Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Immunol ; 203(4): 899-910, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31285277

ABSTRACT

The kynurenine pathway (KP) is a key regulator of many important physiological processes and plays a harmful role in cancer, many neurologic conditions, and chronic viral infections. In HIV infection, KP activity is consistently associated with reduced CD4 T cell counts and elevated levels of T cell activation and viral load; it also independently predicts mortality and morbidity from non-AIDS events. Kynurenine 3-monooxygenase (KMO) is a therapeutically important target in the KP. Using the nonhuman primate model of SIV infection in rhesus macaques, we investigated whether KMO inhibition could slow the course of disease progression. We used a KMO inhibitor, CHDI-340246, to perturb the KP during early acute infection and followed the animals for 1 y to assess clinical outcomes and immune phenotype and function during pre-combination antiretroviral therapy acute infection and combination antiretroviral therapy-treated chronic infection. Inhibition of KMO in acute SIV infection disrupted the KP and prevented SIV-induced increases in downstream metabolites, improving clinical outcome as measured by both increased CD4+ T cell counts and body weight. KMO inhibition increased naive T cell frequency and lowered PD-1 expression in naive and memory T cell subsets. Importantly, early PD-1 expression during acute SIV infection predicted clinical outcomes of body weight and CD4+ T cell counts. Our data indicate that KMO inhibition in early acute SIV infection provides clinical benefit and suggest a rationale for testing KMO inhibition as an adjunctive treatment in SIV/HIV infection to slow the progression of the disease and improve immune reconstitution.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Programmed Cell Death 1 Receptor/biosynthesis , Pyrimidines/pharmacology , Simian Acquired Immunodeficiency Syndrome/immunology , Animals , Anti-Retroviral Agents/pharmacology , Body Weight/drug effects , CD4-Positive T-Lymphocytes/drug effects , Enzyme Inhibitors/pharmacology , Macaca mulatta , Programmed Cell Death 1 Receptor/drug effects , Simian Acquired Immunodeficiency Syndrome/metabolism
2.
J Virol ; 90(14): 6255-6262, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27122585

ABSTRACT

UNLABELLED: Strategies aimed at eliminating persistent viral reservoirs from HIV-1-infected individuals have focused on CD4(+) T-cell reservoirs. However, very little attention has been given to approaches that could promote elimination of tissue macrophage reservoirs. HIV-1 infection of macrophages induces phosphorylation of colony-stimulating factor 1 receptor (CSF-1R), which confers resistance to apoptotic pathways driven by tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), thereby promoting viral persistence. In this study, we assessed whether CSF-1R antagonists (PLX647, PLX3397, and PLX5622) restored apoptotic sensitivity of HIV-1-infected macrophages in vitro PLX647, PLX3397, and PLX5622 at clinically relevant concentrations blocked the activation of CSF-1R and reduced the viability of infected macrophages, as well as the extent of viral replication. Our data show that strategies targeting monocyte colony-stimulating factor (MCSF) signaling could be used to promote elimination of HIV-1-infected myeloid cells and to contribute to the elimination of persistent viral reservoirs. IMPORTANCE: As the HIV/AIDS research field explores approaches to eliminate HIV-1 in individuals on suppressive antiviral therapy, those approaches will need to eliminate both CD4(+) T-cell and myeloid cell reservoirs. Most of the attention has focused on CD4(+) T-cell reservoirs, and scant attention has been paid to myeloid cell reservoirs. The distinct nature of the infection in myeloid cells versus CD4(+) T cells will likely dictate different approaches in order to achieve their elimination. For CD4(+) T cells, most strategies focus on promoting virus reactivation to promote immune-mediated clearance and/or elimination by viral cytopathicity. Macrophages resist viral cytopathic effects and CD8(+) T-cell killing. Therefore, we have explored clearance strategies that render macrophages sensitive to viral cytopathicity. This research helps inform the design of strategies to promote clearance of the macrophage reservoir in infected individuals on suppressive antiviral therapy.


Subject(s)
Aminopyridines/pharmacology , HIV Infections/immunology , HIV-1/immunology , Macrophages/immunology , Pyrroles/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , TNF-Related Apoptosis-Inducing Ligand/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , HIV Infections/drug therapy , HIV Infections/virology , Humans , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/drug effects , Macrophages/pathology , Macrophages/virology , Virus Latency , Virus Replication/drug effects , Virus Replication/immunology
3.
Proc Natl Acad Sci U S A ; 109(7): 2549-54, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22308487

ABSTRACT

Cell cycle entry is commonly considered to positively regulate HIV-1 infection of CD4 T cells, raising the question as to how quiescent lymphocytes, representing a large portion of the viral reservoir, are infected in vivo. Factors such as the homeostatic cytokine IL-7 have been shown to render quiescent T cells permissive to HIV-1 infection, presumably by transiently stimulating their entry into the cell cycle. However, we show here that at physiological oxygen (O(2)) levels (2-5% O(2) tension in lymphoid organs), IL-7 stimulation generates an environment permissive to HIV-1 infection, despite a significantly attenuated level of cell cycle entry. We identify the IL-7-induced increase in Glut1 expression, resulting in augmented glucose uptake, as a key factor in rendering these T lymphocytes susceptible to HIV-1 infection. HIV-1 infection of human T cells is abrogated either by impairment of Glut1 signal transduction or by siRNA-mediated Glut1 down-regulation. Consistent with this, we show that the susceptibility of human thymocyte subsets to HIV-1 infection correlates with Glut1 expression; single-round infection is markedly higher in the Glut1-expressing double-positive thymocyte population than in any of the Glut1-negative subsets. Thus, our studies reveal the Glut1-mediated metabolic pathway as a critical regulator of HIV-1 infection in human CD4 T cells and thymocytes.


Subject(s)
Glucose Transporter Type 1/metabolism , Glucose/metabolism , HIV Infections/physiopathology , Adult , Biological Transport , CD4-Positive T-Lymphocytes/metabolism , Humans , Signal Transduction
4.
Semin Immunol ; 22(5): 270-5, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20630771

ABSTRACT

The thymus forms as an alymphoid thymic primordium with T cell differentiation requiring the seeding of this anlage. This review will focus on the characteristics of the hematopoietic progenitors which colonize the thymus and their subsequent commitment/differentiation, both in mice and men. Within the thymus, the interplay between Notch1 and IL-7 signals is crucial for the orchestration of T cell development, but the precise requirements for these factors in murine and human thympoeisis are not synonymous. Recent advances in our understanding of the mechanisms regulating precursor entry and their maintenance in the thymus will also be presented.


Subject(s)
Cell Lineage , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Cell Differentiation , Humans , Interleukin-7/immunology , Interleukin-7/metabolism , Receptor, Notch1/immunology , Receptor, Notch1/metabolism
5.
Arthritis Rheum ; 64(8): 2451-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22392608

ABSTRACT

OBJECTIVE: CD4+FoxP3+ Treg cells suppress effector T cells and prevent autoimmune disease. Treg cell function is deficient in active rheumatoid arthritis (RA), a loss which may play a role in the pathogenesis of this disease. We previously showed that a single-nucleotide polymorphism in the FCRL3 gene led to higher expression of Fc receptor-like 3 (FcRL3) on Treg cells and that FcRL3+ Treg cells are functionally deficient in comparison to FcRL3- Treg cells. This study was undertaken to investigate the potential role of FcRL3 in RA. METHODS: A cross-sectional study was performed to evaluate the FCRL3 -169 genotype and FcRL3 expression on T cell subsets, including Treg cells, in peripheral blood samples from 51 patients with RA enrolled in the University of California, San Francisco (UCSF) RA Cohort. Clinical data were obtained from the UCSF RA Cohort database. RESULTS: Patients with the FCRL3 -169C allele (genotype C/C or C/T) expressed higher levels of FcRL3 on Treg cells, and on CD8+ and γ/δ T cells, in comparison to RA patients with the T/T genotype. Higher FcRL3 expression on these T cell subpopulations correlated with RA disease activity in patients harboring the FCRL3 -169C allele. Furthermore, FcRL3 expression on Treg cells was higher in patients with erosive RA, and the FCRL3 -169C allele was overrepresented in patients with erosive RA. CONCLUSION: Our findings indicate that FcRL3 expression, which is strongly associated with the presence of the FCRL3 -169C allele, may serve as a biomarker for RA disease activity.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Polymorphism, Single Nucleotide/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Severity of Illness Index , T-Lymphocyte Subsets/metabolism , Adult , Alleles , Arthritis, Rheumatoid/pathology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Biomarkers/metabolism , CD8 Antigens/metabolism , Cell Count , Cross-Sectional Studies , Female , Genotype , Humans , Male , Middle Aged , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
6.
J Immunol ; 184(7): 3639-47, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20190142

ABSTRACT

CD4(+)FoxP3(+) regulatory T cells (T(reg)) play a critical role in maintaining self-tolerance and inhibiting autoimmune disease. Despite being a major focus of modern immunological investigation, many aspects of T(reg) biology remain unknown. In a screen for novel candidate genes involved in human T(reg) function, we detected the expression of an autoimmune susceptibility gene, FcRL3, in T(reg) but not in conventional CD4(+) T cells. FcRL3 is an orphan receptor of unknown function with structural homology to classical Fc receptors. Numerous genetic studies have demonstrated a link between a single nucleotide polymorphism in the FCRL3 promoter and both overexpression of FcRL3 and autoimmune diseases such as rheumatoid arthritis. Given the critical role of T(reg) in suppressing autoimmunity, we sought to ascertain how expression of FcRL3 relates to the phenotype, differentiation, and function of T(reg). We show in this study that FcRL3 is expressed on a population of thymically derived T(reg) that exhibits a memory phenotype and high levels of programmed cell death-1. Purified FcRL3(+) T(reg) are less responsive to antigenic stimulation in the presence of IL-2 than their FcRL3(-) counterparts, despite intact proximal and distal IL-2 signaling as determined by phosphorylation of Stat-5 and upregulation of Bcl2. In vitro suppression assays demonstrated that FcRL3(+) T(reg) have reduced capacity to suppress the proliferation of effector T cells. These data suggest that FcRL3 expression is associated with T(reg) dysfunction that may, in turn, contribute to the loss of self-tolerance and the development of autoimmunity.


Subject(s)
Antigens, CD/immunology , Apoptosis Regulatory Proteins/immunology , Receptors, Immunologic/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Antigens, CD/biosynthesis , Apoptosis Regulatory Proteins/biosynthesis , Cell Differentiation/immunology , Cell Separation , Flow Cytometry , Humans , Lymphocyte Activation/immunology , Phenotype , Polymorphism, Single Nucleotide , Programmed Cell Death 1 Receptor , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Self Tolerance/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism
7.
JCI Insight ; 7(5)2022 03 08.
Article in English | MEDLINE | ID: mdl-35104248

ABSTRACT

Type I IFNs (TI-IFNs) drive immune effector functions during acute viral infections and regulate cell cycling and systemic metabolism. That said, chronic TI-IFN signaling in the context of HIV infection treated with antiretroviral therapy (ART) also facilitates viral persistence, in part by promoting immunosuppressive responses and CD8+ T cell exhaustion. To determine whether inhibition of IFN-α might provide benefit in the setting of chronic, ART-treated SIV infection of rhesus macaques, we administered an anti-IFN-α antibody followed by an analytical treatment interruption (ATI). IFN-α blockade was well-tolerated and associated with lower expression of TI-IFN-inducible genes (including those that are antiviral) and reduced tissue viral DNA (vDNA). The reduction in vDNA was further accompanied by higher innate proinflammatory plasma cytokines, expression of monocyte activation genes, IL-12-induced effector CD8+ T cell genes, increased heme/metabolic activity, and lower plasma TGF-ß levels. Upon ATI, SIV-infected, ART-suppressed nonhuman primates treated with anti-IFN-α displayed lower levels of weight loss and improved erythroid function relative to untreated controls. Overall, these data demonstrated that IFN-α blockade during ART-treated SIV infection was safe and associated with the induction of immune/erythroid pathways that reduced viral persistence during ART while mitigating the weight loss and anemia that typically ensue after ART interruption.


Subject(s)
HIV Infections , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Anti-Retroviral Agents/pharmacology , Anti-Retroviral Agents/therapeutic use , DNA, Viral , HIV Infections/drug therapy , Immunity , Interferon-alpha , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/drug therapy , Weight Loss
8.
PLoS Pathog ; 5(2): e1000295, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19214220

ABSTRACT

Chronic immune activation and progression to AIDS are observed after SIV infection in macaques but not in natural host primate species. To better understand this dichotomy, we compared acute pathogenic SIV infection in pigtailed macaques (PTs) to non-pathogenic infection in African green monkeys (AGMs). SIVagm-infected PTs, but not SIVagm-infected AGMs, rapidly developed systemic immune activation, marked and selective depletion of IL-17-secreting (Th17) cells, and loss of the balance between Th17 and T regulatory (Treg) cells in blood, lymphoid organs, and mucosal tissue. The loss of Th17 cells was found to be predictive of systemic and sustained T cell activation. Collectively, these data indicate that loss of the Th17 to Treg balance is related to SIV disease progression.


Subject(s)
Interleukin-17/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Analysis of Variance , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Chlorocebus aethiops , Colon/immunology , Colon/metabolism , Cytokines/immunology , Cytokines/metabolism , Flow Cytometry , Forkhead Transcription Factors/metabolism , Interleukin-17/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Activation , Macaca nemestrina , Male , Oligonucleotide Array Sequence Analysis , RNA, Viral/analysis , RNA, Viral/blood , RNA, Viral/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus , Statistics, Nonparametric , T-Lymphocytes, Helper-Inducer/virology , T-Lymphocytes, Regulatory/virology , Viral Load
9.
Methods Mol Biol ; 506: 171-90, 2009.
Article in English | MEDLINE | ID: mdl-19110627

ABSTRACT

The thymus provides a specialized environment allowing the differentiation of T lymphocytes from bone marrow-derived progenitor cells. We and others have demonstrated that gene transfer into distinct thymocyte populations can be obtained, both in vivo and ex vivo, using lentiviral vectors. Here, we describe techniques for intrathymic lentiviral transduction in mice, using a surgical approach wherein the thoracic cavity is exposed as well as a significantly less invasive strategy wherein virions are directly injected through the skin. Moreover, thymocyte differentiation from murine and human progenitors is now feasible in vitro, under conditions wherein the Notch and IL-7 signaling pathways are activated. We describe methods allowing transduction of murine and human progenitors and their subsequent differentiation into more mature thymocytes. Conditions for lentiviral gene transfer into more differentiated human thymocyte subsets are also presented. Optimization of technologies for HIV-based gene transfer into murine and human thymocyte progenitors will advance strategies aimed at modulating T-cell differentiation and function in-vivo; approaches potentially targeting patients with genetic and acquired immunodeficiencies as well as immune-sensitive tumors. Furthermore, this technology will foster the progression of basic research aimed at elucidating molecular aspects of T-cell differentiation in mice and humans.


Subject(s)
Gene Transfer Techniques , Thymus Gland/metabolism , Animals , Antigens, CD34/immunology , Base Sequence , Cell Line , DNA Primers , Humans , Mice , Thymus Gland/cytology , Thymus Gland/immunology , Transduction, Genetic
10.
Methods Mol Biol ; 415: 301-20, 2008.
Article in English | MEDLINE | ID: mdl-18370162

ABSTRACT

Gene transfer into mammalian cells has been of crucial importance for studies determining the role of specific genes in the differentiation and cell fate of various hematopoietic lineages. Until recently, the majority of these studies were performed in transformed cell lines due to difficulties in achieving levels of transfection of greater than 1-3% in primary hematopoietic cells. Vectors based on retrovirus and lentivirus backbones have revolutionized our ability to transfer genes into primary hematopoietic cells. These vectors have allowed extensive ex vivo and in vivo studies following introduction of a gene of interest and have been used clinically in individuals suffering from cancers, infections, and genetic diseases. Ex vivo lentiviral gene transfer can result in efficient transduction of progenitor cells (>80%) that can then be further differentiated into immune lineage cells including T, B, dendritic, or natural killer cells. Alternatively, differentiated immune cells can themselves be transduced ex vivo with lentiviral vectors. Here, we discuss optimization of technologies for human immunodeficiency virus (HIV)-based gene transfer into murine and human progenitor and immune cell lineages.


Subject(s)
Lentivirus/genetics , Leukocytes, Mononuclear/virology , Transduction, Genetic/methods , Animals , B-Lymphocytes/virology , Cells, Cultured , Centrifugation , Dendritic Cells/virology , Genetic Vectors/genetics , Hematopoietic Stem Cells/virology , Mice , Plasmids/genetics , Promoter Regions, Genetic/genetics , T-Lymphocytes/virology , Titrimetry , Transfection , Transgenes , Virion/metabolism
11.
Retrovirology ; 4: 31, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17504522

ABSTRACT

BACKGROUND: We previously identified the glucose transporter Glut-1, a member of the multimembrane-spanning facilitative nutrient transporter family, as a receptor for both HTLV-1 and HTLV-2. However, a recent report concluded that Glut-1 cannot serve as a receptor for HTLV-1 on CD4 T cells: This was based mainly on their inability to detect Glut-1 on this lymphocyte subset using the commercial antibody mAb1418. It was therefore of significant interest to thoroughly assess Glut-1 expression on CD4 and CD8 T cells, and its association with HTLV-1 and -2 envelope binding. RESULTS: As previously reported, ectopic expression of Glut-1 but not Glut-3 resulted in significantly augmented binding of tagged proteins harboring the receptor binding domains of either HTLV-1 or HTLV-2 envelope glycoproteins (H1RBD or H2RBD). Using antibodies raised against the carboxy-terminal peptide of Glut-1, we found that Glut-1 expression was significantly increased in both CD4 and CD8 cells following TCR stimulation. Corresponding increases in the binding of H1RBD as well as H2RBD, not detected on quiescent T cells, were observed following TCR engagement. Furthermore, increased Glut-1 expression was accompanied by a massive augmentation in glucose uptake in TCR-stimulated CD4 and CD8 lymphocytes. Finally, we determined that the apparent contradictory results obtained by Takenouchi et al were due to their monitoring of Glut-1 with a mAb that does not bind cells expressing endogenous Glut-1, including human erythrocytes that harbor 300,000 copies per cell. CONCLUSION: Transfection of Glut-1 directly correlates with the capacities of HTLV-1 and HTLV-2 envelope-derived ligands to bind cells. Moreover, Glut-1 is induced by TCR engagement, resulting in massive increases in glucose uptake and binding of HTLV-1 and -2 envelopes to both CD4 and CD8 T lymphocytes. Therefore, Glut-1 is a primary binding receptor for HTLV-1 and HTLV-2 envelopes on activated CD4 as well as CD8 lymphocytes.


Subject(s)
CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Excitatory Amino Acid Transporter 2/metabolism , Human T-lymphotropic virus 1/physiology , Human T-lymphotropic virus 2/physiology , Viral Envelope Proteins/metabolism , Virus Attachment , Antibodies, Monoclonal/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cells, Cultured , Excitatory Amino Acid Transporter 2/biosynthesis , Flow Cytometry , Gene Expression Regulation , Glucose/metabolism , Humans , Lymphocyte Activation , Protein Binding , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/immunology , Receptors, Virus/metabolism
12.
Nat Med ; 23(11): 1271-1276, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28967921

ABSTRACT

In the quest for a functional cure or the eradication of HIV infection, it is necessary to know the sizes of the reservoirs from which infection rebounds after treatment interruption. Thus, we quantified SIV and HIV tissue burdens in tissues of infected nonhuman primates and lymphoid tissue (LT) biopsies from infected humans. Before antiretroviral therapy (ART), LTs contained >98% of the SIV RNA+ and DNA+ cells. With ART, the numbers of virus (v) RNA+ cells substantially decreased but remained detectable, and their persistence was associated with relatively lower drug concentrations in LT than in peripheral blood. Prolonged ART also decreased the levels of SIV- and HIV-DNA+ cells, but the estimated size of the residual tissue burden of 108 vDNA+ cells potentially containing replication-competent proviruses, along with evidence of continuing virus production in LT despite ART, indicated two important sources for rebound following treatment interruption. The large sizes of these tissue reservoirs underscore challenges in developing 'HIV cure' strategies targeting multiple sources of virus production.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Infections/virology , HIV/isolation & purification , Viral Load , DNA, Viral/analysis , HIV/genetics , HIV Infections/blood , Humans , Lymphoid Tissue/virology , RNA, Viral/analysis
13.
Sci Transl Med ; 7(300): 300ra125, 2015 Aug 12.
Article in English | MEDLINE | ID: mdl-26268312

ABSTRACT

HIV disease progression appears to be driven by increased immune activation. Given observations that fetal exposure to infectious pathogens in utero can result in reduced immune responses, or tolerance, to those pathogens postnatally, we hypothesized that fetal exposure to HIV may render the fetus tolerant to the virus, thus reducing damage caused by immune activation during infection later in life. To test this hypothesis, fetal rhesus macaques (Macaca mulatta) were injected with the attenuated virus SIVmac1A11 in utero and challenged with pathogenic SIVmac239 1 year after birth. SIVmac1A11-injected animals had significantly reduced plasma RNA viral loads (P < 0.02) up to 35 weeks after infection. Generalized estimating equations analysis was performed to identify immunologic and clinical measurements associated with plasma RNA viral load. A positive association with plasma RNA viral load was observed with the proportion of CD8(+) T cells expressing the transcription factor, FoxP3, and the proportion of CD4(+) T cells producing the lymphoproliferative cytokine, IL-2. In contrast, an inverse relationship was found with the frequencies of circulating CD4(+) and CD8(+) T cells displaying intermediate expression of the proliferation marker, Ki-67. Animals exposed to simian immunodeficiency virus (SIV) in utero appeared to have enhanced SIV-specific immune responses, a lower proportion of CD8(+) T cells expressing the exhaustion marker PD-1, and more circulating TH17 cells than controls. Although the development of tolerance was not demonstrated, these data suggest that rhesus monkeys exposed to SIVmac1A11 in utero had distinct immune responses associated with the control of viral replication after postnatal challenge.


Subject(s)
Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , Viral Load/immunology , Administration, Oral , Animals , Animals, Newborn , Cell Count , Epitopes/immunology , Female , Immune Tolerance , Ki-67 Antigen/metabolism , Macaca mulatta , RNA, Viral/blood , Simian Acquired Immunodeficiency Syndrome/blood , Th17 Cells/immunology
14.
Cell Rep ; 13(8): 1589-97, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26586432

ABSTRACT

Gut microbes can profoundly modulate mucosal barrier-promoting Th17 cells in mammals. A salient feature of HIV/simian immunodeficiency virus (SIV) immunopathogenesis is the loss of Th17 cells, which has been linked to increased activity of the immunomodulatory enzyme, indoleamine 2,3-dioxygenase 1 (IDO 1). The role of gut microbes in this system remains unknown, and the SIV-infected rhesus macaque provides a well-described model for HIV-associated Th17 loss and mucosal immune disruption. We observed a specific depletion of gut-resident Lactobacillus during acute and chronic SIV infection of rhesus macaques, which was also seen in early HIV-infected humans. This depletion in rhesus macaques correlated with increased IDO1 activity and Th17 loss. Macaques supplemented with a Lactobacillus-containing probiotic exhibited decreased IDO1 activity during chronic SIV infection. We propose that Lactobacillus species inhibit mammalian IDO1 and thus may help to preserve Th17 cells during pathogenic SIV infection, providing support for Lactobacillus species as modulators of mucosal immune homeostasis.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Lactobacillus/immunology , Macaca mulatta/immunology , Simian Immunodeficiency Virus/immunology , Th17 Cells/immunology , Animals , Female , HIV Infections/immunology , HIV Infections/microbiology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/microbiology , Th17 Cells/microbiology
15.
Front Biosci ; 9: 3218-41, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15353351

ABSTRACT

We identified the ubiquitous glucose transporter GLUT1 as a receptor for Deltaretroviruses HTLV-1 and HTLV-2 envelopes (Env), mediating viral binding and entry. Here, we review the context and key observations that led us to this finding: functional modules of HTLV SU are similar to those of Gammaretrovirus Env which use multimembrane-spanning nutrient transporters as receptors; the HTLV Env receptor is an early marker of T lymphocyte activation; and HTLV Env inhibits glucose transport. We review several molecular, viral, cellular and physiological aspects of HTLV infection in relation to the in vivo and in vitro properties of GLUT1. Also, we examine the implications of HTLV-1 Env-GLUT1 interactions and altered glucose transport on the two major HTLV-1-induced diseases, adult T cell leukemia (ATL) and neurodegenerative tropical spastic paraparesis/HTLV-associated myelopathy (TSP/HAM). Complementary to the classical models of disease progression, we propose new schemes that emphasize the potential metabolic alterations caused in different cellular compartments. Finally, we review the potential use of HTLV Env-derived constructs as tools for labeling GLUT1 in vivo and inhibiting GLUT1 transport in tumor cells.


Subject(s)
Deltaretrovirus Infections/metabolism , Gene Products, env/physiology , Glucose/metabolism , Human T-lymphotropic virus 1/metabolism , Human T-lymphotropic virus 2/metabolism , Animals , Biological Transport , Gene Products, env/metabolism , Glucose Transporter Type 1/metabolism , Humans , Hydrogen-Ion Concentration , Leukemia, T-Cell/virology , Lymphocyte Activation , Paraparesis, Tropical Spastic/virology , Protein Binding
16.
Anat Rec (Hoboken) ; 297(8): 1392-406, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24867874

ABSTRACT

Establishment of a functional immune system has important implications for health and disease, yet questions remain regarding the mechanism, location, and timing of development of myeloid and lymphoid cell compartments. The goal of this study was to characterize the ontogeny of the myeloid-lymphoid system in rhesus monkeys to enhance current knowledge of the developmental sequence of B-cell (CD20, CD79), T-cell (CD3, CD4, CD8, FoxP3), dendritic cell (CD205), and macrophage (CD68) lineages in the fetus and infant. Immunohistochemical assessments addressed the temporal and spatial expression of select phenotypic markers in the developing liver, thymus, spleen, lymph nodes, gut-associated lymphoid tissue (GALT), and bone marrow with antibodies known to cross-react with rhesus cells. CD3 was the earliest lymphoid marker identified in the first trimester thymus and, to a lesser extent, in the spleen. T-cell markers were also expressed midgestation on cells of the liver, spleen, thymus, and in Peyer's patches of the small and large intestine, and where CCR5 expression was noted. A myeloid marker, CD68, was found on hepatic cells near blood islands in the late first trimester. B-cell markers were observed mid-second trimester in the liver, spleen, thymus, lymph nodes, bone marrow spaces, and occasionally in GALT. By the late third trimester and postnatally, secondary follicles with germinal centers were present in the thymus, spleen, and lymph nodes. These results suggest that immune ontogeny in monkeys is similar in temporal and anatomical sequence when compared to humans, providing important insights for translational studies.


Subject(s)
Cell Lineage , Lymphoid Tissue/embryology , Myeloid Cells/cytology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Biomarkers/analysis , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , Immunoenzyme Techniques , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Macaca mulatta , Macrophages/cytology , Macrophages/metabolism , Myeloid Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
17.
PLoS One ; 8(12): e84091, 2013.
Article in English | MEDLINE | ID: mdl-24391889

ABSTRACT

HIV-mediated immune dysfunction may influence CD4(+) T cell recovery during suppressive antiretroviral therapy (ART). We analyzed cellular biomarkers of immunological inflammation, maturation, and senescence in HIV-infected subjects on early suppressive ART. We performed longitudinal analyses of peripheral immunological biomarkers of subjects on suppressive ART (n = 24) from early treatment (median 6.4 months, interquartile range [IQR] 4.8-13.9 months) to 1-2 years of follow-up (median 19.8 months, IQR 18.3-24.6 months). We performed multivariate regression to determine which biomarkers were associated with and/or predictive of CD4(+) T cell recovery. After adjusting for the pre-ART CD4(+) T cell count, age, proximal CD4(+) T cell count, and length of ART medication, the percentage of CD27(+)CD8(+) T cells remained significantly associated with the CD4(+) T cell recovery rate (ß = 0.092 cells/ul/month, P = 0.028). In HIV-infected subjects starting suppressive ART, patients with the highest percentage of CD8(+) T cells expressing CD27 had the greatest rate of CD4(+) T cell recovery.


Subject(s)
Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Viral Load/immunology , Adult , Biomarkers/analysis , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Flow Cytometry , Follow-Up Studies , HIV Infections/drug therapy , HIV Infections/metabolism , HIV-1/drug effects , Humans , Longitudinal Studies , Middle Aged , RNA, Viral/genetics , Risk Factors , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
18.
Blood ; 109(3): 1034-42, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17023582

ABSTRACT

The IL-7 cytokine promotes the survival of a diverse T-cell pool, thereby ensuring an efficient immune response. Moreover, IL-7 induces the proliferation of recent thymic emigrants (RTEs) in neonates. Here, we demonstrate that the survival and proliferative effects of IL-7 on human RTEs can be distinguished on the basis of dose as well as duration of IL-7 administration. A dose of 0.1 ng/mL IL-7 is sufficient to promote viability, whereas cell-cycle entry is observed only at doses higher than 1 ng/mL. Moreover, a short 1-hour exposure to high-dose IL-7 (10 ng/mL) induces long-term survival but continuous IL-7 exposure is necessary for optimal cell-cycle entry and proliferation. We find that distinct signaling intermediates are activated under conditions of IL-7-induced survival and proliferation; STAT5 tyrosine phosphorylation does not correlate with proliferation, whereas up-regulation of the glucose transporter Glut-1 as well as increased glucose uptake are markers of IL-7-induced cell cycle entry. Glut-1 is directly regulated by PI3K and, indeed, inhibiting PI3K activity abrogates IL-7-induced proliferation. Our finding that the survival and proliferation of RTEs are differentially modulated by the dose and kinetics of exogenous IL-7 has important implications for the clinical use of this cytokine.


Subject(s)
Cell Movement , Cell Proliferation/drug effects , Interleukin-7/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Thymus Gland/cytology , Cell Cycle/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Glucose Transporter Type 1/analysis , Glucose Transporter Type 1/drug effects , Humans , Signal Transduction , T-Lymphocytes/cytology
19.
J Immunol ; 176(11): 6702-8, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16709829

ABSTRACT

IL-7 plays a major role in T lymphocyte homeostasis and has been proposed as an immune adjuvant for lymphopenic patients. This prospect is based, at least in part, on the short-term expansion of peripheral T cells in rIL7-treated mice and primates. Nevertheless, in vivo, following initial increases in T cell proliferation and numbers, lymphocytes return to a quiescent state. As the bases for this cell cycle exit have not yet been elucidated, it is important to assess the long-term biological effects of IL-7 on quiescent human T lymphocyte subsets. In this study, we find that IL-7-stimulated CD4+ naive lymphocytes enter into cell cycle with significantly delayed kinetics as compared with the memory population. Importantly though, these lymphocytes exit from the cell cycle despite the continuous replenishment of rIL-7. This response is distinct in memory and naive CD4+ lymphocytes with memory cells starting to exit from cycle by day 10 vs day 18 for naive cells. Return to quiescence is associated with a cessation in IL-7R signaling as demonstrated by an abrogation of STAT-5 phosphorylation, despite an up-regulation of surface IL-7Ralpha. Indeed, an initial 10-fold decrease in IL-7Ralpha mRNA levels is followed by increased IL-7Ralpha expression in naive as well as memory T cells, with kinetics paralleling cell cycle exit. Altogether, our data demonstrate that IL-7 promotes the extended survival of both naive and memory CD4+ T cells, whereas cycling of these two subsets is distinct and transient. Thus, IL-7 therapy should be designed to allow optimal responsiveness of naive and memory T cell subsets.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Cycle/immunology , Gene Expression Regulation/immunology , Interleukin-7/physiology , Lymphocyte Activation/immunology , Receptors, Interleukin-7/biosynthesis , Receptors, Interleukin-7/genetics , Adult , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Cycle/genetics , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Humans , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Protein Subunits/antagonists & inhibitors , Protein Subunits/biosynthesis , Protein Subunits/genetics , Receptors, Interleukin-7/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Time Factors
20.
Proc Natl Acad Sci U S A ; 102(36): 12867-72, 2005 Sep 06.
Article in English | MEDLINE | ID: mdl-16126902

ABSTRACT

GLUT1, the major glucose transporter in peripheral T lymphocytes, is induced upon T cell receptor activation. However, the role of GLUT1 during human thymocyte differentiation remains to be evaluated. Our identification of GLUT1 as the human T lymphotrophic virus (HTLV) receptor has enabled us to use tagged HTLV-receptor-binding domain fusion proteins to specifically monitor surface GLUT1 expression. Here, we identify a unique subset of CD4+ CD8+ double-positive (DP) thymocytes, based on their GLUT1 surface expression. Whereas these cells express variable levels of CD8, they express uniformly high levels of CD4. Glucose uptake was 7-fold higher in CD4(hi) DP thymocytes than in CD4(lo) DP thymocytes (P = 0.0002). Further analyses indicated that these GLUT1+ thymocytes are early post-beta-selection, as demonstrated by low levels of T cell receptor (TCR)alphabeta and CD3. This population of immature GLUT1+ DP cells is rapidly cycling and can be further distinguished by specific expression of the transferrin receptor. Importantly, the CXCR4 chemokine receptor is expressed at 15-fold higher levels on GLUT1+ DP thymocytes, as compared with the DP GLUT1- subset, and the former cells show enhanced chemotaxis to the CXCR4 ligand CXCL12. Thus, during human thymopoiesis, GLUT1 is up-regulated after beta-selection, and these immature DP cells constitute a population with distinct metabolic and chemotactic properties.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Cycle , Chemotaxis, Leukocyte , Monosaccharide Transport Proteins/metabolism , Receptors, CXCR4/metabolism , Thymus Gland/cytology , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/cytology , Cell Movement , Chemokine CXCL12 , Chemokines, CXC/metabolism , Chemotaxis, Leukocyte/drug effects , Child , Child, Preschool , Glucose/pharmacology , Glucose Transporter Type 1 , HIV-1/metabolism , Humans , Infant , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Transferrin/metabolism , Thymus Gland/drug effects , Thymus Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL