Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 75(6): 1229-1242.e5, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31377117

ABSTRACT

Interferon gamma (IFN-γ), critical for host defense and tumor surveillance, requires tight control of its expression. Multiple cis-regulatory elements exist around Ifng along with a non-coding transcript, Ifng-as1 (also termed NeST). Here, we describe two genetic models generated to dissect the molecular functions of this locus and its RNA product. DNA deletion within the Ifng-as1 locus disrupted chromatin organization of the extended Ifng locus, impaired Ifng response, and compromised host defense. Insertion of a polyA signal ablated the Ifng-as1 full-length transcript and impaired host defense, while allowing proper chromatin structure. Transient knockdown of Ifng-as1 also reduced IFN-γ production. In humans, discordant expression of IFNG and IFNG-AS1 was evident in memory T cells, with high expression of this long non-coding RNA (lncRNA) and low expression of the cytokine. These results establish Ifng-as1 as an important regulator of Ifng expression, as a DNA element and transcribed RNA, involved in dynamic and cell state-specific responses to infection.


Subject(s)
Gene Expression Regulation/immunology , Immunologic Memory , Infections/immunology , Interferon-gamma/immunology , RNA, Untranslated/immunology , T-Lymphocytes/immunology , Animals , Chromatin/genetics , Chromatin/immunology , Female , Gene Knockdown Techniques , Infections/genetics , Infections/pathology , Interferon-gamma/genetics , Mice , RNA, Untranslated/genetics , T-Lymphocytes/pathology
2.
J Virol ; 97(1): e0155622, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36541799

ABSTRACT

Increased demand for novel, highly effective vaccination strategies necessitates a better understanding of long-lived memory CD8 T cell differentiation. To achieve this understanding, we used the mouse model of acute lymphocytic choriomeningitis virus (LCMV) infection. We reexamined classical memory CD8 T cell subsets and performed in-depth, longitudinal analysis of their phenotype, transcriptional programming, and anatomic location within the spleen. All analyses were performed at multiple time points from 8 days to 1 year postinfection. Memory subsets are conventionally defined by their expression of KLRG1 and IL-7Rα, as follows: KLRG1+IL-7Rα- terminal effectors (TEs) and KLRG1-IL-7Rα+ memory precursors (MPs). But we also characterized a third KLRG1+IL-7Rα+ subset which we refer to as KLRG1+ MPs. In these analyses, we defined a comprehensive memory phenotype that is associated with higher levels of CD28 expression. We also demonstrated that MPs, KLRG1+ MPs, and TEs have distinct localization programs within the spleen. We found that MPs became preferentially enriched in the white pulp as early as 1 to 2 weeks postinfection, and their predominance in the white pulp was maintained throughout the course of a year. On the other hand, KLRG1+ MPs and TEs localized to the red pulp just as early, and they consistently localized to the red pulp thereafter. These findings indicate that location may be crucial for memory formation and that white pulp-derived signals may contribute to long-term memory survival. Achieving robust memory responses following vaccination may require more deliberate consideration of which memory phenotypes are induced, as well as where they traffic, as these factors could impact their longevity. IMPORTANCE CD8 T cells play a critical role in viral immunity and it is important to understand how memory cells are formed and what processes lead to their long-term maintenance. Here, we use a mouse model of acute infection to perform an in-depth, longitudinal analysis of memory CD8 T cell differentiation, examining the phenotype and location of memory cells out to 1 year postinfection.


Subject(s)
Lymphocytic Choriomeningitis , T-Lymphocyte Subsets , Animals , Mice , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus , Mice, Inbred C57BL , Phenotype , Vaccination , CD28 Antigens/genetics , Transcriptome , Antigens, Surface/genetics , Viral Vaccines/immunology
3.
Nature ; 552(7685): 362-367, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29236685

ABSTRACT

The differentiation of human memory CD8 T cells is not well understood. Here we address this issue using the live yellow fever virus (YFV) vaccine, which induces long-term immunity in humans. We used in vivo deuterium labelling to mark CD8 T cells that proliferated in response to the virus and then assessed cellular turnover and longevity by quantifying deuterium dilution kinetics in YFV-specific CD8 T cells using mass spectrometry. This longitudinal analysis showed that the memory pool originates from CD8 T cells that divided extensively during the first two weeks after infection and is maintained by quiescent cells that divide less than once every year (doubling time of over 450 days). Although these long-lived YFV-specific memory CD8 T cells did not express effector molecules, their epigenetic landscape resembled that of effector CD8 T cells. This open chromatin profile at effector genes was maintained in memory CD8 T cells isolated even a decade after vaccination, indicating that these cells retain an epigenetic fingerprint of their effector history and remain poised to respond rapidly upon re-exposure to the pathogen.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Epigenesis, Genetic , Immunologic Memory/immunology , Yellow Fever Vaccine/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Proliferation , Chromatin/genetics , Chromatin/metabolism , DNA Methylation , Deuterium , Gene Expression Profiling , Half-Life , Humans , Immunologic Memory/genetics , Lymphocyte Count , Mice , Radioisotope Dilution Technique , Transcription, Genetic , Yellow Fever/immunology , Yellow Fever/virology , Yellow fever virus/immunology
4.
Immunity ; 38(4): 805-17, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23583644

ABSTRACT

CD4(+) T follicular helper (Tfh) cells provide the required signals to B cells for germinal center reactions that are necessary for long-lived antibody responses. However, it remains unclear whether there are CD4(+) memory T cells committed to the Tfh cell lineage after antigen clearance. By using adoptive transfer of antigen-specific memory CD4(+) T cell subpopulations in the lymphocytic choriomeningitis virus infection model, we found that there are distinct memory CD4(+) T cell populations with commitment to either Tfh- or Th1-cell lineages. Our conclusions are based on gene expression profiles, epigenetic studies, and phenotypic and functional analyses. Our findings indicate that CD4(+) memory T cells "remember" their previous effector lineage after antigen clearance, being poised to reacquire their lineage-specific effector functions upon antigen reencounter. These findings have important implications for rational vaccine design, where improving the generation and engagement of memory Tfh cells could be used to enhance vaccine-induced protective immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , Adoptive Transfer , Animals , Antigens, Viral/immunology , Cell Differentiation , Cell Lineage , Cells, Cultured , DNA Methylation/immunology , Epigenesis, Genetic/immunology , Granzymes/genetics , Immunologic Memory , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CXCR5/metabolism , Transcriptome
5.
PLoS Comput Biol ; 17(10): e1009468, 2021 10.
Article in English | MEDLINE | ID: mdl-34648489

ABSTRACT

Understanding how immunological memory lasts a lifetime requires quantifying changes in the number of memory cells as well as how their division and death rates change over time. We address these questions by using a statistically powerful mixed-effects differential equations framework to analyze data from two human studies that follow CD8 T cell responses to the yellow fever vaccine (YFV-17D). Models were first fit to the frequency of YFV-specific memory CD8 T cells and deuterium enrichment in those cells 42 days to 1 year post-vaccination. A different dataset, on the loss of YFV-specific CD8 T cells over three decades, was used to assess out of sample predictions of our models. The commonly used exponential and bi-exponential decline models performed relatively poorly. Models with the cell loss following a power law (exactly or approximately) were most predictive. Notably, using only the first year of data, these models accurately predicted T cell frequencies up to 30 years post-vaccination. Our analyses suggest that division rates of these cells drop and plateau at a low level (0.1% per day, ∼ double the estimated values for naive T cells) within one year following vaccination, whereas death rates continue to decline for much longer. Our results show that power laws can be predictive for T cell memory, a finding that may be useful for vaccine evaluation and epidemiological modeling. Moreover, since power laws asymptotically decline more slowly than any exponential decline, our results help explain the longevity of immune memory phenomenologically.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Yellow Fever Vaccine/immunology , Yellow fever virus/immunology , Computational Biology , Humans , Models, Immunological
6.
Nat Immunol ; 10(1): 116-125, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19029902

ABSTRACT

A major challenge in vaccinology is to prospectively determine vaccine efficacy. Here we have used a systems biology approach to identify early gene 'signatures' that predicted immune responses in humans vaccinated with yellow fever vaccine YF-17D. Vaccination induced genes that regulate virus innate sensing and type I interferon production. Computational analyses identified a gene signature, including complement protein C1qB and eukaryotic translation initiation factor 2 alpha kinase 4-an orchestrator of the integrated stress response-that correlated with and predicted YF-17D CD8(+) T cell responses with up to 90% accuracy in an independent, blinded trial. A distinct signature, including B cell growth factor TNFRS17, predicted the neutralizing antibody response with up to 100% accuracy. These data highlight the utility of systems biology approaches in predicting vaccine efficacy.


Subject(s)
Gene Expression Profiling/methods , Immunity, Innate/genetics , Systems Biology/methods , Yellow Fever Vaccine/immunology , Yellow Fever/prevention & control , Yellow fever virus/immunology , Adolescent , Adult , Antibodies, Viral/blood , CD8-Positive T-Lymphocytes/immunology , Carrier Proteins/genetics , Cells, Cultured , Controlled Clinical Trials as Topic , Humans , Immunity, Active/genetics , Middle Aged , Mitochondrial Proteins/genetics , Multivariate Analysis , Neutralization Tests , Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/genetics , Vaccination , Yellow Fever Vaccine/therapeutic use , Young Adult
7.
Immunity ; 35(3): 400-12, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21943489

ABSTRACT

Functionally exhausted T cells have high expression of the PD-1 inhibitory receptor, and therapies that block PD-1 signaling show promise for resolving chronic viral infections and cancer. By using human and murine systems of acute and chronic viral infections, we analyzed epigenetic regulation of PD-1 expression during CD8(+) T cell differentiation. During acute infection, naive to effector CD8(+) T cell differentiation was accompanied by a transient loss of DNA methylation of the Pdcd1 locus that was directly coupled to the duration and strength of T cell receptor signaling. Further differentiation into functional memory cells coincided with Pdcd1 remethylation, providing an adapted program for regulation of PD-1 expression. In contrast, the Pdcd1 regulatory region was completely demethylated in exhausted CD8(+) T cells and remained unmethylated even when virus titers decreased. This lack of DNA remethylation leaves the Pdcd1 locus poised for rapid expression, potentially providing a signal for premature termination of antiviral functions.


Subject(s)
Antigens, CD/metabolism , Apoptosis Regulatory Proteins/metabolism , CD8-Positive T-Lymphocytes/immunology , DNA Methylation , Promoter Regions, Genetic , Virus Diseases/pathology , Animals , Antigens, CD/genetics , Antigens, Surface/genetics , Antigens, Surface/metabolism , Apoptosis Regulatory Proteins/genetics , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation , Cells, Cultured , Chronic Disease , Epigenomics , Humans , Immunologic Memory , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor , Signal Transduction
8.
Proc Natl Acad Sci U S A ; 114(19): 4993-4998, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28446615

ABSTRACT

Exhausted T cells in chronic infections and cancer have sustained expression of the inhibitory receptor programmed cell death 1 (PD-1). Therapies that block the PD-1 pathway have shown promising clinical results in a significant number of advanced-stage cancer patients. Nonetheless, a better understanding of the immunological responses induced by PD-1 blockade in cancer patients is lacking. Identification of predictive biomarkers is a priority in the field, but whether peripheral blood analysis can provide biomarkers to monitor or predict patients' responses to treatment remains to be resolved. In this study, we analyzed longitudinal blood samples from advanced stage non-small cell lung cancer (NSCLC) patients (n = 29) receiving PD-1-targeted therapies. We detected an increase in Ki-67+ PD-1+ CD8 T cells following therapy in ∼70% of patients, and most responses were induced after the first or second treatment cycle. This T-cell activation was not indiscriminate because we observed only minimal effects on EBV-specific CD8 T cells, suggesting that responding cells may be tumor specific. These proliferating CD8 T cells had an effector-like phenotype (HLA-DR+, CD38+, Bcl-2lo), expressed costimulatory molecules (CD28, CD27, ICOS), and had high levels of PD-1 and coexpression of CTLA-4. We found that 70% of patients with disease progression had either a delayed or absent PD-1+ CD8 T-cell response, whereas 80% of patients with clinical benefit exhibited PD-1+ CD8 T-cell responses within 4 wk of treatment initiation. Our results suggest that peripheral blood analysis may provide valuable insights into NSCLC patients' responses to PD-1-targeted therapies.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung , Cell Proliferation/drug effects , Lung Neoplasms , Lymphocyte Activation/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Male , Nivolumab
9.
Clin Infect Dis ; 69(2): 348-351, 2019 07 02.
Article in English | MEDLINE | ID: mdl-30668661

ABSTRACT

An immunocompetent adult received corticosteroids for chest pain, which later was clinically found to be herpes zoster (HZ). She developed severe disease and rapid viral dissemination that elicited an exceptionally strong varicella zoster virus-specific B-cell and CD8 T-cell response. Clinicians should consider atypical HZ presentation prior to corticosteroid administration.


Subject(s)
Adaptive Immunity , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Encephalitis, Varicella Zoster/immunology , Herpesvirus 3, Human/immunology , Female , Humans , Young Adult
10.
Proc Natl Acad Sci U S A ; 113(10): 2702-7, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26908875

ABSTRACT

Significantly higher levels of plasma CXCL13 [chemokine (C-X-C motif) ligand 13] were associated with the generation of broadly neutralizing antibodies (bnAbs) against HIV in a large longitudinal cohort of HIV-infected individuals. Germinal centers (GCs) perform the remarkable task of optimizing B-cell Ab responses. GCs are required for almost all B-cell receptor affinity maturation and will be a critical parameter to monitor if HIV bnAbs are to be induced by vaccination. However, lymphoid tissue is rarely available from immunized humans, making the monitoring of GC activity by direct assessment of GC B cells and germinal center CD4(+) T follicular helper (GC Tfh) cells problematic. The CXCL13-CXCR5 [chemokine (C-X-C motif) receptor 5] chemokine axis plays a central role in organizing both B-cell follicles and GCs. Because GC Tfh cells can produce CXCL13, we explored the potential use of CXCL13 as a blood biomarker to indicate GC activity. In a series of studies, we found that plasma CXCL13 levels correlated with GC activity in draining lymph nodes of immunized mice, immunized macaques, and HIV-infected humans. Furthermore, plasma CXCL13 levels in immunized humans correlated with the magnitude of Ab responses and the frequency of ICOS(+) (inducible T-cell costimulator) Tfh-like cells in blood. Together, these findings support the potential use of CXCL13 as a plasma biomarker of GC activity in human vaccine trials and other clinical settings.


Subject(s)
Biomarkers/blood , Chemokine CXCL13/blood , Chemokine CXCL13/immunology , Germinal Center/immunology , Animals , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , HIV Infections/blood , HIV Infections/immunology , Humans , Lymph Nodes/immunology , Macaca , Mice, Inbred C57BL , Vaccination
11.
Proc Natl Acad Sci U S A ; 112(10): 3050-5, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25713354

ABSTRACT

CD8 T cells are a potent tool for eliminating intracellular pathogens and tumor cells. Thus, eliciting robust CD8 T-cell immunity is the basis for many vaccines under development. However, the relationship between antigen load and the magnitude of the CD8 T-cell response is not well-described in a human immune response. Here we address this issue by quantifying viral load and the CD8 T-cell response in a cohort of 80 individuals immunized with the live attenuated yellow fever vaccine (YFV-17D) by sampling peripheral blood at days 0, 1, 2, 3, 5, 7, 9, 11, 14, 30, and 90. When the virus load was below a threshold (peak virus load < 225 genomes per mL, or integrated virus load < 400 genome days per mL), the magnitude of the CD8 T-cell response correlated strongly with the virus load (R(2) ∼ 0.63). As the virus load increased above this threshold, the magnitude of the CD8 T-cell responses saturated. Recent advances in CD8 T-cell-based vaccines have focused on replication-incompetent or single-cycle vectors. However, these approaches deliver relatively limited amounts of antigen after immunization. Our results highlight the requirement that T-cell-based vaccines should deliver sufficient antigen during the initial period of the immune response to elicit a large number of CD8 T cells that may be needed for protection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Viral Load , Yellow Fever Vaccine/immunology , Cohort Studies , Gene Expression Profiling , Humans , Yellow fever virus/genetics , Yellow fever virus/immunology , Yellow fever virus/isolation & purification
12.
Proc Natl Acad Sci U S A ; 112(15): 4719-24, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25775592

ABSTRACT

Four Ebola patients received care at Emory University Hospital, presenting a unique opportunity to examine the cellular immune responses during acute Ebola virus infection. We found striking activation of both B and T cells in all four patients. Plasmablast frequencies were 10-50% of B cells, compared with less than 1% in healthy individuals. Many of these proliferating plasmablasts were IgG-positive, and this finding coincided with the presence of Ebola virus-specific IgG in the serum. Activated CD4 T cells ranged from 5 to 30%, compared with 1-2% in healthy controls. The most pronounced responses were seen in CD8 T cells, with over 50% of the CD8 T cells expressing markers of activation and proliferation. Taken together, these results suggest that all four patients developed robust immune responses during the acute phase of Ebola virus infection, a finding that would not have been predicted based on our current assumptions about the highly immunosuppressive nature of Ebola virus. Also, quite surprisingly, we found sustained immune activation after the virus was cleared from the plasma, observed most strikingly in the persistence of activated CD8 T cells, even 1 mo after the patients' discharge from the hospital. These results suggest continued antigen stimulation after resolution of the disease. From these convalescent time points, we identified CD4 and CD8 T-cell responses to several Ebola virus proteins, most notably the viral nucleoprotein. Knowledge of the viral proteins targeted by T cells during natural infection should be useful in designing vaccines against Ebola virus.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Immunity, Cellular/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Ebolavirus/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Hemorrhagic Fever, Ebola/blood , Hemorrhagic Fever, Ebola/virology , Humans , Immunity, Humoral/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunophenotyping , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Viral Proteins/immunology , Viral Proteins/metabolism
13.
J Infect Dis ; 215(12): 1862-1872, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28863472

ABSTRACT

A nurse who acquired Lassa virus infection in Togo in the spring of 2016 was repatriated to the United States for care at Emory University Hospital. Serial sampling from this patient permitted the characterization of several aspects of the innate and cellular immune responses to Lassa virus. Although most of the immune responses correlated with the kinetics of viremia resolution, the CD8 T-cell response was of surprisingly high magnitude and prolonged duration, implying prolonged presentation of viral antigens. Indeed, long after viremia resolution, there was persistent viral RNA detected in the semen of the patient, accompanied by epididymitis, suggesting the male reproductive tract as 1 site of antigen persistence. Consistent with the magnitude of acute T-cell responses, the patient ultimately developed long-term, polyfunctional memory T-cell responses to Lassa virus.


Subject(s)
Antibodies, Viral/blood , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Lassa Fever/immunology , Lassa virus/immunology , Lassa virus/isolation & purification , Adult , Amides/therapeutic use , Antigens, Viral/immunology , Antiviral Agents/therapeutic use , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Class Switching/genetics , Lassa Fever/blood , Lymphocyte Activation , Male , Pyrazines/therapeutic use , Ribavirin/therapeutic use , Viremia/blood
14.
J Virol ; 90(24): 11259-11278, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27707928

ABSTRACT

Epidemiological studies suggest that India has the largest number of dengue virus infection cases worldwide. However, there is minimal information about the immunological responses in these patients. CD8 T cells are important in dengue, because they have been implicated in both protection and immunopathology. Here, we provide a detailed analysis of HLA-DR+ CD38+ and HLA-DR- CD38+ effector CD8 T cell subsets in dengue patients from India and Thailand. Both CD8 T cell subsets expanded and expressed markers indicative of antigen-driven proliferation, tissue homing, and cytotoxic effector functions, with the HLA-DR+ CD38+ subset being the most striking in these effector qualities. The breadth of the dengue-specific CD8 T cell response was diverse, with NS3-specific cells being the most dominant. Interestingly, only a small fraction of these activated effector CD8 T cells produced gamma interferon (IFN-γ) when stimulated with dengue virus peptide pools. Transcriptomics revealed downregulation of key molecules involved in T cell receptor (TCR) signaling. Consistent with this, the majority of these CD8 T cells remained IFN-γ unresponsive even after TCR-dependent polyclonal stimulation (anti-CD3 plus anti-CD28) but produced IFN-γ by TCR-independent polyclonal stimulation (phorbol 12-myristate 13-acetate [PMA] plus ionomycin). Thus, the vast majority of these proliferating, highly differentiated effector CD8 T cells probably acquire TCR refractoriness at the time the patient is experiencing febrile illness that leads to IFN-γ unresponsiveness. Our studies open novel avenues for understanding the mechanisms that fine-tune the balance between CD8 T cell-mediated protective versus pathological effects in dengue. IMPORTANCE: Dengue is becoming a global public health concern. Although CD8 T cells have been implicated both in protection and in the cytokine-mediated immunopathology of dengue, how the balance is maintained between these opposing functions remains unknown. We comprehensively characterized CD8 T cell subsets in dengue patients from India and Thailand and show that these cells expand massively and express phenotypes indicative of overwhelming antigenic stimulus and tissue homing/cytotoxic-effector functions but that a vast majority of them fail to produce IFN-γ in vitro Interestingly, the cells were fully capable of producing the cytokine when stimulated in a T cell receptor (TCR)-independent manner but failed to do so in TCR-dependent stimulation. These results, together with transcriptomics, revealed that the vast majority of these CD8 T cells from dengue patients become cytokine unresponsive due to TCR signaling insufficiencies. These observations open novel avenues for understanding the mechanisms that fine-tune the balance between CD8-mediated protective versus pathological effects.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Dengue Virus/drug effects , T-Lymphocyte Subsets/immunology , Transcriptome/immunology , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/immunology , Adolescent , Antibodies/pharmacology , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/genetics , CD28 Antigens/immunology , CD3 Complex/genetics , CD3 Complex/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/virology , Cell Proliferation/drug effects , Child , Child, Preschool , Dengue Virus/genetics , Dengue Virus/growth & development , Dengue Virus/metabolism , Female , Gene Expression Regulation , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Humans , Immunity, Cellular , India , Infant , Interferon-gamma/genetics , Interferon-gamma/immunology , Ionomycin/pharmacology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Primary Cell Culture , RNA Helicases/genetics , RNA Helicases/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Signal Transduction , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/virology , Tetradecanoylphorbol Acetate/pharmacology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology
15.
Immunity ; 28(5): 710-22, 2008 May.
Article in English | MEDLINE | ID: mdl-18468462

ABSTRACT

To explore the human T cell response to acute viral infection, we performed a longitudinal analysis of CD8(+) T cells responding to the live yellow fever virus and smallpox vaccines--two highly successful human vaccines. Our results show that both vaccines generated a brisk primary effector CD8(+) T cell response of substantial magnitude that could be readily quantitated with a simple set of four phenotypic markers. Secondly, the vaccine-induced T cell response was highly specific with minimal bystander effects. Thirdly, virus-specific CD8(+) T cells passed through an obligate effector phase, contracted more than 90% and gradually differentiated into long-lived memory cells. Finally, these memory cells were highly functional and underwent a memory differentiation program distinct from that described for human CD8(+) T cells specific for persistent viruses. These results provide a benchmark for CD8(+) T cell responses induced by two of the most effective vaccines ever developed.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Smallpox Vaccine/immunology , T-Lymphocyte Subsets/immunology , Yellow Fever Vaccine/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Lymphocyte Activation , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Vaccination , Vaccinia virus/immunology , Yellow Fever Vaccine/metabolism
16.
J Immunol ; 191(2): 540-4, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23772031

ABSTRACT

Ag-specific CD8 T cells play a critical role in controlling HIV infection but eventually lose antiviral functions in part because of expression and signaling through the inhibitory programmed death-1 (PD-1) receptor. To better understand the impact of prolonged TCR ligation on regulation of PD-1 expression in HIV-specific CD8 T cells, we investigated the capacity of virus-specific CD8 T cells to modify the PD-1 epigenetic program after reduction in viral load. We observed that the transcriptional regulatory region was unmethylated in the PD-1(hi) HIV-specific CD8 T cells, whereas it remained methylated in donor-matched naive cells at acute and chronic stages of infection. Surprisingly, the PD-1 promoter remained unmethylated in HIV-specific CD8 T cells from subjects with a viral load controlled by antiviral therapy for >2 y or from elite controllers. Together, these data demonstrate that the epigenetic program at the PD-1 locus becomes fixed after prolonged exposure to HIV virus.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , DNA Methylation , HIV Infections/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Antiretroviral Therapy, Highly Active , CD8-Positive T-Lymphocytes/metabolism , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/immunology , Humans , Promoter Regions, Genetic , Transcription, Genetic , Viral Load
17.
Eur J Immunol ; 43(12): 3219-32, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24030473

ABSTRACT

CD4(+) T follicular helper (TFH) cells are central for generation of long-term B-cell immunity. A defining phenotypic attribute of TFH cells is the expression of the chemokine R CXCR5, and TFH cells are typically identified by co-expression of CXCR5 together with other markers such as PD-1, ICOS, and Bcl-6. Herein, we report high-level expression of the nutrient transporter folate R 4 (FR4) on TFH cells in acute viral infection. Distinct from the expression profile of conventional TFH markers, FR4 was highly expressed by naive CD4(+) T cells, was downregulated after activation and subsequently re-expressed on TFH cells. Furthermore, FR4 expression was maintained, albeit at lower levels, on memory TFH cells. Comparative gene expression profiling of FR4(hi) versus FR4(lo) Ag-specific CD4(+) effector T cells revealed a molecular signature consistent with TFH and TH1 subsets, respectively. Interestingly, genes involved in the purine metabolic pathway, including the ecto-enzyme CD73, were enriched in TFH cells compared with TH1 cells, and phenotypic analysis confirmed expression of CD73 on TFH cells. As there is now considerable interest in developing vaccines that would induce optimal TFH cell responses, the identification of two novel cell surface markers should be useful in characterization and identification of TFH cells following vaccination and infection.


Subject(s)
Gene Expression Regulation/immunology , Receptors, Cell Surface/immunology , T-Lymphocytes, Helper-Inducer/immunology , 5'-Nucleotidase/biosynthesis , 5'-Nucleotidase/genetics , 5'-Nucleotidase/immunology , Acute Disease , Animals , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Gene Expression Regulation/genetics , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/immunology , Mice , Mice, Transgenic , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Proto-Oncogene Proteins c-bcl-6 , Receptors, CXCR5/biosynthesis , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/metabolism
18.
J Virol ; 86(6): 2911-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22238318

ABSTRACT

Humoral immune responses are thought to play a major role in dengue virus-induced immunopathology; however, little is known about the plasmablasts producing these antibodies during an ongoing infection. Herein we present an analysis of plasmablast responses in patients with acute dengue virus infection. We found very potent plasmablast responses that often increased more than 1,000-fold over the baseline levels in healthy volunteers. In many patients, these responses made up as much 30% of the peripheral lymphocyte population. These responses were largely dengue virus specific and almost entirely made up of IgG-secreting cells, and plasmablasts reached very high numbers at a time after fever onset that generally coincided with the window where the most serious dengue virus-induced pathology is observed. The presence of these large, rapid, and virus-specific plasmablast responses raises the question as to whether these cells might have a role in dengue immunopathology during the ongoing infection. These findings clearly illustrate the need for a detailed understanding of the repertoire and specificity of the antibodies that these plasmablasts produce.


Subject(s)
Dengue Virus/physiology , Dengue/immunology , Immunity, Humoral , Plasma Cells/immunology , Acute Disease , Adolescent , Adult , Antibodies, Viral/immunology , Child , Child, Preschool , Cohort Studies , Dengue/virology , Dengue Virus/immunology , Female , Humans , Infant , Male , Middle Aged , Plasma Cells/virology , Species Specificity , Young Adult
19.
J Infect Dis ; 206(9): 1372-85, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22904336

ABSTRACT

Progressive vaccinia (PV) is a rare but potentially lethal complication that develops in smallpox vaccine recipients with severely impaired cellular immunity. We describe a patient with PV who required treatment with vaccinia immune globulin and who received 2 investigational agents, ST-246 and CMX001. We describe the various molecular, pharmacokinetic, and immunologic studies that provided guidance to escalate and then successfully discontinue therapy. Despite development of resistance to ST-246 during treatment, the patient had resolution of PV. This case demonstrates the need for continued development of novel anti-orthopoxvirus pharmaceuticals and the importance of both intensive and timely clinical and laboratory support in management of PV.


Subject(s)
Antibodies, Viral/administration & dosage , Antiviral Agents/administration & dosage , Benzamides/administration & dosage , Cytosine/analogs & derivatives , Isoindoles/administration & dosage , Organophosphonates/administration & dosage , Vaccinia virus/isolation & purification , Vaccinia/diagnosis , Vaccinia/drug therapy , Adult , Antiviral Agents/pharmacology , Cytosine/administration & dosage , Drug Resistance, Viral , Humans , Immunoglobulins/administration & dosage , Male , Smallpox Vaccine/administration & dosage , Smallpox Vaccine/adverse effects , Treatment Outcome
20.
J Virol ; 85(22): 11770-80, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21900169

ABSTRACT

Although cellular immunity to acute lymphocytic choriomeningitis virus (LCMV) infection has been well characterized in experimental studies in mice, the T cell response to this virus in humans is incompletely understood. Thus, we analyzed the breadths, magnitudes, and differentiation phenotypes of memory LCMV-specific CD8(+) and CD4(+) T cells in three human donors displaying a variety of disease outcomes after accidental needle stick injury or exposure to LCMV. Although only a small cohort of donors was analyzed at a single time point postinfection, several interesting observations were made. First, we were able to detect LCMV-specific CD8(+) and CD4(+) T cell responses directly ex vivo at 4 to 8 years after exposure, demonstrating the longevity of T cell memory in humans. Second, unlike in murine models of LCMV infection, we found that the breadths of memory CD8(+) and CD4(+) T cell responses were not significantly different from one another. Third, it seemed that the overall CD8(+) T cell response was augmented with increasing severity of disease, while the LCMV-specific CD4(+) T cell response magnitude was highly variable between the three different donors. Next, we found that LCMV-specific CD8(+) T cells in the three donors analyzed seemed to undergo an effector memory differentiation program distinct from that of CD4(+) T cells. Finally, the levels of expression of memory, costimulatory, and inhibitory receptors on CD8(+) and CD4(+) T cell subsets, in some instances, correlated with disease outcome. These data demonstrate for the first time LCMV-specific CD8(+) and CD4(+) T cells in infected humans and begin to provide new insights into memory T cell responses following an acute virus infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Adult , Human Experimentation , Humans , Male , Needlestick Injuries/complications , Occupational Exposure/adverse effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL