Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(13)2023 Jul 03.
Article in English | MEDLINE | ID: mdl-37446195

ABSTRACT

Like other chronic viral infections, HIV-1 persistence inhibits the development of antigen-specific memory T-cells, resulting in the exhaustion of the immune response and chronic inflammation. Autophagy is a major lysosome-dependent mechanism of intracellular large-target degradation such as lipid and protein aggregates, damaged organelles, and intracellular pathogens. Although it is known that autophagy may target HIV-1 for elimination, knowledge of its function as a metabolic contributor in such viral infection is only in its infancy. Recent data show that elite controllers (EC), who are HIV-1-infected subjects with natural and long-term antigen (Ag)-specific T-cell protection against the virus, are characterized by distinct metabolic autophagy-dependent features in their T-cells compared to other people living with HIV-1 (PLWH). Despite durable viral control with antiretroviral therapy (ART), HIV-1-specific immune dysfunction does not normalize in non-controller PLWH. Therefore, the hypothesis of inducing autophagy to strengthen their Ag-specific T-cell immunity against HIV-1 starts to be an enticing concept. The aim of this review is to critically analyze promises and potential limitations of pharmacological and dietary interventions to activate autophagy in an attempt to rescue Ag-specific T-cell protection among PLWH.


Subject(s)
HIV Infections , HIV-1 , Humans , HIV-1/physiology , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Inflammation/metabolism
2.
JCI Insight ; 8(13)2023 07 10.
Article in English | MEDLINE | ID: mdl-37227774

ABSTRACT

HIV-1 infection is characterized by inflammation and a progressive decline in CD4+ T cell count. Despite treatment with antiretroviral therapy (ART), the majority of people living with HIV (PLWH) maintain residual levels of inflammation, a low degree of immune activation, and higher sensitivity to cell death in their memory CD4+ T cell compartment. To date, the mechanisms responsible for this high sensitivity remain elusive. We have identified the transcription factor IRF-5 to be involved in impairing the maintenance of murine CD4+ T cells during chronic infection. Here, we investigate whether IRF-5 also contributes to memory CD4+ T cell loss during HIV-1 infection. We show that TLR7 and IRF-5 were upregulated in memory CD4+ T cells from PLWH, when compared with naturally protected elite controllers and HIVfree participants. TLR7 was upstream of IRF-5, promoting Caspase 8 expression in CD4+ T cells from ART HIV-1+ but not from HIVfree donors. Interestingly, the TLR7/IRF-5 axis acted synergistically with the Fas/FasL pathway, suggesting that TLR7 and IRF-5 expression in ART HIV-1+ memory CD4+ T cells represents an imprint that predisposes cells to Fas-mediated apoptosis. This predisposition could be blocked using IRF-5 inhibitory peptides, suggesting IRF-5 blockade as a possible therapy to prevent memory CD4+ T cell loss in PLWH.


Subject(s)
HIV Infections , HIV-1 , Humans , Mice , Animals , CD4-Positive T-Lymphocytes , Toll-Like Receptor 7/metabolism , Apoptosis , Inflammation/metabolism
3.
Autophagy ; 18(6): 1256-1273, 2022 06.
Article in English | MEDLINE | ID: mdl-34612140

ABSTRACT

The maintenance of a strong IL21 production in memory CD4 T cells, especially in HIV-1-specific cells, represents a major correlate of natural immune protection against the virus. However, the molecular mechanisms underlying IL21 production during HIV-1 infection, which is only elevated among the naturally protected elite controllers (EC), are still unknown. We recently found out that lipophagy is a critical immune mediator that control an antiviral metabolic state following CD8A T cell receptor engagement, playing an important role in the natural control of HIV-1 infection. This led us to investigate whether the beneficial role of a strong macroautophagy/autophagy, could also be used to ensure effective IL21 production as well. Herein, we confirm that after both polyclonal and HIV-1-specific activation, memory CD4 T cells (Mem) from EC display enhanced activity of the autophagy-mediated proteolysis compared to ART. Our results indicate that the enhanced autophagy activity in EC was controlled by the energy-sensing PRKAA1 (protein kinase AMP-activated catalytic subunit alpha 1). We further confirmed the critical role of the autophagy-mediated proteolysis in the strong IL21 production in EC by using BECN1 gene silencing as well as protease, PRKAA1, and lysosomal inhibitors. Finally, we established that high autophagy-mediated proteolysis in EC fuels their cellular rates of mitochondrial respiration due to glutaminolysis. Our data confirm the critical role of autophagy in dictating the metabolic input, which is required not only to ensure protective cytotoxic CD8A T cell responses, but also to provide strong IL21 production among antiviral CD4 T cells.Abbreviations: AKG: alpha-ketoglutarate; ART: patients under antiretroviral therapy; ATG7: autophagy related 7; BaF: bafilomycin A1; BECN1: beclin 1; Chloro.: chloroquine; EC: elite controllers; EIF4EBP1: eukaryotic translation initiation factor 4E binding protein 1; FOXO3: forkhead box O3; GLS: glutaminase; GLUD1: glutamate dehydrogenase 1; HIVneg: HIV-1-uninfected control donors; IFNG/IFN-γ: interferon gamma; IL21: interleukin 21; MTOR: mechanistic target of rapamycin kinase; PBMC: peripheral blood mononuclear cells; PRKAA1: protein kinase AMP-activated catalytic subunit alpha 1; SQSTM1: sequestosome 1; TCA: tricarboxylic acid cycle; ULK1: unc-51 like autophagy activating kinase.


Subject(s)
Autophagy , HIV-1 , Adenosine Monophosphate , Antiviral Agents/pharmacology , Autophagy/physiology , CD4-Positive T-Lymphocytes , Humans , Interleukins , Leukocytes, Mononuclear , Protein Kinases
4.
Cell Microbiol ; 23(12): e13398, 2021 12.
Article in English | MEDLINE | ID: mdl-34697890

ABSTRACT

Canid herpesvirus 1 (CHV-1) is a Varicellovirus that causes self-limiting infections in adult dogs but morbidity and mortality in puppies. Using a multipronged approach, we discovered the CHV-1 entry pathway into Madin-Darby canine kidney (MDCK) epithelial cells. We found that CHV-1 triggered extensive host cell membrane lamellipodial ruffling and rapid internalisation of virions in large, uncoated vacuoles, suggestive of macropinocytosis. Treatment with inhibitors targeting key macropinocytosis factors, including inhibitors of Na+ /H+ exchangers, F-actin, myosin light-chain kinase, protein kinase C, p21-activated kinase, phosphatidylinositol-3-kinase and focal adhesion kinase, significantly reduced viral replication. Moreover, the effect was restricted to exposure to the inhibitors early in infection, confirming a role for the macropinocytic machinery during entry. The profile of inhibitors also suggested a role for signalling via integrins and receptor tyrosine kinases in viral entry. In contrast, inhibitors of clathrin, caveolin, microtubules and endosomal acidification did not affect CHV-1 entry into MDCK cells. We found that the virus colocalised with the fluid-phase uptake marker dextran; however, surprisingly, CHV-1 infection did not enhance the uptake of dextran. Thus, our results indicate that CHV-1 uses a macropinocytosis-like, pH-independent entry pathway into MDCK cells, which nevertheless is not based on stimulation of fluid uptake. TAKE AWAYS: CHV-1 enters epithelial cells via a macropinocytosis-like mechanism. CHV-1 induces extensive lamellipodial ruffling. CHV-1 entry into MDCK cells is pH-independent.


Subject(s)
Herpesvirus 1, Canid , Varicellovirus , Animals , Cell Line , Dogs , Hydrogen-Ion Concentration , Kidney , Madin Darby Canine Kidney Cells
5.
ACS Infect Dis ; 7(11): 3034-3051, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34658235

ABSTRACT

The antimicrobial medication malarone (atovaquone/proguanil) is used as a fixed-dose combination for treating children and adults with uncomplicated malaria or as chemoprophylaxis for preventing malaria in travelers. It is an inexpensive, efficacious, and safe drug frequently prescribed around the world. Following anecdotal evidence from 17 patients in the provinces of Quebec and Ontario, Canada, suggesting that malarone/atovaquone may present some benefits in protecting against COVID-19, we sought to examine its antiviral potential in limiting the replication of SARS-CoV-2 in cellular models of infection. In VeroE6 expressing human TMPRSS2 and human lung Calu-3 epithelial cells, we show that the active compound atovaquone at micromolar concentrations potently inhibits the replication of SARS-CoV-2 and other variants of concern including the alpha, beta, and delta variants. Importantly, atovaquone retained its full antiviral activity in a primary human airway epithelium cell culture model. Mechanistically, we demonstrate that the atovaquone antiviral activity against SARS-CoV-2 is partially dependent on the expression of TMPRSS2 and that the drug can disrupt the interaction of the spike protein with the viral receptor, ACE2. Additionally, spike-mediated membrane fusion was also reduced in the presence of atovaquone. In the United States, two clinical trials of atovaquone administered alone or in combination with azithromycin were initiated in 2020. While we await the results of these trials, our findings in cellular infection models demonstrate that atovaquone is a potent antiviral FDA-approved drug against SARS-CoV-2 and other variants of concern in vitro.


Subject(s)
COVID-19 , SARS-CoV-2 , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Atovaquone/pharmacology , Humans , United States
6.
PLoS Pathog ; 17(5): e1009617, 2021 05.
Article in English | MEDLINE | ID: mdl-34043736

ABSTRACT

Urinary tract infections (UTIs) are a common bacterial infectious disease in humans, and strains of uropathogenic Escherichia coli (UPEC) are the most frequent cause of UTIs. During infection, UPEC must cope with a variety of stressful conditions in the urinary tract. Here, we demonstrate that the small RNA (sRNA) RyfA of UPEC strains is required for resistance to oxidative and osmotic stresses. Transcriptomic analysis of the ryfA mutant showed changes in expression of genes associated with general stress responses, metabolism, biofilm formation and genes coding for cell surface proteins. Inactivation of ryfA in UPEC strain CFT073 decreased urinary tract colonization in mice and the ryfA mutant also had reduced production of type 1 and P fimbriae (pili), adhesins which are known to be important for UTI. Furthermore, loss of ryfA also reduced UPEC survival in human macrophages. Thus, ryfA plays a key regulatory role in UPEC adaptation to stress, which contributes to UTI and survival in macrophages.


Subject(s)
Biofilms/growth & development , Escherichia coli Infections/microbiology , RNA, Small Untranslated/genetics , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/genetics , Adaptation, Physiological , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Animals , Fimbriae, Bacterial/metabolism , Gene Expression Profiling , Humans , Macrophages/microbiology , Mice , Osmoregulation , Oxidative Stress , RNA, Bacterial/genetics , Sequence Deletion , Uropathogenic Escherichia coli/growth & development , Uropathogenic Escherichia coli/physiology , Virulence
7.
Cytokine Growth Factor Rev ; 59: 111-117, 2021 06.
Article in English | MEDLINE | ID: mdl-33658144

ABSTRACT

One of the most essential and important building blocks of life is the tryptophan amino acid. As such, the pathways surrounding its metabolism are often crucial for the maintenance of proper cell activity and homeostasis. The ratios of tryptophan to kynurenine, mainly mediated by indoleamine 2,3-dioxygenase activity, is a key parameter in the inflammation as well as immunomodulation of both aseptic and septic diseases. As a result, several studies have been published to better understand the mechanisms by which the tryptophan pathways lead to such outcomes. Many have focused on gut health and cells associated with the given environment, the majority of which constitute regulatory T cells and T helper 17 cells. However, recent studies have highlighted the role of this molecular pathway on its capacity to modulate B cells functions and humoral immunity. Accordingly, the focus of this short review is to examine the key tryptophan pathways and their impact on B cells demonstrated by those studies. A better understanding of the role of tryptophan and its metabolites is crucial for its use in disease prevention and treatments.


Subject(s)
B-Lymphocytes , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase , Inflammation , Kynurenine , Tryptophan
8.
Autophagy ; 17(11): 3408-3423, 2021 11.
Article in English | MEDLINE | ID: mdl-33459125

ABSTRACT

Although macroautophagy/autophagy has been proposed as a critical defense mechanism against HIV-1 by targeting viral components for degradation, its contribution as a catabolic process in providing optimal anti-HIV-1 immunity has never been addressed. The failure to restore proper antiviral CD8A/CD8 T-cell immunity, especially against HIV-1, is still the major limitation of current antiretroviral therapies. Consequently, it is of clinical imperative to provide new strategies to enhance the function of HIV-1-specific CD8A T-cells in patients under antiretroviral treatments (ART). Here, we investigated whether targeting autophagy activity could be an optional solution to make this possible. Our data show that, after both polyclonal and HIV-1-specific activation, CD8A T-cells from ART displayed reduced autophagy-dependent degradation of lysosomal contents when compared to naturally HIV-1 protected elite controllers (EC). We further confirmed in EC, by using specific BECN1 gene silencing and lysosomal inhibitors, the critical role of active autophagy in superior CD8A T-cell protection against HIV-1. More importantly, we found that an IL21 treatment was effective in rescuing the antiviral CD8A T-cell immunity from ART in an autophagy-dependent manner. Finally, we established that IL21-dependent rescue occurred due to the enhanced degradation of endogenous lipids via autophagy, referred to as lipophagy, which fueled the cellular rates of mitochondrial beta-oxidation. In summary, our data show that autophagy/lipophagy can be considered as a therapeutic tool to elicit functional antiviral CD8 T-cell responses. Our results also provide additional insights toward the development of improved T-cell-based prevention and cure strategies against HIV-1.Abbreviations: ART: patients under antiretroviral therapy; BaF: bafilomycin A1; BECN1: beclin 1; CEF: cytomegalo-, Epstein-Barr- and flu-virus peptide pool; Chloro.: chloroquine; EC: elite controllers; FAO: fatty acid beta-oxidation; HIVneg: HIV-1-uninfected control donors; IFNG/IFN-γ: interferon gamma; IL21: interleukin 21; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; PBMC: peripheral blood mononuclear cells; SQSTM1: sequestosome 1; ULK1: unc-51 like autophagy activating kinase 1.


Subject(s)
Autophagy/immunology , Autophagy/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , HIV-1/immunology , Adult , Anti-HIV Agents/therapeutic use , Autophagy/drug effects , Beclin-1/antagonists & inhibitors , Beclin-1/genetics , Beclin-1/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/drug effects , Case-Control Studies , HIV Infections/drug therapy , HIV Infections/immunology , HIV Non-Progressors , Humans , In Vitro Techniques , Interleukins/immunology , Lipid Metabolism/immunology , Lymphocyte Activation , Middle Aged , Mitochondria/metabolism , Oxidation-Reduction
9.
Viruses ; 12(8)2020 08 11.
Article in English | MEDLINE | ID: mdl-32796517

ABSTRACT

This review examines the impact of cannabinoids on viral infections, as well as its effects on the mitochondria of the nervous and immune system. The paper conveys information about the beneficial and negative impacts of cannabinoids on viral infections, especially HIV-1. These include effects on the inflammatory response as well as neuroprotective effects. We also explore non-apoptotic mitochondrial pathways modulated by the activity of cannabinoids, resulting in modifications to cellular functions. As a large part of the literature derives from studies of the nervous system, we first compile the information related to mitochondrial functions in this system, particularly through the CB1 receptor. Finally, we reflect on how this knowledge could complement what has been demonstrated in the immune system, especially in the context of the CB2 receptor and Ca2+ uptake. The overall conclusion of the review is that cannabinoids have the potential to affect a broad range of cell types through mitochondrial modulation, be it through receptor-specific action or not, and that this pathway has a potential implication in cases of viral infection.


Subject(s)
Cannabinoids/immunology , Immunomodulation , Mitochondria/drug effects , Virus Diseases/immunology , Animals , Cannabinoids/administration & dosage , Humans , Immune System/drug effects , Mice , Mitochondria/physiology , Nervous System/drug effects , Receptor, Cannabinoid, CB1/immunology , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/immunology , Receptor, Cannabinoid, CB2/metabolism
10.
Cytokine Growth Factor Rev ; 55: 26-36, 2020 10.
Article in English | MEDLINE | ID: mdl-32151523

ABSTRACT

The notion of immuno-metabolism refers to the crosstalk between key metabolic pathways and the development/maintenance of protective immunity in the context of physiological processes and anti-microbial defenses. Enthusiasm for immuno-metabolism in the context of HIV-1 infection, especially among T-cell lineages, continues to grow over time as science opens new therapeutic perspectives to limit viral pathogenesis and to boost anti-viral responses. The idea of "metabolism as a therapeutic target" is called metabolic reprogramming and is based on the use of specific metabolism-targeting drugs that are currently available for cancer therapy. In this review, we will focus on the evidence that shows the key role of mitochondria, the cell's powerhouses, and their ability to use diverse metabolic resources (referred to as metabolic plasticity) in providing optimal immune T-cell protection among HIV-1-infected patients. Conversely, we highlight observations indicating that mitochondria metabolic dysfunction associated with excessive glucose dependency, a phenomenon reported as "Warburg effect", results in the inability to mount and maintain effective T-cell-dependent immunity during persistent HIV-1 infection. Therefore, helping mitochondria to regain the metabolic plasticity and allow specific T-cells to adapt and thrive under unfavorable environmental conditions during HIV-1 infection may represent the next generation of combinatory treatment options for patients.


Subject(s)
HIV Infections , HIV-1 , Mitochondria , T-Lymphocytes , Cell Plasticity , HIV Infections/metabolism , Humans , Metabolic Networks and Pathways , Mitochondria/metabolism , T-Lymphocytes/metabolism
11.
PLoS Pathog ; 15(10): e1008060, 2019 10.
Article in English | MEDLINE | ID: mdl-31658294

ABSTRACT

The loss of Memory CD4 T-cells (Mem) is a major hallmark of HIV-1 immuno-pathogenesis and occurs early during the first months of primary infection. A lot of effort has been put into understanding the molecular mechanisms behind this loss, yet they still have not been fully identified. In this study, we unveil the unreported role of USP18 in the deleterious effects of sustained type I IFN signaling on Mem, including HIV-1-specific CD4 T-cells. We find that interfering with IFN-I signaling pathway in infected patients, notably by targeting the interferon-stimulated gene USP18, resulted in reduced PTEN expression similar to those observed in uninfected control donors. We show that AKT activation in response to cytokine treatment, T-cell receptor (TcR) triggering, as well as HIV-1 Gag stimulation was significantly improved in infected patients when PTEN or USP18 were inhibited. Finally, our data demonstrate that higher USP18 in Mem from infected patients prevent proper cell survival and long-lasting maintenance in an AKT-dependent manner. Altogether, we establish a direct role for type I IFN/USP18 signaling in the maintenance of total and virus-specific Mem and provide a new mechanism for the reduced survival of these populations during primary HIV-1 infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV-1/immunology , Immunologic Memory/immunology , Interferon Type I/immunology , PTEN Phosphohydrolase/biosynthesis , Ubiquitin Thiolesterase/metabolism , Apoptosis/immunology , Cell Survival , HIV Infections/immunology , HIV Infections/pathology , HIV-1/pathogenicity , Humans , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Ubiquitin Thiolesterase/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
12.
Cell Rep ; 24(5): 1163-1175, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30067973

ABSTRACT

The transcription factor interferon regulatory factor 5 (IRF-5) plays an important function in innate immunity and in initiating pro-inflammatory responses against pathogens. IRF-5 is constitutively expressed in several cell types, including plasmacytoid dendritic cells, monocytes, and B cells. We have previously reported that IRF-5 is also expressed in T cells during infection. The role of IRF-5 in T cells is yet unknown. Here, we demonstrate that IRF-5 is increasingly expressed in interferon (IFN)-γ+ CD4 T cells over the course of L. donovani infection. This transcription factor is induced by apoptotic material via Toll-like receptor 7 (TLR7) and promotes the expression of death receptor 5 (DR5). IRF-5 activation sensitizes CD4 T cells to cell death. Because tissue disruption and chronic inflammation are common characteristics of persistent infections, activation of IRF-5 in CD4 T cells may represent a common pathway that leads to suppression of protective CD4 T cell responses, favoring the establishment of chronic infection.


Subject(s)
Apoptosis , CD4-Positive T-Lymphocytes/metabolism , Interferon Regulatory Factors/metabolism , Leishmaniasis, Visceral/metabolism , Animals , Cells, Cultured , Female , Interferon Regulatory Factors/genetics , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Toll-Like Receptor 7/metabolism
13.
Cytokine Growth Factor Rev ; 40: 90-98, 2018 04.
Article in English | MEDLINE | ID: mdl-29778137

ABSTRACT

Antiretroviral therapy (ART) has dramatically reduced HIV-1-associated morbidity and mortality, and has transformed HIV-1 infection into a manageable chronic condition by suppressing viral replication. However, despite recent patient care improvements, ART still fails to cure HIV-1 infection due to the inability to counteract immune defects and metabolic disturbances that are associated with residual inflammation alongside viral persistence. Life-long drug administration also results in multiple side-effects in patients including lipodystrophy and insulin resistance. Thus, it is critical to find new ways to reduce the length of treatment and facilitate the termination of ART, for example by boosting protective immunity. The rare ability of some individuals to naturally control HIV-1 infection despite residual inflammation could be exploited to identify molecular mechanisms involved in host protection that may function as potential therapeutic targets. In this review, we highlight evidence illustrating the molecular and metabolic advantages of HIV-1 controllers over ART treated patients that contribute to the maintenance of effective antiviral immunity.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV-1/drug effects , HIV-1/immunology , Viral Load/drug effects , Viral Load/immunology , Anti-HIV Agents/adverse effects , Antiretroviral Therapy, Highly Active , HIV Infections/immunology , Host-Pathogen Interactions/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Virus Replication/drug effects
14.
Viruses ; 10(1)2017 12 30.
Article in English | MEDLINE | ID: mdl-29301196

ABSTRACT

Type I interferons (IFN-I) are one of the primary immune defenses against viruses. Similar to all other molecular mechanisms that are central to eliciting protective immune responses, IFN-I expression is subject to homeostatic controls that regulate cytokine levels upon clearing the infection. However, in the case of established persistent viral infection, sustained elevation of IFN-I expression bears deleterious effects to the host and is today considered as the major driver of inflammation and immunosuppression. In fact, numerous emerging studies place sustained IFN-I expression as a common nexus in the pathogenesis of multiple chronic diseases including persistent infections with the human immunodeficiency virus type 1 (HIV-1), simian immunodeficiency virus (SIV), as well as the rodent-borne lymphocytic choriomeningitis virus clone 13 (LCMV clone 13). In this review, we highlight recent studies illustrating the molecular dysregulation and resultant cellular dysfunction in both innate and adaptive immune responses driven by sustained IFN-I expression. Here, we place particular emphasis on the efficacy of IFN-I receptor (IFNR) blockade towards improving immune responses against viral infections given the emerging therapeutic approach of blocking IFNR using neutralizing antibodies (Abs) in chronically infected patients.


Subject(s)
Interferon Type I/genetics , Signal Transduction/immunology , Virus Diseases/immunology , Animals , Antiviral Agents/therapeutic use , Chronic Disease , Humans , Immune Tolerance/immunology , Interferon Type I/metabolism , Models, Immunological , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/drug effects , Virus Diseases/drug therapy , Virus Diseases/genetics
15.
J Virol ; 90(17): 7967-79, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27356894

ABSTRACT

UNLABELLED: Early HIV-1 infection is characterized by enhanced tryptophan catabolism, which contributes to immune suppression and disease progression. However, the mechanism by which kynurenine, a tryptophan-related metabolite, induces immune suppression remains poorly understood. Herein, we show that the increased production of kynurenine correlates with defective interleukin-2 (IL-2) signaling in memory CD4 T cells from HIV-infected subjects. Defective IL-2 signaling in these subjects, which drives reduced protection from Fas-mediated apoptosis, was also associated with memory CD4 T-cell loss. Treatment of memory CD4 T cells with the concentration of kynurenine found in plasma inhibited IL-2 signaling through the production of reactive oxygen species. We further show that IL-2 signaling in memory CD4 T cells is improved by the antioxidant N-acetylcysteine. Early initiation of antiretroviral therapy restored the IL-2 response in memory CD4 T cells by reducing reactive oxygen species and kynurenine production. The study findings provide a kynurenine-dependent mechanism through IL-2 signaling for reduced CD4 T-cell survival, which can be reversed by early treatment initiation in HIV-1 infection. IMPORTANCE: The persistence of functional memory CD4 T cells represents the basis for long-lasting immune protection in individuals after exposure to HIV-1. Unfortunately, primary HIV-1 infection results in the massive loss of these cells within weeks of infection, which is mainly driven by inflammation and massive infection by the virus. These new findings show that the enhanced production of kynurenine, a metabolite related to tryptophan catabolism, also impairs memory CD4 T-cell survival and interferes with IL-2 signaling early during HIV-1 infection.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Immunosuppressive Agents/metabolism , Interleukin-2/metabolism , Kynurenine/metabolism , Adult , Cell Survival/drug effects , Humans , Middle Aged , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Young Adult
16.
Sci Rep ; 6: 22902, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26978598

ABSTRACT

HIV-infected slow progressors (SP) represent a heterogeneous group of subjects who spontaneously control HIV infection without treatment for several years while showing moderate signs of disease progression. Under conditions that remain poorly understood, a subgroup of these subjects experience failure of spontaneous immunological and virological control. Here we determined the frequency of SP subjects who showed loss of HIV control within our Canadian Cohort of HIV(+) Slow Progressors and identified the proinflammatory cytokine IL-32 as a robust biomarker for control failure. Plasmatic levels of the proinflammatory isoforms of IL-32 (mainly ß and γ) at earlier clinic visits positively correlated with the decline of CD4 T-cell counts, increased viral load, lower CD4/CD8 ratio and levels of inflammatory markers (sCD14 and IL-6) at later clinic visits. We present here a proof-of-concept for the use of IL-32 as a predictive biomarker for disease progression in SP subjects and identify IL-32 as a potential therapeutic target.


Subject(s)
Biomarkers/blood , HIV Infections/blood , Inflammation Mediators/blood , Interleukins/blood , Adult , CD4 Lymphocyte Count , CD4-CD8 Ratio , Cells, Cultured , Cohort Studies , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Profiling/methods , HIV Infections/genetics , HIV Infections/immunology , HIV-1/immunology , Humans , Interleukin-6/blood , Interleukin-6/genetics , Interleukins/genetics , Lipopolysaccharide Receptors/blood , Lipopolysaccharide Receptors/genetics , Male , Oligonucleotide Array Sequence Analysis , Protein Isoforms/blood , Protein Isoforms/genetics , Viral Load/immunology
17.
Cytokine Growth Factor Rev ; 28: 1-10, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26851985

ABSTRACT

HIV-1 infection leads to a depletion of CD4 T-cells associated with a persistent immune inflammation and changes in cellular metabolism. Most effort of managing HIV infection with combination of antiretroviral therapies (ART) has been focused on CD4 T-cell recovery, while control of persistent immune inflammation and metabolism were relatively underappreciated in the past. Recent discoveries on the interplay between innate immunity, inflammation (especially the inflammasome) and metabolic changes in the context of cancer and autoimmunity provide an emerging field for chronic viral infections including HIV-1. In a previous review, we described the deregulated metabolism contributing to immune dysfunctions such as alteration of memory T-cell responses, mucosal protection, and dendritic cell-related antigen presentation. Here, we summarize the latest knowledge on the detrimental influence of long-lasting inflammation and inflammasome activation induced by HIV-1, gut dysbiosis, and bacterial translocation, on metabolism during the course of viral infection. We also report on the inability of ART to fully counteract inflammation, resulting in partial metabolic improvement and leading to an insufficient decrease in the risk of non-AIDS events. Further advances in our understanding of the relationship between inflammation, altered metabolism, and long-term ART is warranted. Additionally, there is a critical need for developing new strategies to regulate the pro-inflammatory signals to enhance cellular metabolism and immune functions in order to improve the quality of life of individuals living with HIV-1.


Subject(s)
HIV Infections/metabolism , HIV-1/immunology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , Humans , Immunity, Innate/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation/virology , Quality of Life
18.
J Immunol ; 195(12): 5625-36, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26546609

ABSTRACT

Despite the overwhelming benefits of antiretroviral therapy (ART) in curtailing viral load in HIV-infected individuals, ART does not fully restore cellular and humoral immunity. HIV-infected individuals under ART show reduced responses to vaccination and infections and are unable to mount an effective antiviral immune response upon ART cessation. Many factors contribute to these defects, including persistent inflammation, especially in lymphoid tissues, where T follicular helper (Tfh) cells instruct and help B cells launch an effective humoral immune response. In this study we investigated the phenotype and function of circulating memory Tfh cells as a surrogate of Tfh cells in lymph nodes and found significant impairment of this cell population in chronically HIV-infected individuals, leading to reduced B cell responses. We further show that these aberrant memory Tfh cells exhibit an IL-2-responsive gene signature and are more polarized toward a Th1 phenotype. Treatment of functional memory Tfh cells with IL-2 was able to recapitulate the detrimental reprogramming. Importantly, this defect was reversible, as interfering with the IL-2 signaling pathway helped reverse the abnormal differentiation and improved Ab responses. Thus, reversible reprogramming of memory Tfh cells in HIV-infected individuals could be used to enhance Ab responses. Altered microenvironmental conditions in lymphoid tissues leading to altered Tfh cell differentiation could provide one explanation for the poor responsiveness of HIV-infected individuals to new Ags. This explanation has important implications for the development of therapeutic interventions to enhance HIV- and vaccine-mediated Ab responses in patients under ART.


Subject(s)
B-Lymphocytes/immunology , HIV Infections/immunology , HIV , Interleukin-2/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adult , Antibodies, Viral/immunology , Antibody Formation , B-Lymphocytes/virology , Cell Differentiation , Cells, Cultured , Cellular Reprogramming , Chronic Disease , Humans , Immunologic Memory , Middle Aged , Signal Transduction , T-Lymphocytes, Helper-Inducer/virology , Young Adult
19.
J Gen Virol ; 96(Pt 6): 1463-1477, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25626682

ABSTRACT

CTLA-4 is a negative regulator of T-cell receptor-mediated CD4(+) T-cell activation and function. Upregulation of CTLA-4 during human immunodeficiency virus type 1 (HIV-1) infection on activated T cells, particularly on HIV-specific CD4(+) T cells, correlates with immune dysfunction and disease progression. As HIV-1 infects and replicates in activated CD4(+) T cells, we investigated mechanisms by which HIV-1 modulates CTLA-4 expression to establish productive viral infection in these cells. Here, we demonstrate that HIV-1 infection in activated CD4(+) T cells was followed by Nef-mediated downregulation of CTLA-4. This was associated with a decreased T-cell activation threshold and significant resistance to CTLA-4 triggering. In line with these in vitro results, quantification of pro-viral HIV DNA from treatment-naive HIV-infected subjects demonstrated a preferential infection of memory CD4(+)CTLA-4(+) T cells, thus identifying CTLA-4 as a biomarker for HIV-infected cells in vivo. As transcriptionally active HIV-1 and Nef expression in vivo were previously shown to take place mainly in the CD3(+)CD4(-)CD8(-) [double-negative (DN)] cells, we further quantified HIV DNA in the CTLA-4(+) and CTLA-4(-) subpopulations of these cells. Our results showed that DN T cells lacking CTLA-4 expression were enriched in HIV DNA compared with DN CTLA-4(+) cells. Together, these results suggested that HIV-1 preferential infection of CD4(+)CTLA-4(+) T cells in vivo was followed by Nef-mediated concomitant downregulation of both CD4 and CTLA-4 upon transition to productive infection. This also highlights the propensity of HIV-1 to evade restriction of the key negative immune regulator CTLA-4 on cell activation and viral replication, and therefore contributes to the overall HIV-1 pathogenesis.


Subject(s)
CD4-Positive T-Lymphocytes/virology , CTLA-4 Antigen/biosynthesis , HIV-1/physiology , Host-Pathogen Interactions , Immune Evasion , T-Lymphocytes, Cytotoxic/immunology , nef Gene Products, Human Immunodeficiency Virus/metabolism , Adult , CD4-Positive T-Lymphocytes/immunology , Female , Gene Expression Regulation , HIV-1/immunology , Humans , Male
20.
PLoS Pathog ; 10(12): e1004575, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25521510

ABSTRACT

The mechanisms involved in the persistence of activated CD4+ T lymphocytes following primary human T leukemia/lymphoma virus type 1 (HTLV-1) infection remain unclear. Here, we demonstrate that the HTLV-1 Tax oncoprotein modulates phosphorylation and transcriptional activity of the FOXO3a transcription factor, via upstream activation of the AKT pathway. De novo HTLV-1 infection of CD4+ T cells or direct lentiviral-mediated introduction of Tax led to AKT activation and AKT-dependent inactivation of FOXO3a, via phosphorylation of residues Ser253 and Thr32. Inhibition of FOXO3a signalling led to the long-term survival of a population of highly activated, terminally differentiated CD4+Tax+CD27negCCR7neg T cells that maintained the capacity to disseminate infectious HTLV-1. CD4+ T cell persistence was reversed by chemical inhibition of AKT activity, lentiviral-mediated expression of a dominant-negative form of FOXO3a or by specific small interfering RNA (siRNA)-mediated silencing of FOXO3a. Overall this study provides new mechanistic insight into the strategies used by HTLV-1 to increase long-term maintenance of Tax+CD4+ T lymphocytes during the early stages of HTLV-1 pathogenesis.


Subject(s)
CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , Forkhead Transcription Factors/antagonists & inhibitors , Gene Products, tax/physiology , HTLV-I Infections/physiopathology , Human T-lymphotropic virus 1/physiology , Cell Differentiation , Cell Survival/physiology , Cells, Cultured , Forkhead Box Protein O3 , Forkhead Transcription Factors/drug effects , Forkhead Transcription Factors/physiology , HTLV-I Infections/pathology , Humans , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , RNA, Small Interfering/pharmacology , Signal Transduction/physiology , Viral Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...