Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 117(8): 4292-4299, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32034098

ABSTRACT

The migratory patterns of virus-specific CD8 T cells during chronic viral infection are not well understood. To address this issue, we have done parabiosis experiments during chronic lymphocytic choriomeningitis virus (LCMV) infection of mice. We found that despite the high frequency of virus-specific CD8 T cells in both lymphoid and nonlymphoid tissues there was minimal migration of virus-specific CD8 T cells between the chronically infected conjoined parabiont mice. This was in contrast to parabionts between mice that had undergone an acute LCMV infection where virus-specific CD8 T cells established equilibrium demonstrating circulation of memory T cells generated after viral clearance. We have identified a population of PD-1+ TCF1+CXCR5+Tim-3- stemlike virus-specific CD8 T cells that reside in lymphoid tissues and act as resource cells for maintaining the T cell response during chronic infection. These are the cells that proliferate and give rise to the more terminally differentiated PD-1+ CXCR5-Tim-3+ CD8 T cells. Both the stemlike CD8 T cells and their terminally differentiated progeny showed minimal migration during chronic infection and the few LCMV-specific CD8 T cells that were present in circulation were the recently emerging progeny from the stemlike CD8 T cells. The PD-1+ TCF1+CXCR5+ stemlike CD8 T cells were truly resident in lymphoid tissues and did not circulate in the blood. We propose that this residency in specialized niches within lymphoid tissues is a key aspect of their biology and is essential for maintaining their quiescence and stemlike program under conditions of a chronic viral infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Lymphoid Tissue/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Female , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/immunology , Humans , Immunologic Memory , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/genetics , Lymphoid Tissue/virology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology
2.
Immunity ; 51(6): 1043-1058.e4, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31810882

ABSTRACT

T cell dysfunction is a characteristic feature of chronic viral infection and cancer. Recent studies in chronic lymphocytic choriomeningitis virus (LCMV) infection have defined a PD-1+ Tcf-1+ CD8+ T cell subset capable of self-renewal and differentiation into more terminally differentiated cells that downregulate Tcf-1 and express additional inhibitory molecules such as Tim3. Here, we demonstrated that expression of the glycoprotein CD101 divides this terminally differentiated population into two subsets. Stem-like Tcf-1+ CD8+ T cells initially differentiated into a transitory population of CD101-Tim3+ cells that later converted into CD101+ Tim3+ cells. Recently generated CD101-Tim3+ cells proliferated in vivo, contributed to viral control, and were marked by an effector-like transcriptional signature including expression of the chemokine receptor CX3CR1, pro-inflammatory cytokines, and granzyme B. PD-1 pathway blockade increased the numbers of CD101-Tim3+ CD8+ T cells, suggesting that these newly generated transitional cells play a critical role in PD-1-based immunotherapy.


Subject(s)
Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Programmed Cell Death 1 Receptor/metabolism , Animals , Biomarkers/metabolism , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Female , Granzymes/genetics , Granzymes/metabolism , Hepatitis A Virus Cellular Receptor 2/biosynthesis , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Lymphocytic Choriomeningitis/virology , Male , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics
3.
Nature ; 552(7685): 404-409, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29236683

ABSTRACT

Memory CD8 T cells that circulate in the blood and are present in lymphoid organs are an essential component of long-lived T cell immunity. These memory CD8 T cells remain poised to rapidly elaborate effector functions upon re-exposure to pathogens, but also have many properties in common with naive cells, including pluripotency and the ability to migrate to the lymph nodes and spleen. Thus, memory cells embody features of both naive and effector cells, fuelling a long-standing debate centred on whether memory T cells develop from effector cells or directly from naive cells. Here we show that long-lived memory CD8 T cells are derived from a subset of effector T cells through a process of dedifferentiation. To assess the developmental origin of memory CD8 T cells, we investigated changes in DNA methylation programming at naive and effector cell-associated genes in virus-specific CD8 T cells during acute lymphocytic choriomeningitis virus infection in mice. Methylation profiling of terminal effector versus memory-precursor CD8 T cell subsets showed that, rather than retaining a naive epigenetic state, the subset of cells that gives rise to memory cells acquired de novo DNA methylation programs at naive-associated genes and became demethylated at the loci of classically defined effector molecules. Conditional deletion of the de novo methyltransferase Dnmt3a at an early stage of effector differentiation resulted in reduced methylation and faster re-expression of naive-associated genes, thereby accelerating the development of memory cells. Longitudinal phenotypic and epigenetic characterization of the memory-precursor effector subset of virus-specific CD8 T cells transferred into antigen-free mice revealed that differentiation to memory cells was coupled to erasure of de novo methylation programs and re-expression of naive-associated genes. Thus, epigenetic repression of naive-associated genes in effector CD8 T cells can be reversed in cells that develop into long-lived memory CD8 T cells while key effector genes remain demethylated, demonstrating that memory T cells arise from a subset of fate-permissive effector T cells.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Dedifferentiation , Immunologic Memory , Animals , DNA (Cytosine-5-)-Methyltransferases/deficiency , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA Methyltransferase 3A , Epigenesis, Genetic , Female , Immunologic Memory/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL
4.
Nat Immunol ; 18(9): 1046-1057, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28714979

ABSTRACT

Translation is a critical process in protein synthesis, but translational regulation in antigen-specific T cells in vivo has not been well defined. Here we have characterized the translatome of virus-specific CD8+ effector T cells (Teff cells) during acute infection of mice with lymphocytic choriomeningitis virus (LCMV). Antigen-specific T cells exerted dynamic translational control of gene expression that correlated with cell proliferation and stimulation via the T cell antigen receptor (TCR). The translation of mRNAs that encode translation machinery, including ribosomal proteins, was upregulated during the T cell clonal-expansion phase, followed by inhibition of the translation of those transcripts when the CD8+ Teff cells stopped dividing just before the contraction phase. That translational suppression was more pronounced in terminal effector cells than in memory precursor cells and was regulated by antigenic stimulation and signals from the kinase mTOR. Our studies show that translation of transcripts encoding ribosomal proteins is regulated during the differentiation of CD8+ Teff cells and might have a role in fate 'decisions' involved in the formation of memory cells.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Protein Biosynthesis/immunology , Animals , Arenaviridae Infections/genetics , Arenaviridae Infections/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Flow Cytometry , Gene Expression Regulation , Immunologic Memory/immunology , Interferon-gamma/immunology , Lymphocytic choriomeningitis virus , Mice , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/immunology , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , TOR Serine-Threonine Kinases/immunology
5.
Science ; 355(6332): 1423-1427, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28280249

ABSTRACT

Programmed cell death-1 (PD-1)-targeted therapies enhance T cell responses and show efficacy in multiple cancers, but the role of costimulatory molecules in this T cell rescue remains elusive. Here, we demonstrate that the CD28/B7 costimulatory pathway is essential for effective PD-1 therapy during chronic viral infection. Conditional gene deletion showed a cell-intrinsic requirement of CD28 for CD8 T cell proliferation after PD-1 blockade. B7-costimulation was also necessary for effective PD-1 therapy in tumor-bearing mice. In addition, we found that CD8 T cells proliferating in blood after PD-1 therapy of lung cancer patients were predominantly CD28-positive. Taken together, these data demonstrate CD28-costimulation requirement for CD8 T cell rescue and suggest an important role for the CD28/B7 pathway in PD-1 therapy of cancer patients.


Subject(s)
B7-1 Antigen/metabolism , CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Lymphocytic Choriomeningitis/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , B7-1 Antigen/genetics , CD28 Antigens/genetics , Gene Deletion , Humans , Immunotherapy , Metabolic Networks and Pathways , Mice
6.
J Virol ; 91(5)2017 03 01.
Article in English | MEDLINE | ID: mdl-28003483

ABSTRACT

Adenovirus serotype 5 (Ad5) is one of the most widely used viral vectors and is known to generate potent T cell responses. While many previous studies have characterized Ad5-induced CD8 T cell responses, there is a relative lack of detailed studies that have analyzed CD4 T cells elicited by Ad5 vaccination. Here, we immunized mice with Ad5 vectors encoding lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP) and examined GP-specific CD4 T cell responses elicited by Ad5 vectors and compared them to those induced by an acute LCMV infection. In contrast to LCMV infection, where balanced CD4 T helper 1 (Th1) and T follicular helper (Tfh) responses were induced, Ad5 immunization resulted in a significantly reduced frequency of Th1 cells. CD4 T cells elicited by Ad5 vectors expressed decreased levels of Th1 markers, such as Tim3, SLAM, T-bet, and Ly6C, had smaller amounts of cytotoxic molecules like granzyme B, and produced less interferon gamma than CD4 T cells induced by LCMV infection. This defective CD4 Th1 response appeared to be intrinsic for Ad5 vectors and not a reflection of comparing a nonreplicating vector to a live viral infection, since immunization with a DNA vector expressing LCMV-GP generated efficient CD4 Th1 responses. Analysis at early time points (day 3 or 4) after immunization with Ad5 vectors revealed a defect in the expression of CD25 (interleukin-2 [IL-2] receptor alpha chain) on Ad5-elicited CD4 T cells, and administration of exogenous IL-2 following Ad5 immunization partially restored CD4 Th1 responses. These results suggest that impairment of Th1 commitment after Ad5 immunization could be due to reduced IL-2-mediated signaling.IMPORTANCE During viral infection, generating balanced responses of Th1 and Tfh cells is important to induce effective cell-mediated responses and provide optimal help for antibody responses. In this study, to investigate vaccine-induced CD4 T cell responses, we characterized CD4 T cells after immunization with Ad5 vectors expressing LCMV-GP in mice. Ad5 vectors led to altered effector differentiation of LCMV GP-specific CD4 T cells compared to that during LCMV infection. CD4 T cells following Ad5 immunization exhibited impaired Th1 lineage commitment, generating significantly decreased Th1 responses than those induced by LCMV infection. Our results suggest that suboptimal IL-2 signaling possibly plays a role in reduced Th1 development following Ad5 immunization.


Subject(s)
Adenoviridae/immunology , Lymphocytic Choriomeningitis/prevention & control , Lymphocytic choriomeningitis virus/immunology , Th1 Cells/immunology , Vaccination , Viral Vaccines/administration & dosage , Administration, Intravenous , Animals , Antibodies, Viral/blood , Cell Differentiation/immunology , Female , Glycoproteins/immunology , Injections, Intramuscular , Lymphocytic Choriomeningitis/blood , Lymphocytic Choriomeningitis/immunology , Mice, Inbred C57BL , Viral Proteins/immunology , Viral Vaccines/immunology
7.
Nat Immunol ; 15(12): 1152-61, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25362489

ABSTRACT

The importance of autophagy in the generation of memory CD8(+) T cells in vivo is not well defined. We report here that autophagy was dynamically regulated in virus-specific CD8(+) T cells during acute infection of mice with lymphocytic choriomeningitis virus. In contrast to the current paradigm, autophagy decreased in activated proliferating effector CD8(+) T cells and was then upregulated when the cells stopped dividing just before the contraction phase. Consistent with those findings, deletion of the gene encoding either of the autophagy-related molecules Atg5 or Atg7 had little to no effect on the proliferation and function of effector cells, but these autophagy-deficient effector cells had survival defects that resulted in compromised formation of memory T cells. Our studies define when autophagy is needed during effector and memory differentiation and warrant reexamination of the relationship between T cell activation and autophagy.


Subject(s)
Autophagy/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Immunologic Memory/immunology , Animals , Cell Separation , Cell Survival/immunology , Chromatography, Liquid , Flow Cytometry , Immunoblotting , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/immunology , Mass Spectrometry , Mice , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Transduction, Genetic
8.
J Exp Med ; 211(9): 1905-18, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25113973

ABSTRACT

Regulatory T (T reg) cells are critical for preventing autoimmunity mediated by self-reactive T cells, but their role in modulating immune responses during chronic viral infection is not well defined. To address this question and to investigate a role for T reg cells in exhaustion of virus-specific CD8 T cells, we depleted T reg cells in mice chronically infected with lymphocytic choriomeningitis virus (LCMV). T reg cell ablation resulted in 10-100-fold expansion of functional LCMV-specific CD8 T cells. Rescue of exhausted CD8 T cells was dependent on cognate antigen, B7 costimulation, and conventional CD4 T cells. Despite the striking recovery of LCMV-specific CD8 T cell responses, T reg cell depletion failed to diminish viral load. Interestingly, T reg cell ablation triggered up-regulation of the molecule programmed cell death ligand-1 (PD-L1), which upon binding PD-1 on T cells delivers inhibitory signals. Increased PD-L1 expression was observed especially on LCMV-infected cells, and combining T reg cell depletion with PD-L1 blockade resulted in a significant reduction in viral titers, which was more pronounced than that upon PD-L1 blockade alone. These results suggest that T reg cells effectively maintain CD8 T cell exhaustion, but blockade of the PD-1 inhibitory pathway is critical for elimination of infected cells.


Subject(s)
Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chronic Disease , Dendritic Cells/immunology , Lymphocyte Activation , Lymphocyte Depletion , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Up-Regulation
9.
J Virol ; 87(13): 7737-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23637417

ABSTRACT

Long-lived plasma cells that reside in the bone marrow constitutively produce antibody in the absence of antigen and are the cellular basis of durable humoral immunity. The generation of these long-lived plasma cells depends upon a series of highly orchestrated interactions between antigen-specific CD4 T cells and B cells and the formation of germinal centers (GCs). In this study, we have examined the role of the cytokine interleukin-21 (IL-21) in regulating humoral immunity during acute viral infections. Using IL-21 receptor-deficient (IL-21R(-/-)) mice, we found that virus-specific CD4 T cells were generated after infection with lymphocytic choriomeningitis virus (LCMV) and that these CD4 T cells differentiated into T follicular helper (TFH)-like cells in the absence of IL-21 signaling. There was also no defect in the formation of GCs, although after day 15 these GCs disappeared faster in IL-21R(-/-) mice than in wild-type mice. Isotype switching and the initial LCMV-specific IgG response were normal in IL-21R(-/-) mice. However, these mice exhibited a profound defect in generating long-lived plasma cells and in sustaining antibody levels over time. Similar results were seen after infection of IL-21R(-/-) mice with vesicular stomatitis virus and influenza virus. Using chimeric mice containing wild-type or IL-21R(-/-) CD4 T cells and B cells, we showed that both B and CD4 T cells need IL-21 signaling for generating long-term humoral immunity. Taken together, our results highlight the importance of IL-21 in humoral immunity to viruses.


Subject(s)
Cell Differentiation/immunology , Immunity, Humoral/immunology , Interleukins/immunology , Plasma Cells/immunology , Virus Diseases/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Germinal Center/immunology , Hemagglutination Tests , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutralization Tests , Plasma Cells/virology , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-21/genetics
10.
Methods Mol Biol ; 821: 305-16, 2012.
Article in English | MEDLINE | ID: mdl-22125074

ABSTRACT

RNA interference (RNAi) is an intracellular mechanism for silencing gene expression utilizing short fragments of double-strand RNA that are complementary to the target messenger RNA. This gene silencing technique has now become an invaluable research tool due to its specific and strong repressive effect on a target transcript. We have recently applied a retrovirus-based RNAi system to investigate the in vivo role of the mammalian target of rapamycin (mTOR) in antigen-specific CD8 T cells, and have found that mTOR regulates memory CD8 T-cell differentiation. Here, we provide a detailed protocol for knocking down mTOR and its related molecules (raptor and FKBP12) in antigen-specific CD8 T cells. In our protocol, a mouse model of lymphocytic choriomeningitis virus infection is used, but the methods can be extended to other viral and bacterial infections as well as vaccinations. Also, the similar approach can be applied to analysis of CD4 T-cell responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Knockdown Techniques , Immunity, Innate/genetics , RNA Interference , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , Adaptor Proteins, Signal Transducing , Animals , CD8-Positive T-Lymphocytes/cytology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation , Cell Line , Disease Models, Animal , Flow Cytometry , Gene Expression Regulation , Gene Silencing/immunology , Genetic Vectors , Humans , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Regulatory-Associated Protein of mTOR , Retroviridae , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Protein 1A/metabolism , Transduction, Genetic
11.
Immunity ; 35(2): 285-98, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21856186

ABSTRACT

To design successful vaccines for chronic diseases, an understanding of memory CD8(+) T cell responses to persistent antigen restimulation is critical. However, most studies comparing memory and naive cell responses have been performed only in rapidly cleared acute infections. Herein, by comparing the responses of memory and naive CD8(+) T cells to acute and chronic lymphocytic choriomeningitis virus infection, we show that memory cells dominated over naive cells and were protective when present in sufficient numbers to quickly reduce infection. In contrast, when infection was not rapidly reduced, because of high antigen load or persistence, memory cells were quickly lost, unlike naive cells. This loss of memory cells was due to a block in sustaining cell proliferation, selective regulation by the inhibitory receptor 2B4, and increased reliance on CD4(+) T cell help. Thus, emphasizing the importance of designing vaccines that elicit effective CD4(+) T cell help and rapidly control infection.


Subject(s)
Antigens, CD/metabolism , Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/metabolism , Lymphocytic choriomeningitis virus/physiology , Receptors, Immunologic/metabolism , T-Lymphocyte Subsets/metabolism , Acute Disease , Adoptive Transfer , Animals , Antigens, CD/immunology , Arenaviridae Infections/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cell Proliferation , Cells, Cultured , Chronic Disease , Cytokines/immunology , Cytokines/metabolism , Immunologic Memory , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Paracrine Communication , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Immunologic/immunology , Signaling Lymphocytic Activation Molecule Family , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/virology , Viral Load , Viral Vaccines
12.
J Exp Med ; 205(3): 625-40, 2008 Mar 17.
Article in English | MEDLINE | ID: mdl-18316415

ABSTRACT

An important question in memory development is understanding the differences between effector CD8 T cells that die versus effector cells that survive and give rise to memory cells. In this study, we provide a comprehensive phenotypic, functional, and genomic profiling of terminal effectors and memory precursors. Using killer cell lectin-like receptor G1 as a marker to distinguish these effector subsets, we found that despite their diverse cell fates, both subsets possessed remarkably similar gene expression profiles and functioned as equally potent killer cells. However, only the memory precursors were capable of making interleukin (IL) 2, thus defining a novel effector cell that was cytotoxic, expressed granzyme B, and produced inflammatory cytokines in addition to IL-2. This effector population then differentiated into long-lived protective memory T cells capable of self-renewal and rapid recall responses. Experiments to understand the signals that regulate the generation of terminal effectors versus memory precursors showed that cells that continued to receive antigenic stimulation during the later stages of infection were more likely to become terminal effectors. Importantly, curtailing antigenic stimulation toward the tail end of the acute infection enhanced the generation of memory cells. These studies support the decreasing potential model of memory differentiation and show that the duration of antigenic stimulation is a critical regulator of memory formation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory , Animals , CD8-Positive T-Lymphocytes/classification , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Proliferation , Down-Regulation , Gene Expression Profiling , Granzymes/biosynthesis , Interleukin-2/biosynthesis , Interleukin-7 Receptor alpha Subunit/genetics , Interleukin-7 Receptor alpha Subunit/metabolism , Lectins, C-Type , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism
13.
Science ; 317(5838): 670-4, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17673664

ABSTRACT

The chemokines CCL21 and CXCL13 are immune factors that dictate homing and motility of lymphocytes and dendritic cells in lymphoid tissues. However, the means by which these chemokines are regulated and how they influence cell trafficking during immune responses remain unclear. We show that CCL21 and CXCL13 are transiently down-regulated within lymphoid tissues during immune responses by a mechanism controlled by the cytokine interferon-gamma. This modulation was found to alter the localization of lymphocytes and dendritic cells within responding lymphoid tissues. As a consequence, priming of T cell responses to a second distinct pathogen after chemokine modulation became impaired. We propose that this transient chemokine modulation may help orchestrate local cellularity, thus minimizing competition for space and resources in activated lymphoid tissues.


Subject(s)
Chemokines, CC/metabolism , Chemokines, CXC/metabolism , Listeriosis/immunology , Lymph Nodes/immunology , Spleen/immunology , Virus Diseases/immunology , Adoptive Transfer , Animals , Arenaviridae Infections/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Chemokine CCL19 , Chemokine CCL21 , Chemokine CXCL13 , Chemokines, CC/genetics , Chemokines, CXC/genetics , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Down-Regulation , Female , Homeostasis , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocytic choriomeningitis virus , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
J Immunol ; 172(12): 7813-20, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15187165

ABSTRACT

Transplanted organs fail due to either acute or chronic rejection. The prevailing view is that the nature or magnitude of the recipient's immune response to donor Ags determines the type of rejection. In variance with this view, we show in this study that the status of the graft itself plays a dominant role in defining the type of rejection even in the face of an established alloimmune response. Using adoptive transfer mouse models in which the graft is exposed to a constant number of effector lymphocytes, we found that newly transplanted heart allografts are rejected acutely, while healed-in allografts undergo chronic rejection. Acute rejection of healed-in allografts was largely recapitulated by subjecting the grafts to ischemia-reperfusion injury similar to that present in newly transplanted organs. Ischemia-Reperfusion injury altered the outcome of rejection by enhancing the accumulation of effector T cells within the graft. The accumulation of effector T cells in the graft was dependent on the presence of both ischemia-reperfusion injury (inflammation) and alloantigens. These findings demonstrate that the graft plays a dominant role in shaping the outcome of rejection by controlling the trafficking of effector T cells.


Subject(s)
Graft Rejection/immunology , Heart Transplantation/immunology , Immunity, Cellular , T-Lymphocytes , Acute Disease , Adoptive Transfer , Animals , Cell Adhesion Molecules/biosynthesis , Cell Movement , Chemokines/biosynthesis , Chronic Disease , Graft Rejection/diagnosis , Mice , Mice, Inbred Strains , Reperfusion Injury , Time Factors , Transplantation, Homologous
15.
Nat Immunol ; 4(12): 1191-8, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14625547

ABSTRACT

A major unanswered question is what distinguishes the majority of activated CD8 T cells that die after an acute viral infection from the small fraction (5-10%) that survive to become long-lived memory cells. In this study we show that increased expression of the interleukin 7 receptor alpha-chain (IL-7Ralpha) identifies the effector CD8 T cells that will differentiate into memory cells. IL-7R(hi) effector cells contained increased amounts of antiapoptotic molecules, and adoptive transfer of IL-7R(hi) and IL-7R(lo) effector cells showed that IL-7R(hi) cells preferentially gave rise to memory cells that could persist and confer protective immunity. Thus, selective expression of IL-7R identifies memory cell precursors, and this marker may be useful in predicting the number of memory T cells generated after infection or immunization.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Immunologic Memory , Receptors, Interleukin-7/biosynthesis , T-Lymphocyte Subsets/cytology , Adoptive Transfer , Animals , Antigens, Surface , CD8-Positive T-Lymphocytes/physiology , Cell Lineage/immunology , Gene Expression/immunology , Lymphocyte Activation , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Signal Transduction/immunology , T-Lymphocyte Subsets/physiology
16.
Proc Natl Acad Sci U S A ; 99(9): 6175-80, 2002 Apr 30.
Article in English | MEDLINE | ID: mdl-11983909

ABSTRACT

The allospecifc T cell population responding to a transplanted organ consists of both naive and memory lymphocytes. Although it is established that naive T cells are activated by antigen within the organized structures of secondary lymphoid organs (the spleen, lymph nodes, and mucosal lymphoid tissues), it is not clear whether memory T cell activation and propagation depend on homing to these organs. To answer this question, we investigated whether allospecific naive or memory T cells can mediate acute cardiac allograft rejection in mutant mice that lack all of their secondary lymphoid tissues. The results of our experiments demonstrated that antigen-experienced memory T cells have two advantages over naive T cells: (i) memory T cells mount a vigorous immune response that leads to allograft rejection independent of secondary lymphoid organs; and (ii) memory T cells generate more memory T cells without homing to secondary lymphoid organs. These unique properties of memory T cells were further confirmed by showing that memory-like T cells that arise from the homeostatic proliferation of naive T cells in the absence of antigenic stimulation are suboptimal at rejecting allografts and do not generate memory T cells in mice devoid of secondary lymphoid tissues.


Subject(s)
Immunologic Memory , Lymph Nodes/immunology , Mucous Membrane/immunology , Spleen/immunology , T-Lymphocyte Subsets/immunology , Animals , Cell Transplantation , Flow Cytometry , Immunophenotyping , Mice , Mice, Inbred BALB C , Mutation , Recombination, Genetic , T-Lymphocytes, Cytotoxic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL