Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Immunometabolism ; 4(2)2022.
Article in English | MEDLINE | ID: mdl-35633761

ABSTRACT

People living with HIV (PLWH) who are immune non-responders (INR) to therapy are unable to restore their CD4 T-cell count and remain at great risk of morbidity and mortality. Here the mitochondrial defects that characterize memory CD4 T-cells in INR and causes of this mitochondrial exhaustion are reviewed. This review also describes the various reagents used to induce the expression of the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), the master regulator of mitochondrial biogenesis, which can restore mitochondria fitness and CD4 T-cell proliferation in INR. Due to sustained heightened inflammation in INR, the mitochondrial network is unable to be rejuvenated and requires attenuation of mediators of inflammation to rescue mitochondria and CD4 T-cell counts in INR.

2.
J Clin Invest ; 132(9)2022 05 02.
Article in English | MEDLINE | ID: mdl-35316209

ABSTRACT

People living with HIV (PLWH) who are immune nonresponders (INRs) are at greater risk of comorbidity and mortality than are immune responders (IRs) who restore their CD4+ T cell count after antiretroviral therapy (ART). INRs have low CD4+ T cell counts (<350 c/µL), heightened systemic inflammation, and increased CD4+ T cell cycling (Ki67+). Here, we report the findings that memory CD4+ T cells and plasma samples of INRs from several cohorts are enriched in gut-derived bacterial solutes p-cresol sulfate (PCS) and indoxyl sulfate (IS) that both negatively correlated with CD4+ T cell counts. In vitro PCS or IS blocked CD4+ T cell proliferation, induced apoptosis, and diminished the expression of mitochondrial proteins. Electron microscopy imaging revealed perturbations of mitochondrial networks similar to those found in INRs following incubation of healthy memory CD4+ T cells with PCS. Using bacterial 16S rDNA, INR stool samples were found enriched in proteolytic bacterial genera that metabolize tyrosine and phenylalanine to produce PCS. We propose that toxic solutes from the gut bacterial flora may impair CD4+ T cell recovery during ART and may contribute to CD4+ T cell lymphopenia characteristic of INRs.


Subject(s)
Bacterial Toxins , HIV Infections , HIV-1 , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes , Humans , Lymphopenia , Mitochondria
3.
Front Immunol ; 12: 638010, 2021.
Article in English | MEDLINE | ID: mdl-33868264

ABSTRACT

Background: Immune non-responders (INR) are HIV+, ART-controlled (>2 yrs) people who fail to reconstitute their CD4 T cell numbers. Systemic inflammation and markers of T cell senescence and exhaustion are observed in INR. This study aims to investigate T cell senescence and exhaustion and their possible association with soluble immune mediators and to understand the immune profile of HIV-infected INR. Selected participants were <50 years old to control for the confounder of older age. Methods: Plasma levels of IL-6, IP10, sCD14, sCD163, and TGF-ß and markers of T cell exhaustion (PD-1, TIGIT) and senescence (CD57, KLRG-1) were measured in ART-treated, HIV+ participants grouped by CD4 T cell counts (n = 63). Immune parameters were also measured in HIV-uninfected, age distribution-matched controls (HC; n = 30). Associations between T cell markers of exhaustion and senescence and plasma levels of immune mediators were examined by Spearman rank order statistics. Results: Proportions of CD4 T cell subsets expressing markers of exhaustion (PD-1, TIGIT) and senescence (CD57, KLRG-1) were elevated in HIV+ participants. When comparing proportions between INR and IR, INR had higher proportions of CD4 memory PD-1+, EM CD57+, TEM TIGIT+ and CD8 EM and TEM TIGIT+ cells. Plasma levels of IL-6, IP10, and sCD14 were elevated during HIV infection. IP10 was higher in INR. Plasma TGF-ß levels and CD4 cycling proportions of T regulatory cells were lower in INR. Proportions of CD4 T cells expressing TIGIT, PD-1, and CD57 positively correlated with plasma levels of IL-6. Plasma levels of TGF-ß negatively correlated with proportions of TIGIT+ and PD-1+ T cell subsets. Conclusions: INR have lower levels of TGF-ß and decreased proportions of cycling CD4 T regulatory cells and may have difficulty controlling inflammation. IP10 is elevated in INR and is linked to higher proportions of T cell exhaustion and senescence seen in INR.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cellular Senescence/immunology , HIV Infections/immunology , T-Lymphocyte Subsets/immunology , Transforming Growth Factor beta/blood , Adult , Anti-Retroviral Agents/therapeutic use , Antigens, CD/blood , Antigens, Differentiation, Myelomonocytic/blood , Biomarkers/blood , CD4 Lymphocyte Count , CD57 Antigens/blood , Female , Humans , Interleukin-6/blood , Lectins, C-Type/blood , Lipopolysaccharide Receptors/blood , Lymphocyte Activation/immunology , Male , Middle Aged , Programmed Cell Death 1 Receptor/blood , Receptors, Cell Surface/blood , Receptors, Cytokine/blood , Receptors, Immunologic/blood , Young Adult
4.
PLoS Pathog ; 16(9): e1008885, 2020 09.
Article in English | MEDLINE | ID: mdl-32976527

ABSTRACT

Atherosclerotic cardiovascular disease (ASCVD) remains an important cause of morbidity in the general population and risk for ASCVD is increased approximately 2-fold in persons living with HIV infection (PLWH). This risk is linked to elevated CD8 T cell counts that are abundant in atherosclerotic plaques and have been implicated in disease pathogenesis yet the mechanisms driving T cell recruitment to and activation within plaques are poorly defined. Here we investigated the role of CD8 T cells in atherosclerosis in a non-human primate model of HIV infection and in the HIV-uninfected elderly; we sought to identify factors that promote the activation, function, and recruitment to endothelium of CX3CR1+ CD8 T cells. We measured elevated expression of CX3CL1 and IL-15, and increased CD8 T cell numbers in the aortas of rhesus macaques infected with SIV or SHIV, and demonstrated similar findings in atherosclerotic vessels of HIV-uninfected humans. We found that recombinant TNF enhanced the production and release of CX3CL1 and bioactive IL-15 from aortic endothelial cells, but not from aortic smooth muscle cells. IL-15 in turn promoted CX3CR1 surface expression on and TNF synthesis by CD8 T cells, and IL-15-treated CD8 T cells exhibited enhanced CX3CL1-dependent chemoattraction toward endothelial cells in vitro. Finally, we show that CD8 T cells in human atherosclerotic plaques have an activated, resident phenotype consistent with in vivo IL-15 and CX3CL1 exposure. In this report, we define a novel model of CD8 T cell involvement in atherosclerosis whereby CX3CL1 and IL-15 operate in tandem within the vascular endothelium to promote infiltration by activated CX3CR1+ memory CD8 T cells that drive further endothelial activation via TNF. We propose that these interactions are prevalent in aging and in PLWH, populations where circulating activated CX3CR1+ CD8 T cell numbers are often expanded.


Subject(s)
Atherosclerosis/metabolism , CD8-Positive T-Lymphocytes/metabolism , Chemokine CX3CL1/metabolism , HIV Infections/metabolism , Interleukin-15/metabolism , Aged , Animals , Endothelial Cells/metabolism , Humans , Macaca mulatta/metabolism , Receptors, Chemokine/metabolism
5.
JCI Insight ; 5(11)2020 06 04.
Article in English | MEDLINE | ID: mdl-32369455

ABSTRACT

HIV infection is associated with an increase in the proportion of activated CD8+ memory T cells (Tmem) that express CX3CR1, but how these cells are generated and maintained in vivo is unclear. We demonstrate that increased CX3CR1 expression on CD8+ Tmem in people living with HIV (PLWH) is dependent on coinfection with human CMV, and CX3CR1+CD8+ Tmem are enriched for a putatively immunosenescent CD57+CD28- phenotype. The cytokine IL-15 promotes the phenotype, survival, and proliferation of CX3CR1+CD57+CD8+ Tmem in vitro, whereas T cell receptor stimulation leads to their death. IL-15-driven survival is dependent on STAT5 and Bcl-2 activity, and IL-15-induced proliferation requires STAT5 and mTORC1. Thus, we identify mechanistic pathways that could explain how "inflammescent" CX3CR1+CD57+ CD8+ Tmem dominate the overall memory T cell pool in CMV-seropositive PLWH and that support reevaluation of immune senescence as a nonproliferative dead end.


Subject(s)
CD57 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CX3C Chemokine Receptor 1/immunology , HIV Infections/immunology , HIV-1/immunology , Immunologic Memory , Interleukin-15/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Cell Survival/immunology , HIV Infections/pathology , Humans
6.
J Immunol ; 204(10): 2722-2733, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32229536

ABSTRACT

Cytotoxic CD4 T cells are linked to cardiovascular morbidities and accumulate in both HIV and CMV infections, both of which are associated with increased risk of cardiovascular disease (CVD). In this study, we identify CMV coinfection as a major driver of the cytotoxic phenotype, characterized by elevated CD57 expression and reduced CD28 expression, in circulating CD4 T cells from people living with HIV infection, and investigate potential mechanisms linking this cell population to CVD. We find that human CD57+ CD4 T cells express high levels of the costimulatory receptor CD2 and that CD2/LFA-3 costimulation results in a more robust and polyfunctional effector response to TCR signals, compared with CD28-mediated costimulation. CD57+ CD4 T cells also express the vascular endothelium-homing receptor CX3CR1 and migrate toward CX3CL1-expressing endothelial cells in vitro. IL-15 promotes the cytotoxic phenotype, elevates CX3CR1 expression, and enhances the trafficking of CD57+ CD4 T cells to endothelium and may therefore be important in linking these cells to cardiovascular complications. Finally, we demonstrate the presence of activated CD57+ CD4 T cells and expression of CX3CL1 and LFA-3 in atherosclerotic plaque tissues from HIV-uninfected donors. Our findings are consistent with a model in which cytotoxic CD4 T cells contribute to CVD in HIV/CMV coinfection and in atherosclerosis via CX3CR1-mediated trafficking and CD2/LFA-3-mediated costimulation. This study identifies several targets for therapeutic interventions and may help bridge the gap in understanding how CMV infection and immunity are linked to increased cardiovascular risk in people living with HIV infection.


Subject(s)
Blood Vessels/physiology , CD4-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/physiology , HIV Infections/immunology , HIV-1/physiology , Plaque, Atherosclerotic/immunology , CD28 Antigens/metabolism , CD57 Antigens/metabolism , CD58 Antigens/metabolism , CX3C Chemokine Receptor 1/metabolism , Cell Movement , Chemokine CX3CL1/metabolism , Coinfection , Cytotoxicity, Immunologic , Humans , Receptors, CXCR3/metabolism , Risk
7.
J Clin Invest ; 128(11): 5083-5094, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30320604

ABSTRACT

Immune nonresponder (INR) HIV-1-infected subjects are characterized by their inability to reconstitute the CD4+ T cell pool after antiretroviral therapy. This is linked to poor clinical outcome. Mechanisms underlying immune reconstitution failure are poorly understood, although, counterintuitively, INRs often have increased frequencies of circulating CD4+ T cells in the cell cycle. While cycling CD4+ T cells from healthy controls and HIV+ patients with restored CD4+ T cell numbers complete cell division in vitro, cycling CD4+ T cells from INRs do not. Here, we show that cells with the phenotype and transcriptional profile of Tregs were enriched among cycling cells in health and in HIV infection. Yet there were diminished frequencies and numbers of Tregs among cycling CD4+ T cells in INRs, and cycling CD4+ T cells from INR subjects displayed transcriptional profiles associated with the impaired development and maintenance of functional Tregs. Flow cytometric assessment of TGF-ß activity confirmed the dysfunction of Tregs in INR subjects. Transcriptional profiling and flow cytometry revealed diminished mitochondrial fitness in Tregs among INRs, and cycling Tregs from INRs had low expression of the mitochondrial biogenesis regulators peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1α) and transcription factor A for mitochondria (TFAM). In vitro exposure to IL-15 allowed cells to complete division, restored the expression of PGC1α and TFAM, and regenerated mitochondrial fitness in the cycling Tregs of INRs. Our data suggest that rescuing mitochondrial function could correct the immune dysfunction characteristic of Tregs in HIV-1-infected subjects who fail to restore CD4+ T cells during antiretroviral therapy.


Subject(s)
DNA-Binding Proteins/immunology , HIV Infections/immunology , HIV-1 , Interleukin-15/immunology , Mitochondria/immunology , Mitochondrial Proteins/immunology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/immunology , T-Lymphocytes, Regulatory/immunology , Transcription Factors/immunology , Adult , Anti-Retroviral Agents/administration & dosage , CD4 Lymphocyte Count , Female , HIV Infections/drug therapy , HIV Infections/pathology , Humans , Male , Middle Aged , Mitochondria/pathology , T-Lymphocytes, Regulatory/pathology
8.
J Infect Dis ; 214(12): 1808-1816, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27703039

ABSTRACT

Increases in inflammation, coagulation, and CD8+ T-cell numbers are associated with an elevated cardiovascular disease (CVD) risk in human immunodeficiency virus (HIV)-infected antiretroviral therapy (ART) recipients. Circulating memory CD8+ T cells that express the vascular endothelium-homing receptor CX3CR1 (fractalkine receptor) are enriched in HIV-infected ART recipients. Thrombin-activated receptor (PAR-1) expression is increased in HIV-infected ART recipients and is particularly elevated on CX3CR1+ CD8+ T cells, suggesting that these cells could interact with coagulation elements. Indeed, thrombin directly enhanced T-cell receptor-mediated interferon γ production by purified CD8+ T cells but was attenuated by thrombin-induced release of transforming growth factor ß by platelets. We have therefore identified a population of circulating memory CD8+ T cells in HIV infection that may home to endothelium, can be activated by clot-forming elements, and are susceptible to platelet-mediated regulation. Complex interactions between inflammatory elements and coagulation at endothelial surfaces may play an important role in CVD risk in HIV-infected ART recipients.


Subject(s)
Blood Platelets/metabolism , CD8-Positive T-Lymphocytes/immunology , HIV Infections/pathology , Receptors, Chemokine/analysis , T-Lymphocyte Subsets/immunology , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/drug effects , CX3C Chemokine Receptor 1 , HIV Infections/immunology , Humans , T-Lymphocyte Subsets/chemistry , T-Lymphocyte Subsets/drug effects , Transforming Growth Factor beta/metabolism
9.
J Infect Dis ; 214 Suppl 2: S51-7, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27625431

ABSTRACT

Untreated human immunodeficiency virus (HIV) infection is characterized by progressive CD4(+) T-cell depletion and CD8(+) T-cell expansion, and CD4(+) T-cell depletion is linked directly to the risk for opportunistic infections and infection-associated mortality. With suppression of HIV replication by antiretroviral therapy, circulating CD4(+) Tcell numbers typically improve while CD8(+) T-cell expansion persists, and both CD4(+) T-cell cytopenia and CD8(+) T-cell expansion are associated with morbidity and mortality. In this brief review, we report on the role that selected homeostatic and inflammatory cytokines may play both in the failure of CD4(+) T-cell restoration and the CD8(+) T-cell expansion that characterize HIV infection.


Subject(s)
Cytokines/immunology , HIV Infections/immunology , T-Lymphocytes/immunology , Animals , Homeostasis , Humans
10.
Front Immunol ; 7: 228, 2016.
Article in English | MEDLINE | ID: mdl-27379092

ABSTRACT

Residual mucosal inflammation along with chronic systemic immune activation is an important feature in individuals infected with human immunodeficiency virus (HIV), and has been linked to a wide range of co-morbidities, including malignancy, opportunistic infections, immunopathology, and cardiovascular complications. Although combined antiretroviral therapy (cART) can reduce plasma viral loads to undetectable levels, reservoirs of virus persist, and increased mortality is associated with immune dysbiosis in mucosal lymphoid tissues. Immune-based therapies are pursued with the goal of improving CD4(+) T-cell restoration, as well as reducing chronic immune activation in cART-treated patients. However, the majority of research on immune activation has been derived from analysis of circulating T cells. How immune cell alterations in mucosal tissues contribute to HIV immune dysregulation and the associated risk of non-infectious chronic complications is less studied. Given the significant differences between mucosal T cells and circulating T cells, and the immediate interactions of mucosal T cells with the microbiome, more attention should be devoted to mucosal immune cells and their contribution to systemic immune activation in HIV-infected individuals. Here, we will focus on mucosal immune cells with a specific emphasis on CD4(+) T lymphocytes, such as T helper 17 cells and CD4(+)Foxp3(+) regulatory T cells (Tregs), which play crucial roles in maintaining mucosal barrier integrity and preventing inflammation, respectively. We hypothesize that pro-inflammatory milieu in cART-treated patients with immune activation significantly contributes to enhanced loss of Th17 cells and increased frequency of dysregulated Tregs in the mucosa, which in turn may exacerbate immune dysfunction in HIV-infected patients. We also present initial evidence to support this hypothesis. A better comprehension of how pro-inflammatory milieu impacts these two types of cells in the mucosa will shed light on mucosal immune dysfunction and HIV reservoirs, and lead to novel ways to restore immune functions in HIV(+) patients.

11.
J Clin Invest ; 126(7): 2745-56, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27322062

ABSTRACT

In HIV-1-infected patients, increased numbers of circulating CD8+ T cells are linked to increased risk of morbidity and mortality. Here, we identified a bystander mechanism that promotes CD8 T cell activation and expansion in untreated HIV-1-infected patients. Compared with healthy controls, untreated HIV-1-infected patients have an increased population of proliferating, granzyme B+, CD8+ T cells in circulation. Vß expression and deep sequencing of CDR3 revealed that in untreated HIV-1 infection, cycling memory CD8 T cells possess a broad T cell repertoire that reflects the repertoire of the resting population. This suggests that cycling is driven by bystander activation, rather than specific antigen exposure. Treatment of peripheral blood mononuclear cells with IL-15 induced a cycling, granzyme B+ phenotype in CD8+ T cells. Moreover, elevated IL-15 expression in the lymph nodes of untreated HIV-1-infected patients correlated with circulating CD8+ T cell counts and was normalized in these patients following antiretroviral therapy. Together, these results suggest that IL-15 drives bystander activation of CD8+ T cells, which predicts disease progression in untreated HIV-1-infected patients and suggests that elevated IL-15 may also drive CD8+ T cell expansion that is linked to increased morbidity and mortality in treated patients.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , HIV Infections/blood , Interleukin-15/metabolism , Lymphocyte Activation , Adult , Aged , Animals , Anti-Retroviral Agents/therapeutic use , Biopsy , Case-Control Studies , Cell Proliferation , Female , Granzymes/metabolism , HIV-1 , Haplotypes , Humans , Ki-67 Antigen/metabolism , Leukocyte Common Antigens/metabolism , Leukocytes, Mononuclear/cytology , Lymph Nodes/pathology , Male , Mice , Middle Aged , Phenotype , Receptors, CCR7/metabolism
13.
Clin Infect Dis ; 62(3): 392-6, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26400999

ABSTRACT

BACKGROUND: Persistent CD8 T-cell expansion, low CD4/CD8 T-cell ratios, and heightened inflammation persist in antiretroviral therapy (ART)-treated human immunodeficiency virus (HIV) infection and are associated with increased risk of morbid outcomes. We explored the role of cytomegalovirus (CMV) infection in CD8 lymphocytosis and inflammation in ART-treated HIV infection. METHODS: Absolute CD4 and CD8 T-cell counts were abstracted from clinical records and compared among 32 HIV-infected CMV-seronegative subjects, 126 age, CD4 and gender-matched HIV-infected CMV-seropositive subjects, and among 21 HIV-uninfected controls (9 CMV-negative, 12 CMV-positive). Plasma inflammatory indices were measured in a subset by ELISA. RESULTS: Median CD8 counts/µL were higher in HIV-positive/CMV-positive patients (795) than in HIV-positive/CMV-negative subjects (522, P = .006) or in healthy controls (451, P = .0007), whereas CD8 T-cell counts were similar to controls' levels in HIV-positive/CMV-negative subjects. Higher plasma levels of IP-10 (P = .0011), TNF-RII (P = .0002), and D-dimer (P = .0444) were also found in coinfected patients than in HIV-positive/CMV-negative subjects. CONCLUSIONS: CMV infection is associated with higher CD8 T-cell counts, resultant lower CD4/CD8 ratios, and increased systemic inflammation in ART-treated HIV infection. CMV infection may contribute to risk for morbid outcomes in treated HIV infection.


Subject(s)
Anti-Retroviral Agents/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Coinfection/pathology , Cytomegalovirus Infections/pathology , HIV Infections/complications , HIV Infections/drug therapy , Adult , CD4-CD8 Ratio , CD4-Positive T-Lymphocytes/immunology , Cohort Studies , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Female , HIV Infections/pathology , Humans , Lymphocyte Count , Lymphocytosis/pathology , Male , Middle Aged , Plasma/chemistry
14.
J Acquir Immune Defic Syndr ; 71(5): 483-92, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26627102

ABSTRACT

BACKGROUND: HIV-infected patients who fail to normalize CD4 T cells despite suppressive antiretroviral therapy have impaired immune homeostasis: diminished naive T-cell numbers, elevated T-cell turnover, senescence, and inflammation. METHODS: Blood samples from immune failures (n = 60), immune successes (n = 20), and healthy controls (n = 20) were examined for plasma interleukin (IL)-7 levels, for cellular expression of the IL-7Rα chain (CD127), for the exhaustion and senescence markers programed death 1 (PD-1) and CD57, and for the survival factor Bcl2. Because both inflammatory and homeostatic cytokines can induce T-cell cycling, we also examined the effects of these mediators on exhaustion and senescence markers. RESULTS: Plasma levels of IL-7 were elevated and both CD4 and CD8 T-cell CD127 expression was decreased in immune failure. Plasma levels of IL-7 correlated directly with naive CD4 T-cell counts in immune success and inversely with T-cell cycling (Ki67) in healthy controls and immune success, but not in immune failure. CD4 T-cell density of PD-1 was increased and Bcl2+ CD4 T cells were decreased in immune failure but not in immune success, whereas the proportion of T cells expressing CD57 was increased in immune failure. PD-1 and CD57 were induced on CD4 but not CD8 T cells by stimulation in vitro with inflammatory IL-1ß or homeostatic (IL-7) cytokines. CONCLUSIONS: Perturbation of the IL-7/IL-7 receptor axis, increased T-cell turnover, and increased senescence may reflect dysregulated responses to both homeostatic and inflammatory cytokines in immune failure patients.


Subject(s)
HIV Infections/immunology , Interleukin-7/blood , Adult , Anti-HIV Agents , Biomarkers/metabolism , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD57 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Case-Control Studies , Cell Death/physiology , Cellular Senescence/immunology , Female , Humans , Immunophenotyping , Interleukin-7 Receptor alpha Subunit/blood , Lymphocyte Activation/immunology , Male , Treatment Failure , bcl-Associated Death Protein/blood
15.
J Infect Dis ; 213(9): 1419-27, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26671887

ABSTRACT

BACKGROUND: Human immunodeficiency virus (HIV) infection is associated with increased risk of thromboembolic and cardiovascular comorbid conditions. Although systemic inflammation is linked to cardiovascular risk, direct evidence of vascular inflammation and endothelial dysfunction is lacking. METHODS: We examined by immunofluorescence microscopy thoracic aortas from 16 simian immunodeficiency virus (SIV)- or simian-human immunodeficiency virus (SHIV)-infected and 16 uninfected rhesus macaques. RESULTS: Focal endothelial proliferation and subendothelial inflammatory cells were found in sections of all infected animals, compared with minimal changes in sections from the 16 uninfected controls. In the infected animals, we detected increased endothelial levels of bacterial 16s ribosomal DNA as well as increased subendothelial accumulation of CD68(+) monocytes/macrophages (P< .001) and CD8(+) T lymphocytes (P< .001). Endothelial dysfunction was manifested by decreased levels of endothelial nitric oxide synthase (P< .005) and Krüppel-like factor 2 (KLF2) (P< .005). KLF2 expression was decreased in primary human aortic endothelial cells exposed to bacterial lipopolysaccharide or to oxidized low-density lipoprotein in vitro, and this could be prevented by simvastatin. CONCLUSIONS: SIV and SHIV infection lead to endothelial inflammation, dysfunction, and decreased KLF2 expression reflecting early atherosclerotic changes. Translocated bacterial components and lipid oxidation products may induce endothelial dysfunction in HIV infection that could be prevented by statin treatment.


Subject(s)
Endothelium, Vascular/immunology , HIV Infections/immunology , HIV-1/immunology , Kruppel-Like Transcription Factors/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Vasculitis/immunology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/immunology , Aorta, Thoracic/microbiology , Cells, Cultured , DNA, Bacterial/analysis , Endothelium, Vascular/microbiology , HIV Infections/physiopathology , Humans , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/physiopathology
16.
J Virol ; 88(10): 5369-80, 2014 May.
Article in English | MEDLINE | ID: mdl-24522925

ABSTRACT

UNLABELLED: The mechanisms of increased memory CD4+ T cell cycling in HIV disease are incompletely understood but have been linked to antigen stimulation, homeostatic signals, or exposure to microbial products and the inflammatory cytokines that they induce. We examined the phenotype and Vß family distribution in cycling memory CD4+ T cells among 52 healthy and 59 HIV-positive (HIV+) donors. Cycling memory CD4+ T cells were proportionally more frequent in subjects with HIV infection than in controls, more often expressed CD38 and PD-1, and less frequently expressed OX40 and intracellular CD40L. OX40 expression on memory CD4+ T cells was induced in vitro by anti-CD3, interleukin-2 (IL-2), IL-7, or IL-15 but not by Toll-like receptor ligands. In HIV+ donors, memory CD4+ T cell cycling was directly related to plasma lipopolysaccharide (LPS) levels, to plasma HIV RNA levels, and to memory CD8+ T cell cycling and was inversely related to peripheral blood CD4+ T cell counts but not to the levels of IL-2, IL-7, or IL-15, while in HIV-negative donors, memory CD4+ T cell cycling was related to IL-7 levels and negatively related to the plasma levels of LPS. In both controls and HIV+ donors, cycling memory CD4+ T cells had a broad distribution of Vß families comparable to that of noncycling cells. Increased memory CD4+ T cell cycling in HIV disease is reflective of generalized immune activation and not driven primarily by cognate peptide stimulation or exposure to common gamma-chain cytokines. This cycling may be a consequence of exposure to microbial products, to plasma viremia, or, otherwise, to proinflammatory cytokines. IMPORTANCE: This work provides evidence that the increased memory CD4+ T cell cycling in HIV infection is not a result of cognate peptide recognition but, rather, is more likely related to the inflammatory environment of HIV infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , Immunologic Memory , Lymphocyte Activation , Receptors, Antigen, T-Cell/analysis , T-Lymphocyte Subsets/immunology , Adult , CD4-Positive T-Lymphocytes/chemistry , Female , Humans , Immunophenotyping , Male , Middle Aged , T-Lymphocyte Subsets/chemistry
17.
J Immunol ; 192(4): 1896-906, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24453254

ABSTRACT

Mouse mammary tumor virus superantigens (vSAGs) are notorious for defying structural characterization, and a consensus has yet to be reached regarding their ability to bridge the TCR to MHC class II (MHCII). In this study, we determined the topology of the T cell signaling complex by examining the respective relation of vSAG7 with the MHCII molecule, MHCII-associated peptide, and TCR. We used covalently linked peptide/MHCII complexes to demonstrate that vSAG presentation is tolerant to variation in the protruding side chains of the peptide, but can be sensitive to the nature of the protruding N-terminal extension. An original approach in which vSAG was covalently linked to either MHCII chain confirmed that vSAG binds outside the peptide binding groove. Also, whereas the C-terminal vSAG segment binds to the MHCII α-chain in a conformation-sensitive manner, the membrane-proximal N-terminal domain binds the ß-chain. Because both moieties of the mature vSAG remain noncovalently associated after processing, our results suggest that vSAG crosslinks MHCII molecules. Comparing different T cell hybridomas, we identified key residues on the MHCII α-chain that are differentially recognized by the CDR3ß when engaged by vSAG. Finally, we show that the highly conserved tyrosine residue found in the vSAg TGXY motif is required for T cell activation. Our results reveal a novel SAG/MHCII/TCR architecture in which vSAGs coerce a near-canonical docking between MHCII and TCR that allows eschewing of traditional CDR3 binding with the associated peptide in favor of MHCII α-chain binding. Our findings highlight the plasticity of the TCR CDRs.


Subject(s)
Antigens, Viral/immunology , Histocompatibility Antigens Class II/immunology , Mammary Tumor Virus, Mouse/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Superantigens/immunology , Binding Sites/immunology , HEK293 Cells , HeLa Cells , Humans , Jurkat Cells , Lymphocyte Activation/immunology , Protein Binding/immunology , Protein Structure, Tertiary , Signal Transduction/immunology , T-Lymphocytes/immunology
18.
PLoS Biol ; 9(10): e1001171, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22022231

ABSTRACT

Memory phenotype (CD44(bright), CD25(negative)) CD4 spleen and lymph node T cells (MP cells) proliferate rapidly in normal or germ-free donors, with BrdU uptake rates of 6% to 10% per day and Ki-67 positivity of 18% to 35%. The rapid proliferation of MP cells stands in contrast to the much slower proliferation of lymphocytic choriomeningitis virus (LCMV)-specific memory cells that divide at rates ranging from <1% to 2% per day over the period from 15 to 60 days after LCMV infection. Anti-MHC class II antibodies fail to inhibit the in situ proliferation of MP cells, implying a non-T-cell receptor (TCR)-driven proliferation. Such proliferation is partially inhibited by anti-IL-7Rα antibody. The sequence diversity of TCRß CDR3 gene segments is comparable among the proliferating and quiescent MP cells from conventional and germ-free mice, implying that the majority of proliferating MP cells have not recently derived from a small cohort of cells that expand through multiple continuous rounds of cell division. We propose that MP cells constitute a diverse cell population, containing a subpopulation of slowly dividing authentic antigen-primed memory cells and a majority population of rapidly proliferating cells that did not arise from naïve cells through conventional antigen-driven clonal expansion.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Cytokines/immunology , Immunologic Memory/immunology , Animals , Bromodeoxyuridine/metabolism , Germ-Free Life , Histocompatibility Antigens Class II/immunology , Ki-67 Antigen/biosynthesis , Lymphocytic choriomeningitis virus/immunology , Mice , Receptors, Antigen, T-Cell/immunology , Receptors, Interleukin-7/immunology
19.
Eur J Immunol ; 37(7): 1916-23, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17549737

ABSTRACT

CD25(+) regulatory T cells (Treg) are a heterogeneous population that exists as CD44(low) and CD44(high) cells. Here we report that while both CD44(low) and CD44(high) Treg are anergic and express similar levels of Foxp3, CD44(high) Treg are highly proliferative in vivo and are more potent suppressors in vitro than CD44(low) Treg. From analysis of the properties of Treg derived from germ-free mice, it was concluded that peptide antigens derived from intestinal microorganisms are not essential for the generation, in vivo proliferation or suppressive activity of Treg. Our results suggest that gut flora antigens play little or no role in the heterogeneity and homeostatic regulation of Treg.


Subject(s)
Antigens, Bacterial/immunology , Homeostasis/immunology , Immunity, Mucosal , Intestinal Mucosa/microbiology , T-Lymphocytes, Regulatory/immunology , Animals , Flow Cytometry , Hyaluronan Receptors/immunology , Intestinal Mucosa/immunology , Mice
20.
J Immunol ; 178(2): 788-97, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17202340

ABSTRACT

The impact of exposure to Ag on the development and maintenance of human CD4(+) memory T cells in general and HIV infection in particular is partially understood. In this study, we measured HIV-specific CD4(+) T cell proliferative responses against HIV proteins and derived peptides one year after highly active antiretroviral therapy initiation in 39 HIV-infected patients who initiated therapy at different times following infection. We show that a brief exposure to HIV of <1 month does not allow the generation of significant detectable frequencies of HIV-specific CD4(+) memory T cells. Patients having prolonged cumulative exposure to high viral load due to therapy failures also demonstrated limited HIV-specific CD4(+) T cell responses. In contrast, patients exposed to significant levels of virus for periods ranging from 3 to 18 mo showed brisk and broad HIV-specific CD4(+) T cell responses 1 year following the onset of therapy intervention. We also demonstrate that the nadir CD4(+) T cell count before therapy initiation correlated positively with the breadth and magnitude of these responses. Our findings indicate that the loss of proliferative HIV-specific CD4(+) T cell responses is associated with the systemic progression of the disease and that a brief exposure to HIV does not allow the establishment of detectable frequencies of HIV-specific memory CD4(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , HIV Infections/immunology , HIV Infections/virology , Immunologic Memory/immunology , Amino Acid Sequence , Antiretroviral Therapy, Highly Active , Cells, Cultured , Conserved Sequence , Gene Products, gag/chemistry , Gene Products, nef/chemistry , Gene Products, tat/chemistry , HIV Infections/drug therapy , HIV Infections/pathology , Humans , Lymphocyte Count , Molecular Sequence Data , Substrate Specificity , Time Factors , nef Gene Products, Human Immunodeficiency Virus , tat Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...