice.

Conclusion/Significance

Antigen-specific suppressor CD8+ T cells are elicited through immunization with the mycobacterial antigen Ag85B. Ligation of 4-1BB receptor further enhanced their suppressive activity on IFN-γ-secreting CD4+ T cells. The selective expression of 4-1BB only on CD8+ T cells in mice developing a massive, non-protective IFN-γ response opens novel strategies for intervention in tuberculosis pathology and vaccination through T-cell co-stimulatory-based molecular targeting.  相似文献   

10.

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   

11.
Characterization of immune responses induced by live attenuated simian immunodeficiency virus (SIV) strains may yield clues to the nature of protective immunity induced by this vaccine approach. We investigated the ability of CD8+ T lymphocytes from rhesus macaques immunized with the live, attenuated SIV strain SIVmac239Δnef or SIVmac239Δ3 to inhibit SIV replication. CD8+ T lymphocytes from immunized animals were able to potently suppress SIV replication in autologous SIV-infected CD4+ T cells. Suppression of SIV replication by unstimulated CD8+ T cells required direct contact and was major histocompatibility complex (MHC) restricted. However, CD3-stimulated CD8+ T cells produced soluble factors that inhibited SIV replication in an MHC-unrestricted fashion as much as 30-fold. Supernatants from stimulated CD8+ T cells were also able to inhibit replication of both CCR5- and CXCR4-dependent human immunodeficiency virus type 1 (HIV-1) strains. Stimulation of CD8+ cells with cognate cytotoxic T-lymphocyte epitopes also induced secretion of soluble factors able to inhibit SIV replication. Production of RANTES, macrophage inhibitory protein 1α (MIP-1α), or MIP-1β from stimulated CD8+ T cells of vaccinated animals was almost 10-fold higher than that from stimulated CD8+ T cells of control animals. However, addition of antibodies that neutralize these β-chemokines, either alone or in combination, only partly blocked inhibition of SIV and HIV replication by soluble factors produced by stimulated CD8+ T cells. Our results indicate that inhibition of SIV replication by CD8+ T cells from animals immunized with live attenuated SIV strains involves both MHC-restricted and -unrestricted mechanisms and that MHC-unrestricted inhibition of SIV replication is due principally to soluble factors other than RANTES, MIP-1α, and MIP-1β.  相似文献   

12.

Introduction

C-X-C motif chemokine 10 (CXCL10) is a chemokine that plays a critical role in the infiltration of T cells in autoimmune diseases and is reported to be expressed in muscle tissue of polymyositis. To determine the therapeutic efficacy of CXCL10 blockade, we investigated the role of CXCL10 and the effect of anti-CXCL10 antibody treatment in C protein-induced myositis (CIM), an animal model of polymyositis.

Methods

CIM was induced with human skeletal muscle C protein fragment in female C57BL/6 mice. Immunohistochemistry of CXCL10 and C-X-C motif chemokine receptor 3 (CXCR3) and measurement of serum CXCL10 were performed. Cell surface markers and interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) in CIM lymph node cells was investigated by flow cytometry. Mice with CIM were treated with anti-CXCL10 antibody or control antibody (anti-RVG1) and the inflammation in muscle tissue was assessed.

Results

Immunohistochemistry showed increased expression of CXCL10 and CXCR3 in the inflammatory lesions of muscle in CIM. Especially, CD8+ T cells invading myofiber expressed CXCR3. Serum level of CXCL10 was increased in CIM compared to the level in normal mice (normal mouse, 14.3 ± 5.3 pg/ml vs. CIM, 368.5 ± 135.6 pg/ml, P < 0.001). CXCR3 positivity in CD8+ T cells was increased compared to that of CD4+ T cells in the lymph node cells of CIM (CXCR3+ among CD8+ T cell, 65.9 ± 2.1% vs. CXCR3+ among CD4+ T cell, 23.5 ± 4.7%, P <0.001). Moreover, IFN-γ+ cells were increased among CXCR3+CD8+ T cells compared to CXCR3–CD8+ T cells (CXCR3+CD8+ T cell, 28.0 ± 4.2% vs. CXCR3-CD8+ T cell, 9.5 ± 1.5%, P = 0.016). Migration of lymph node cells was increased in response to CXCL10 (chemotactic index was 1.91 ± 0.45). CIM mice treated with anti-CXCL10 antibody showed a lower inflammation score in muscles than those with anti-RVG1 (median, anti-CXCL10 treatment group, 0.625 vs. anti-RVG1 treatment group, 1.25, P = 0.007).

Conclusions

CXCL10/CXCR3 expression was increased in the inflammation of CIM model and its blockade suppressed inflammation in muscle.  相似文献   

13.

Background

Sandhoff disease is a lysosomal storage disorder characterized by the absence of β-hexosaminidase and storage of GM2 ganglioside and related glycolipids. We have previously found that the progressive neurologic disease induced in Hexb −/− mice, an animal model for Sandhoff disease, is associated with the production of pathogenic anti-glycolipid autoantibodies.

Methodology/Principal Findings

In our current study, we report on the alterations in the thymus during the development of mild to severe progressive neurologic disease. The thymus from Hexb −/− mice of greater than 15 weeks of age showed a marked decrease in the percentage of immature CD4+/CD8+ T cells and a significantly increased number of CD4+/CD8 T cells. During involution, the levels of both apoptotic thymic cells and IgG deposits to T cells were found to have increased, whilst swollen macrophages were prominently observed, particularly in the cortex. We employed cDNA microarray analysis to monitor gene expression during the involution process and found that genes associated with the immune responses were upregulated, particularly those expressed in macrophages. CXCL13 was one of these upregulated genes and is expressed specifically in the thymus. B1 cells were also found to have increased in the thy mus. It is significant that these alterations in the thymus were reduced in FcRγ additionally disrupted Hexb −/− mice.

Conclusions/Significance

These results suggest that the FcRγ chain may render the usually poorly immunogenic thymus into an organ prone to autoimmune responses, including the chemotaxis of B1 cells toward CXCL13.  相似文献   

14.
In vitro evidence suggests that memory CD4+ cells are preferentially infected by human immunodeficiency virus type 1 (HIV-1), yet studies of HIV-1-infected individuals have failed to detect preferential memory cell depletion. To explore this paradox, we stimulated CD45RA+ CD4+ (naïve) and CD45RO+ CD4+ (memory) cells with antibodies to CD3 and CD28 and infected them with either CCR5-dependent (R5) or CXCR4-dependent (X4) HIV-1 isolates. Naïve CD4+ cells supported less X4 HIV replication than their memory counterparts. However, naïve cells were susceptible to R5 viral infection, while memory cells remained resistant to infection and viral replication. As with the unseparated cells, mixing the naïve and memory cells prior to infection resulted in cells resistant to R5 infection and highly susceptible to X4 infection. While both naïve and memory CD4+ subsets downregulated CCR5 expression in response to CD28 costimulation, only the memory cells produced high levels of the β-chemokines RANTES, MIP-1α, and MIP-1β upon stimulation. Neutralization of these β-chemokines rendered memory CD4+ cells highly sensitive to infection with R5 HIV-1 isolates, indicating that downregulation of CCR5 is not sufficient to mediate complete protection from CCR5 strains of HIV-1. These results indicate that susceptibility to R5 HIV-1 isolates is determined not only by the level of CCR5 expression but also by the balance of CCR5 expression and β-chemokine production. Furthermore, our results suggest a model of HIV-1 transmission and pathogenesis in which naïve rather than memory CD4+ T cells serve as the targets for early rounds of HIV-1 replication.  相似文献   

15.
Induction of antigen-specific CD8+ T cells bearing a high-avidity T-cell receptor (TCR) is thought to be an important factor in antiviral and antitumor immune responses. However, the relationship between TCR diversity and functional avidity of epitope-specific CD8+ T cells accumulating in the central nervous system (CNS) during viral infection is unknown. Hence, analysis of T-cell diversity at the clonal level is important to understand the fate and function of virus-specific CD8+ T cells. In this study, we examined the Vβ diversity and avidity of CD8+ T cells specific to the predominant epitope (VP2121-130) of Theiler''s murine encephalomyelitis virus. We found that Vβ6+ CD8+ T cells, associated with epitope specificity, predominantly expanded in the CNS during viral infection. Further investigations of antigen-specific Vβ6+ CD8+ T cells by CDR3 spectratyping and sequencing indicated that distinct T-cell clonotypes are preferentially increased in the CNS compared to the periphery. Among the epitope-specific Vβ6+ CD8+ T cells, MGX-Jβ1.1 motif-bearing cells, which could be found at a high precursor frequency in naïve mice, were expanded in the CNS and tightly associated with gamma interferon production. These T cells displayed moderate avidity for the cognate epitope rather than the high avidity normally observed in memory/effector T cells. Therefore, our findings provide new insights into the CD8+ T-cell repertoire during immune responses to viral infection in the CNS.Theiler''s murine encephalomyelitis virus (TMEV) is a member of the Cardiovirus genus within the Picornaviridae family (43). This virus is a common enteric pathogen among wild mice but rarely causes neurological disease (57). However, when it infects susceptible mice (e.g., the SJL/J [SJL] strain) intracerebrally, it reproducibly induces a chronic immune-mediated demyelinating disease that has been studied as an infectious model of human multiple sclerosis (MS) (10, 30). In contrast, infection of resistant mice like those of the C57BL/6 (B6) strain results in strong antiviral immune responses that clear the virus effectively and prevent disease development (24, 31). Therefore, immune responses in B6 mice have been often compared to those in susceptible SJL mice to understand the nature of protective versus pathogenic immunity in these mice.It has been shown that the major histocompatibility complex (MHC) H-2D locus is a critical genetic factor for resistance to TMEV-induced demyelinating disease (9, 49). For example, expression of the H-2Db transgene makes susceptible FVB mice resistant by inducing strong H-2Db-restricted VP2121-130-specific CD8+ T-cell responses (36). This acquired resistance is abolished when VP2121-130-specific T cells are tolerized by introducing the VP2 transgene (45). These results strongly suggest that CD8+ T cells generated in the presence of H-2Db are critical for viral clearance from the central nervous system (CNS). Since the cardinal difference between the resistant B6 and susceptible SJL strains is the quantity, not the quality, of virus-specific CD8+ T cells (23, 32), strong CD8+ T-cell responses are probably required to prevent viral persistence and the consequent development of demyelinating disease. More than threefold more virus-specific CD8+ T cells were found in the CNSs of resistant B6 mice than in those of susceptible SJL mice at the acute phase of infection. Thus, the level of virus-specific CD8+ T cells at an early phase of the immune response may be a critical factor in resistance to the disease.Many recent investigations indicate that oligoclonal CD8+ T cells accumulate in the CNSs of MS patients (4, 38, 51). In addition, CD8+ T cells may also induce the development of experimental autoimmune encephalomyelitis (EAE) (54). Therefore, clonal expansion of certain CD8+ T cells may be associated with the pathogenesis of demyelinating diseases. However, B6 mice, which are resistant to TMEV-induced demyelinating disease, induce strong CD8+ T-cell responses to a single predominant epitope (VP2121-130), i.e., ≥70% of CNS-infiltrating CD8+ T cells (41, 42). These CD8+ T cells result in effective viral clearance yet remain at a low level in the CNS more than 120 days postinfection (dpi) without detectable pathology (42). This inconsistency led us to investigate the shape and quality of the T-cell receptor (TCR) repertoire accumulating in the CNSs of B6 mice.The CD8+ T-cell responses induced after viral infection have previously been investigated with other animal viruses, including influenza virus, lymphocytic choriomeningitis virus (LCMV), mouse hepatitis virus (MHV), and Borna disease virus (11, 14, 35, 47, 58). Among these models, the detailed T-cell Vβ repertoire in the CNS was described only in the MHV model (46). CD8+ T-cell responses against TMEV in B6 mice are primarily against a single predominant epitope (22, 36, 41). However, virtually no study of the TCR Vβ repertoires of virus-specific CD8+ T cells has been reported. Furthermore, it is not yet known whether a particular TCR Vβ repertoire is associated with the avidity and/or function of CD8+ T cells in the CNS. Since protective versus pathogenic CD8+ T cells may correlate with their Vβ repertoire and T-cell function, these studies may help to elucidate the underlying mechanisms of protection versus pathogenesis of CD8+ T cells in the CNS.In this study, we have addressed several important questions about the CD8+ T-cell repertoire in the CNS. First, what is the pattern of Vβ usage in TMEV-infected B6 mice? Second, are there differences in the antigen-specific CD8+ T-cell clonotypes between the CNS and periphery? Third, are the T-cell clonotypes maintained in the CNS during the viral infection? Fourth, what is the functional avidity of T cells accumulating in the CNS during this virus infection? Last, what possible factors are associated with repertoire selection and expansion in the CNS? Our results show that Vβ6+ CD8+ T cells preferentially expand in the CNS during viral infection. Further analyses of the CDR3 region of antigen-specific Vβ6+ CD8+ T cells by spectratyping and sequencing indicate that distinct T-cell clonotypes are expanded in the CNS compared to those in the periphery. T cells expressing a particular Vβ6-CDR3-Jβ1.1 sequence are preferentially retained in the CNS during the course of viral infection. Interestingly, these T cells are capable of producing gamma interferon (IFN-γ) upon stimulation and display moderate avidity for the cognate epitope. We believe that our findings will provide important information regarding the CD8+ T-cell repertoire during viral infection and that these results may help to provide a better understanding of antiviral CD8+ T-cell immunity in the CNS.  相似文献   

16.
Emergence of a new influenza strain leads to a rapid global spread of the virus due to minimal antibody immunity. Pre-existing CD8+ T-cell immunity directed towards conserved internal viral regions can greatly ameliorate the disease. However, mutational escape within the T cell epitopes is a substantial issue for virus control and vaccine design. Although mutations can result in a loss of T cell recognition, some variants generate cross-reactive T cell responses. In this study, we used reverse genetics to modify the influenza NP336–374 peptide at a partially-solvent exposed residue (N->A, NPN3A mutation) to assess the availability, effectiveness and mechanism underlying influenza-specific cross-reactive T cell responses. The engineered virus induced a diminished CD8+ T cell response and selected a narrowed T cell receptor (TCR) repertoire within two Vβ regions (Vβ8.3 and Vβ9). This can be partially explained by the H-2DbNPN3A structure that showed a loss of several contacts between the NPN3A peptide and H-2Db, including a contact with His155, a position known to play an important role in mediating TCR-pMHC-I interactions. Despite these differences, common cross-reactive TCRs were detected in both the naïve and immune NPN3A-specific TCR repertoires. However, while the NPN3A epitope primes memory T-cells that give an equivalent recall response to the mutant or wild-type (wt) virus, both are markedly lower than wt->wt challenge. Such decreased CD8+ responses elicited after heterologous challenge resulted in delayed viral clearance from the infected lung. Furthermore, mice first exposed to the wt virus give a poor, low avidity response following secondary infection with the mutant. Thus, the protective efficacy of cross-reactive CD8+ T cells recognising mutant viral epitopes depend on peptide-MHC-I structural interactions and functional avidity. Our study does not support vaccine strategies that include immunization against commonly selected cross-reactive variants with mutations at partially-solvent exposed residues that have characteristics comparable to NPN3A.  相似文献   

17.
Induction of a functional subset of HIV-specific CD4+ T cells that is resistant to HIV infection could enhance immune protection and decrease the rate of HIV disease progression. CMV-specific CD4+ T cells, which are less frequently infected than HIV-specific CD4+ T cells, are a model for such an effect. To determine the mechanism of this protection, we compared the functional response of HIV gag-specific and CMV pp65-specific CD4+ T cells in individuals co-infected with CMV and HIV. We found that CMV-specific CD4+ T cells rapidly up-regulated production of MIP-1α and MIP-1β mRNA, resulting in a rapid increase in production of MIP-1α and MIP-1β after cognate antigen stimulation. Production of β-chemokines was associated with maturational phenotype and was rarely seen in HIV-specific CD4+ T cells. To test whether production of β-chemokines by CD4+ T cells lowers their susceptibility to HIV infection, we measured cell-associated Gag DNA to assess the in vivo infection history of CMV-specific CD4+ T cells. We found that CMV-specific CD4+ T cells which produced MIP-1β contained 10 times less Gag DNA than did those which failed to produce MIP-1β. These data suggest that CD4+ T cells which produce MIP-1α and MIP-1β bind these chemokines in an autocrine fashion which decreases the risk of in vivo HIV infection.  相似文献   

18.
Interleukin-2 (IL-2) has been implicated as being necessary for the optimal formation of primary CD8+ T cell responses against various pathogens. Here we have examined the role that IL-2 signaling plays in several aspects of a CD8+ T cell response against murine gammaherpesvirus 68 (MHV-68). Exposure to MHV-68 causes a persistent infection, along with infectious mononucleosis, providing a model for studying these processes in mice. Our study indicates that CD25 is necessary for optimal expansion of the antigen-specific CD8+ T cell response but not for the long-term memory response. Contrastingly, IL-2 signaling through CD25 is absolutely required for CD8+ T cell mononucleosis.Members of the gammaherpesvirus family are associated with significant diseases, such as nasopharyngeal carcinoma, lymphoid malignancies, and infectious mononucleosis (16). Murine gammaherpesvirus 68 (MHV-68) is a γ2-herpesvirus related to the human pathogens Epstein-Barr virus (EBV) and Kaposi''s sarcoma virus (19, 21). Intranasal (i.n.) infection of mice with MHV-68 results in acute infection of the lung epithelium, which is eventually controlled; however, the virus also establishes a latent infection in B cells, dendritic cells, and macrophages that is maintained throughout the life of the host (8, 9). Infection with MHV-68 generates a broad array of antigen-specific CD8+ T cells that can control the virus without eliminating persistent infection (5, 12, 13). Additionally, CD4+ T cells and neutralizing antibodies are thought to be critical for the prevention of virus reactivation (3, 6).A major complication of EBV infection is infectious mononucleosis (16), which occurs when infection is delayed until puberty. Signs of disease include dramatic lymph node enlargement and the presence of large numbers of activated CD8+ T cells in the peripheral blood. Similarly to EBV infection, MHV-68 induces a polyclonal activation of B cells upon establishment of latency. Concurrently, a CD8+ T cell-dominated lymphocytosis of the peripheral blood occurs, as seen with EBV. However, there are distinct differences between the two types of infectious mononucleosis. CD8+ T cell lymphocytosis seen with EBV consists of a broad array of T cell receptor specificities, a large proportion of which are specific for EBV epitopes. In contrast, MHV-68-induced mononucleosis is dominated by oligoclonal Vβ4+ CD8+ T cells that are not reactive to MHV-68 epitopes. With MHV-68, the expansion of this population is dramatic, with levels reaching upwards of 60% of the peripheral blood CD8+ T cell population (20). This occurs in different mouse strains, across at least five different major histocompatibility complex (MHC) class I haplotypes. However, it is important to note that infection of wood mice (Apodemus sylvaticus) does not induce splenomegaly, as seen with laboratory strains of mice, indicating a potential lack of Vβ4 expansion that may be species related (14). Interestingly, evidence suggests that Vβ4+ CD8+ T cell expansion does not require classical MHC class Ia antigen presentation (4). Recent studies instead implicate a secreted viral protein, M1, capable of stimulating the Vβ4+ T cell population in a novel manner, and the authors propose a role for Vβ4+ T cells in control of MHV-68 infection (7).We and others have recently shown that IL-2 signaling during the early stages of a response to acute viral and bacterial pathogens is required for optimal expansion and differentiation of CD8+ T cells (15, 17, 18). However, reports with other viruses have shown IL-2-independent primary CD8+ T cell responses (1, 22). Therefore, we wished to determine whether IL-2 signals are necessary for the expansion, maintenance, and/or recall of CD8+ T cell responses during murine gammaherpesvirus infection.We generated chimeric mice through lethal irradiation of C57BL/6 mice followed by adoptive transfer of mixed bone marrow from C57BL/6 wild-type (WT) and CD25−/− donors, as previously described (17). Following previous described protocols, mice were given bone marrow in a 2:1 ratio of CD25−/−/WT to generate equally proportioned congenic populations in recipient mice (see Fig. S1 in the supplemental material) (1, 17). The resultant mice contained CD8+ T cells of both WT and CD25−/− origin, which could be distinguished by congenic markers. Chimeric mice were infected intranasally with 400 PFU of MHV-68, and the kinetics of the CD8+ T cell response were followed by antibody and tetramer staining of peripheral blood for CD8+ T cells specific for the epitopes ORF6487 (p56) and ORF61524 (p79), as previously described (13). While antigen-specific CD25−/− CD8+ T cells were initially able to proliferate in response to infection, the peak response was significantly lower than that of the wild-type cells (Fig. (Fig.11 A and B). This indicates that while CD25 is dispensable for early activation of CD8+ T cells, IL-2 signaling is required for full expansion of the antigen-specific response to MHV-68. Despite this deficit in the acute antiviral response, the resultant memory populations were not statistically different between the groups (Fig. 1A and B). In our previous report, CD25−/− CD8+ T cells were unable to fully differentiate into short-lived effector cells (SLECs), defined as KLRG1high CD127low (17). To determine if MHV-68-specific responses were also unable to fully differentiate, we infected chimeric mice and stained p79+ CD8+ T cells for the cell surface markers KLRG1 and CD127. At the peak of the response (14 days postinfection [p.i.]), p79+ WT cells had differentiated into SLEC (KLRG1high CD127low), memory precursor (MPEC) (KLRG1low CD127high), and doubly positive populations. However, the p79+ CD25−/− cells failed to form the SLEC population and instead had a corresponding increase in the MPEC population, indicating that CD25 is necessary for full effector differentiation of gammaherpesvirus-specific CD8+ T cell responses (Fig. 1C and D).Open in a separate windowFIG. 1.IL-2 signals are necessary for the optimal expansion of MHV-68-specific CD8+ T cells. WT/CD25−/− chimeric mice were infected with MHV-68 intranasally and bled at set time points. The antigen-specific responses against two dominant epitopes, p79 (A) and p56 (B), were determined via tetramer staining of peripheral blood. p79-specific CD8+ T cells from the WT and CD25−/− populations were stained at the peak of the response (day 14 p.i.) for KLRG1 and CD127 to determine their ability to differentiate into short-lived and memory precursor effector cells (C and D). *, P < 0.05; **, P < 0.01; ***, P < 0.001. Error bars represent standard deviations from the means. Four mice were used per group, and data are representative of at least two experiments.To determine whether antigen-specific CD25−/− CD8+ T cells were capable of optimally responding to a secondary challenge, we infected chimeric mice with MHV-68 and waited 60 days before challenging with recombinant vaccinia virus (rVV) expressing the ORF61524 epitope (2 × 106 PFU, intraperitoneal). It is necessary to use a heterologous virus to induce a recall CD8+ T cell response since MHV-68 generates a robust neutralizing antibody response, preventing secondary infection. Previous studies with rVV indicate that the recall response of MHV-68-specific CD8+ T cells is antigen dependent, since administration of rVV expressing an irrelevant epitope had no effect upon the MHV-68-specific populations (2). WT and CD25−/− cells were able to respond to the secondary challenge with similar kinetics (Fig. (Fig.22 A and B), indicating that MHV-68 memory CD8+ T cells are capable of a generating a recall response in the absence of IL-2 signaling. These data, together with our previous report (17), show that the dependence on CD25 for formation of the SLEC population is conserved between both persistent and acute virus infections.Open in a separate windowFIG. 2.CD25−/− CD8+ T cells can respond to secondary challenge. WT/CD25−/− chimeric mice were infected with MHV-68 i.n. After 60 days, the percentage of peripheral blood CD8+ T cells specific for p79 was determined. Mice were then challenged with rVV p79, and the p79+ CD8+ population was determined 5 days postchallenge (A). The numbers in the box represent the averages ± standard deviations. The average fold increase was calculated to determine the ability of WT and CD25−/− CD8+ T cells to respond to a secondary challenge (B). Error bars represent standard deviations from the means. Four mice were used per group, and data are representative of at least two experiments.WT CD8+ T cells underwent a dramatic expansion between days 15 and 21 p.i. (Fig. (Fig.3A),3A), consistent with infectious mononucleosis (10). Interestingly, we did not observe a similar expansion of CD25−/− CD8+ T cells, indicating a role for IL-2 signaling in the expansion of CD8+ T cells during mononucleosis (Fig. (Fig.3A).3A). Since mononucleosis is dominated by Vβ4+ CD8+ T cells, we analyzed these T cells from both naive and infected mice (17 days p.i.) for expression of CD25 by flow cytometry. While Vβ4+ CD8+ T cells from the spleen and peripheral blood of naive mice did not express detectable levels of CD25, mice infected with MHV-68 expressed intermediate levels of CD25 during the time period when dramatic expansion of Vβ4+ T cells occurs (Fig. 3B and C). Consistent with a role for IL-2 signaling in Vβ4 expansion, we observed a severe deficit in expansion in the CD25−/− population of chimeric mice, since the percentage of WT Vβ4+ cells increased dramatically between days 14 and 36 p.i., accompanied by only a small expansion of the CD25−/− Vβ4+ population over the same period (Fig. (Fig.44 A and B).Open in a separate windowFIG. 3.Vβ4+ CD8+ T cells express CD25 upon infection with MHV-68. WT/CD25−/− chimeric mice were infected with MHV-68 i.n., and the percentage of peripheral blood cells that were CD8+ was determined over time for each congenic population (A). Vβ4+ CD8+ T cells from naive and MHV-68-infected mice (day 17 p.i.) were analyzed for expression of CD25 (B and C). Isotype control, filled histogram; naive mice, dashed line; infected mice, solid line (**, P < 0.01). Error bars represent standard deviations from the means. Four mice were used per group, and data are representative of at least two experiments.Open in a separate windowFIG. 4.CD8+ T cell-based infectious mononucleosis does not occur in the absence of IL-2 signaling in MHV-68-infected mice. WT/CD25−/− chimeric mice were infected with MHV-68 i.n., and the percentage of Vβ4+ CD8+ T cells was determined over time for each congenic population. Representative plots from day 36 p.i. (A) or the averages over time (B) are shown. WT and CD25−/− CD8+ T cells from chimeric mice were analyzed for expression of CD62L over time. Representative plots from day 24 p.i. (C) or the averages over time (D) are shown. *, P < 0.05; **, P < 0.01). Error bars represent standard deviations from the means. Four mice were used per group, and data are representative of at least two experiments.During infectious mononucleosis, CD8+ T cells are in a highly activated state and thus express low levels of CD62L (20). Therefore, we analyzed CD8+ T cells from chimeric mice for expression of CD62L. After MHV-68 infection, the majority of WT CD8+ T cells in the peripheral blood were CD62Llow, as previously reported (Fig. 4C and D) (20). Interestingly, CD25−/− CD8+ T cells failed to develop this dominant CD62Llow population, indicating that CD25 is necessary for the activation of the CD8+ T cell compartment in addition to cell expansion during mononucleosis (Fig. 4C and D). When we analyzed the Vβ4+ CD8+ T cell compartment, we observed that WT cells downregulated expression of CD62L. While Vβ4+ cells from the CD25−/− compartment also decreased expression of CD62L, they did so to a lesser extent both as a percentage and on a per-cell basis (see Fig. S2 in the supplemental material).In these studies, we have shown that signaling through CD25 is necessary for the generation of an optimal primary CD8+ T cell response against a gammaherpesvirus, since virus-specific CD8+ T cells were unable to expand as robustly as WT cells and did not fully differentiate into short-lived effector cells. These observations are consistent with previous results from our lab and findings of others using a variety of acute infection models (17, 18). However, not all persistent infections appear to require CD25, since the m45-specific response to murine cytomegalovirus (MCMV) infection occurs normally in the absence of IL-2 signals (1). What allows for some responses to be independent of IL-2 remains unknown. Potential explanations could involve differences in tropism, the route of infection, or the amount of proinflammatory cytokines induced by each infection. Despite the dependence on CD25 for the short-term effector response, the memory CD8+ T cell response remained intact in the absence of IL-2 signaling. In contrast, Vβ4 expansion and mononucleosis never attained normal levels. Unlike the antigen-specific response, which relies upon peptide/MHC interactions for induction, mononucleosis does not rely upon conventional antigen presentation (4). Instead, the M1 protein of MHV-68, expressed during the establishment and expansion of latency in the spleen, appears to drive Vβ4 expansion (7). Interestingly, our evidence shows that both antigen-dependent and -independent CD8+ T cell expansion require CD25. Antigen-specific T cells also undergo an apoptotic contraction phase, followed by a lower frequency of cells surviving as relatively quiescent memory cells. In contrast, during mononucleosis caused by MHV-68, CD8+ T cells remain in an activated state and do not undergo a marked contraction, providing a potential explanation as to why the WT and CD25−/− Vβ4 populations continue to differ in both number and phenotype later in the response.Earlier studies have also identified CD4+ T cells as being critical for the development of MHV-68-induced infectious mononucleosis (11, 20). We have previously shown that CD4+ T cell help was critical for robust expression of CD25 on activated antigen-specific CD8+ T cells. Interestingly, when we measured CD25 expression on Vβ4+ cells from mice lacking CD4+ T cells, we saw a moderate decrease in the level of CD25 expressed (data not shown), indicating one potential reason why CD4-deficient mice do not experience infectious mononucleosis. However, it is likely that other factors involving CD4+ T cells and activation of B cells are also involved (10).In conclusion, the significance of these studies is twofold. First, they shed light on the requirements for MHV-68-induced mononucleosis. Second, our data illustrate that CD25 is required for both antigen-specific and non-antigen-specific activation of CD8+ T cell responses, while being dispensable for memory cell formation. This knowledge may be useful in developing new T cell-based immune therapies to enhance control of persistent gammaherpesvirus infections.   相似文献   

19.

Background

Recently we and others have identified CD8 and CD4 T cell epitopes within the highly expressed M. tuberculosis protein TB10.4. This has enabled, for the first time, a comparative study of the dynamics and function of CD4 and CD8 T cells specific for epitopes within the same protein in various stages of TB infection.

Methods and Findings

We focused on T cells directed to two epitopes in TB10.4; the MHC class I restricted epitope TB10.4 3–11 (CD8/10.4 T cells) and the MHC class II restricted epitope TB10.4 74–88 (CD4/10.4 T cells). CD4/10.4 and CD8/10.4 T cells displayed marked differences in terms of expansion and contraction in a mouse TB model. CD4/10.4 T cells dominated in the early phase of infection whereas CD8/10.4 T cells were expanded after week 16 and reached 5–8 fold higher numbers in the late phase of infection. In the early phase of infection both CD4/10.4 and CD8/10.4 T cells were characterized by 20–25% polyfunctional cells (IL-2+, IFN-γ+, TNF-α+), but whereas the majority of CD4/10.4 T cells were maintained as polyfunctional T cells throughout infection, CD8/10.4 T cells differentiated almost exclusively into effector cells (IFN-γ+, TNF-α+). Both CD4/10.4 and CD8/10.4 T cells exhibited cytotoxicity in vivo in the early phase of infection, but whereas CD4/10.4 cell mediated cytotoxicity waned during the infection, CD8/10.4 T cells exhibited increasing cytotoxic potential throughout the infection.

Conclusions/Significance

Our results show that CD4 and CD8 T cells directed to epitopes in the same antigen differ both in their kinetics and functional characteristics throughout an infection with M. tuberculosis. In addition, the observed strong expansion of CD8 T cells in the late stages of infection could have implications for the development of post exposure vaccines against latent TB.  相似文献   

20.
Conventional methods to induce tolerance in humans have met with limited success. Hence, efforts to redirect tolerogen uptake using reovirus adhesin, protein sigma 1 (pσ1), may circumvent these shortcomings based upon the recent finding that when reovirus pσ1 is engineered to deliver chicken ovalbumin (OVA) mucosally, tolerance is obtained, even with a single dose. To test whether single-dose tolerance can be induced to treat EAE, proteolipid protein (PLP130–151) was genetically fused to OVA to pσ1 (PLP:OVA-pσ1) and shown to significantly ameliorate EAE, suppressing proinflammatory cytokines by IL-10+ forkhead box P3 (FoxP3)+ CD25+CD4+ Treg and IL-4+CD25CD4+ Th2 cells. IL-10R or IL-4 neutralization reversed protection to EAE conferred by PLP:OVA-pσ1, and adoptive transfer of Ag-specific Treg or Th2 cells restored protection against EAE in recipients. Upon assessment of each relative participant, functional inactivation of CD25 impaired PLP:OVA-pσ1''s protective capacity, triggering TGF-β-mediated inflammation; however, concomitant inactivation of TGF-β and CD25 reestablished PLP:OVA-pσ1-mediated protection by IL-28-producing FoxP3+CD25CD4+ T cells. Thus, pσ1-based therapy can resolve EAE independently of or dependently upon CD25 and assigns IL-28 as an alternative therapy for autoimmunity.  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