Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Invest ; 121(10): 3877-88, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21926463

ABSTRACT

Loss of memory B cells occurs from the onset of HIV-1 infection and persists into the chronic stages of infection. Lack of survival of these cells, even in subjects being treated, could primarily be the consequence of an altered local microenvironment induced by HIV infection. In this study we showed that memory B cell survival was significantly decreased in aviremic successfully treated (ST) subjects compared with subjects who control viral load as a result of natural immunity (elite controller [EC]) or with uninfected control (HIV-) subjects. The lower survival levels observed in memory B cells from ST subjects were the result of disrupted IL-2 signaling that led to increased transcriptional activity of Foxo3a and increased expression of its proapoptotic target TRAIL. Notably, memory B cell survival in ST subjects was significantly enhanced by the addition of exogenous IL-2 in a Foxo3a-dependent manner. We further showed that Foxo3a silencing by siRNA resulted in decreased expression of TRAIL and apoptosis levels in memory B cells from ST subjects. Our results thus establish a direct role for Foxo3a/TRAIL signaling in the persistence of memory B cells and provide a mechanism for the reduced survival of memory B cells during HIV infection. This knowledge could be exploited for the development of therapeutic and preventative HIV vaccines.


Subject(s)
B-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , HIV Infections/immunology , Immunologic Memory , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis/immunology , B-Lymphocytes/metabolism , Case-Control Studies , Cell Survival/immunology , Chronic Disease , Forkhead Box Protein O3 , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/genetics , HIV Infections/metabolism , HIV Infections/pathology , HIV Long-Term Survivors , HIV-1 , Humans , Interleukin-2/blood , Interleukin-2/pharmacology , RNA, Small Interfering/genetics , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors
2.
Science ; 332(6030): 687-96, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21551058

ABSTRACT

Flow cytometry is an essential tool for dissecting the functional complexity of hematopoiesis. We used single-cell "mass cytometry" to examine healthy human bone marrow, measuring 34 parameters simultaneously in single cells (binding of 31 antibodies, viability, DNA content, and relative cell size). The signaling behavior of cell subsets spanning a defined hematopoietic hierarchy was monitored with 18 simultaneous markers of functional signaling states perturbed by a set of ex vivo stimuli and inhibitors. The data set allowed for an algorithmically driven assembly of related cell types defined by surface antigen expression, providing a superimposable map of cell signaling responses in combination with drug inhibition. Visualized in this manner, the analysis revealed previously unappreciated instances of both precise signaling responses that were bounded within conventionally defined cell subsets and more continuous phosphorylation responses that crossed cell population boundaries in unexpected manners yet tracked closely with cellular phenotype. Collectively, such single-cell analyses provide system-wide views of immune signaling in healthy human hematopoiesis, against which drug action and disease can be compared for mechanistic studies and pharmacologic intervention.


Subject(s)
Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Flow Cytometry/methods , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Pyrimidines/pharmacology , Signal Transduction , Single-Cell Analysis/methods , Thiazoles/pharmacology , Algorithms , Antibodies , Antigens, Surface/analysis , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cytokines/metabolism , Dasatinib , Hematopoiesis , Humans , Immunophenotyping , Lanthanoid Series Elements , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation , Lymphocyte Subsets/metabolism , Mass Spectrometry , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transition Elements
3.
PLoS One ; 6(4): e19038, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21541329

ABSTRACT

Signaling events affecting thymic selection of un-manipulated polyclonal natural CD25(+)foxp3(+) regulatory T cells (nTreg) have not been established ex vivo. Here, we report a higher frequency of phosphorylated STAT-5 (pSTAT-5) in nTreg cells in the adult murine thymus and to a lesser extent in the periphery, compared to other CD4(+)CD8(-) subsets. In the neonatal thymus, the numbers of pSTAT-5(+) cells in CD25(+)foxp3(-) and nTreg cells increased in parallel, suggesting that pSTAT-5(+)CD25(+)foxp3(-) cells might represent the precursors of foxp3(+) regulatory T cells. This "specific" pSTAT-5 expression detected in nTreg cells ex vivo was likely due to a very recent signal given by IL-2/IL-15 cytokines in vivo since (i) it disappeared rapidly if cells were left unstimulated in vitro and (ii) was also observed if total thymocytes were stimulated in vitro with saturating amounts of IL-2 and/or IL-15 but not IL-7. Interestingly, STAT-5 activation upon IL-2 stimulation correlated better with foxp3 and CD122 than with CD25 expression. Finally, we show that expression of an endogenous superantigen strongly affected the early Treg cell repertoire but not the proportion of pSTAT-5(+) cells within this repertoire. Our results reveal that continuous activation of the CD122/STAT-5 signaling pathway characterize regulatory lineage differentiation in the murine thymus.


Subject(s)
Epitopes/immunology , Interleukin-2 Receptor beta Subunit/immunology , STAT5 Transcription Factor/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/cytology , Aging/metabolism , Animals , Animals, Newborn , Cell Differentiation/immunology , Cell Lineage , Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Kinetics , Mice , Phosphorylation , Thymus Gland/immunology
4.
J Exp Med ; 205(13): 3119-31, 2008 Dec 22.
Article in English | MEDLINE | ID: mdl-19047440

ABSTRACT

Correlates of immune-mediated protection to most viral and cancer vaccines are still unknown. This impedes the development of novel vaccines to incurable diseases such as HIV and cancer. In this study, we have used functional genomics and polychromatic flow cytometry to define the signature of the immune response to the yellow fever (YF) vaccine 17D (YF17D) in a cohort of 40 volunteers followed for up to 1 yr after vaccination. We show that immunization with YF17D leads to an integrated immune response that includes several effector arms of innate immunity, including complement, the inflammasome, and interferons, as well as adaptive immunity as shown by an early T cell response followed by a brisk and variable B cell response. Development of these responses is preceded, as demonstrated in three independent vaccination trials and in a novel in vitro system of primary immune responses (modular immune in vitro construct [MIMIC] system), by the coordinated up-regulation of transcripts for specific transcription factors, including STAT1, IRF7, and ETS2, which are upstream of the different effector arms of the immune response. These results clearly show that the immune response to a strong vaccine is preceded by coordinated induction of master transcription factors that lead to the development of a broad, polyfunctional, and persistent immune response that integrates all effector cells of the immune system.


Subject(s)
Gene Expression Regulation/immunology , Immune System Phenomena , Immunity, Innate/immunology , Vaccination , Yellow Fever Vaccine/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Proliferation , Flow Cytometry , Gene Expression Profiling , Gene Regulatory Networks , Humans , Interleukin-1beta/immunology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/physiology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transcription, Genetic
5.
Nat Med ; 14(3): 266-74, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18311149

ABSTRACT

The persistence of central memory CD4(+) T cells (T(CM) cells) is a major correlate of immunological protection in HIV/AIDS, as the rate of T(CM) cell decline predicts HIV disease progression. In this study, we show that T(CM) cells and effector memory CD4(+) T cells (T(EM) cells) from HIV(+) elite controller (EC) subjects are less susceptible to Fas-mediated apoptosis and persist longer after multiple rounds of T cell receptor triggering when compared to T(CM) and T(EM) cells from aviremic successfully treated (ST) subjects or from HIV(-) donors. We show that persistence of T(CM) cells from EC subjects is a direct consequence of inactivation of the FOXO3a pathway. Silencing the transcriptionally active form of FOXO3a by small interfering RNA or by introducing a FOXO3a dominant-negative form (FOXO3a Nt) extended the long-term survival of T(CM) cells from ST subjects to a length of time similar to that of T(CM) cells from EC subjects. The crucial role of FOXO3a in the survival of memory cells will help shed light on the underlying immunological mechanisms that control viral replication in EC subjects.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , HIV Infections/immunology , HIV Infections/metabolism , Immunologic Memory , Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active , Apoptosis/physiology , CD4-Positive T-Lymphocytes/cytology , Dendritic Cells/immunology , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation , HIV Infections/drug therapy , Humans , Jurkat Cells , RNA, Small Interfering , Virus Replication
6.
J Exp Med ; 204(1): 79-91, 2007 Jan 22.
Article in English | MEDLINE | ID: mdl-17190839

ABSTRACT

The molecular events involved in the establishment and maintenance of CD4+ central memory and effector memory T cells (TCM and TEM, respectively) are poorly understood. In this study, we demonstrate that ex vivo isolated TCM are more resistant to both spontaneous and Fas-induced apoptosis than TEM and have an increased capacity to proliferate and persist in vitro. Using global gene expression profiling, single cell proteomics, and functional assays, we show that the survival of CD4+ TCM depends, at least in part, on the activation and phosphorylation of signal transducer and activator of transcription 5a (STAT5a) and forkhead box O3a (FOXO3a). TCM showed a significant increase in the levels of phosphorylation of STAT5a compared with TEM in response to both IL-2 (P<0.04) and IL-7 (P<0.002); the latter is well known for its capacity to enhance T cell survival. Moreover, ex vivo TCM express higher levels of the transcriptionally inactive phosphorylated forms of FOXO3a and concomitantly lower levels of the proapoptotic FOXO3a target, Bim. Experiments aimed at blocking FOXO3a phosphorylation confirmed the role of this phosphoprotein in protecting TCM from apoptosis. Our results provide, for the first time in humans, an insight into molecular mechanisms that could be responsible for the longevity and persistence of CD4+ TCM.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , Receptors, Antigen, T-Cell/metabolism , Apoptosis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Survival , Dendritic Cells/immunology , Forkhead Box Protein O3 , Gene Expression Profiling , Humans , I-kappa B Kinase/antagonists & inhibitors , Immunologic Memory , In Vitro Techniques , Lymphocyte Activation , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Suppressor Proteins , fas Receptor/metabolism
7.
Nat Med ; 12(10): 1198-202, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16917489

ABSTRACT

The engagement of programmed death 1 (PD-1) to its ligands, PD-L1 and PD-L2, inhibits proliferation and cytokine production mediated by antibodies to CD3 (refs. 5,6,7). Blocking the PD-1-PD-L1 pathway in mice chronically infected with lymphocytic choriomeningitis virus restores the capacity of exhausted CD8(+) T cells to undergo proliferation, cytokine production and cytotoxic activity and, consequently, results in reduced viral load. During chronic HIV infection, HIV-specific CD8(+) T cells are functionally impaired, showing a reduced capacity to produce cytokines and effector molecules as well as an impaired capacity to proliferate. Here, we found that PD-1 was upregulated on HIV-specific CD8(+) T cells; PD-1 expression levels were significantly correlated both with viral load and with the reduced capacity for cytokine production and proliferation of HIV-specific CD8(+) T cells. Notably, cytomegalovirus (CMV)-specific CD8(+) T cells from the same donors did not upregulate PD-1 and maintained the production of high levels of cytokines. Blocking PD-1 engagement to its ligand (PD-L1) enhanced the capacity of HIV-specific CD8(+) T cells to survive and proliferate and led to an increased production of cytokines and cytotoxic molecules in response to cognate antigen. The accumulation of HIV-specific dysfunctional CD8(+) T cells in the infected host could prevent the renewal of a functionally competent HIV-specific CD8(+) repertoire.


Subject(s)
Antigens, CD/biosynthesis , Apoptosis Regulatory Proteins/biosynthesis , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , HIV Infections/blood , HIV Infections/metabolism , Immune System Diseases/immunology , Immune System/pathology , Up-Regulation , Amino Acid Sequence , CD3 Complex/biosynthesis , Cell Differentiation , Cell Proliferation , Cytokines/metabolism , Humans , Immune System Diseases/pathology , Immunophenotyping , Molecular Sequence Data , Programmed Cell Death 1 Receptor , Receptors, Antigen, T-Cell/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...