Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
J Neuroinflammation ; 21(1): 127, 2024 May 13.
Article En | MEDLINE | ID: mdl-38741181

HIV-associated neurocognitive disorders (HAND) are a spectrum of cognitive impairments that continue to affect approximately half of all HIV-positive individuals despite effective viral suppression through antiretroviral therapy (ART). White matter pathologies have persisted in the ART era, and the degree of white matter damage correlates with the degree of neurocognitive impairment in patients with HAND. The HIV protein Nef has been implicated in HAND pathogenesis, but its effect on white matter damage has not been well characterized. Here, utilizing in vivo, ex vivo, and in vitro methods, we demonstrate that Nef-containing extracellular vesicles (Nef EVs) disrupt myelin sheaths and inflict damage upon oligodendrocytes within the murine central nervous system. Intracranial injection of Nef EVs leads to reduced myelin basic protein (MBP) staining and a decreased number of CC1 + oligodendrocytes in the corpus callosum. Moreover, cerebellar slice cultures treated with Nef EVs exhibit diminished MBP expression and increased presence of unmyelinated axons. Primary mixed brain cultures and enriched oligodendrocyte precursor cell cultures exposed to Nef EVs display a decreased number of O4 + cells, indicative of oligodendrocyte impairment. These findings underscore the potential contribution of Nef EV-mediated damage to oligodendrocytes and myelin maintenance in the pathogenesis of HAND.


Extracellular Vesicles , HIV-1 , Mice, Inbred C57BL , Oligodendroglia , nef Gene Products, Human Immunodeficiency Virus , Animals , Oligodendroglia/metabolism , Oligodendroglia/pathology , Oligodendroglia/virology , Mice , Extracellular Vesicles/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , HIV-1/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Central Nervous System/virology , Cells, Cultured , Humans , Male
2.
Cell Rep ; 41(8): 111674, 2022 11 22.
Article En | MEDLINE | ID: mdl-36417867

A possible explanation for chronic inflammation in HIV-infected individuals treated with anti-retroviral therapy is hyperreactivity of myeloid cells due to a phenomenon called "trained immunity." Here, we demonstrate that human monocyte-derived macrophages originating from monocytes initially treated with extracellular vesicles containing HIV-1 protein Nef (exNef), but differentiating in the absence of exNef, release increased levels of pro-inflammatory cytokines after lipopolysaccharide stimulation. This effect is associated with chromatin changes at the genes involved in inflammation and cholesterol metabolism pathways and upregulation of the lipid rafts and is blocked by methyl-ß-cyclodextrin, statin, and an inhibitor of the lipid raft-associated receptor IGF1R. Bone-marrow-derived macrophages from exNef-injected mice, as well as from mice transplanted with bone marrow from exNef-injected animals, produce elevated levels of tumor necrosis factor α (TNF-α) upon stimulation. These phenomena are consistent with exNef-induced trained immunity that may contribute to persistent inflammation and associated co-morbidities in HIV-infected individuals with undetectable HIV load.


Extracellular Vesicles , HIV Infections , HIV Seropositivity , HIV-1 , Humans , Mice , Animals , HIV-1/genetics , nef Gene Products, Human Immunodeficiency Virus/genetics , Extracellular Vesicles/metabolism , Macrophages/metabolism , Inflammation/metabolism
3.
Cardiovasc Diabetol ; 20(1): 44, 2021 02 13.
Article En | MEDLINE | ID: mdl-33581737

BACKGROUND: Endothelial progenitor cells (EPCs) has been shown to be dysfunctional in both type 2 diabetes mellitus (T2DM) and chronic kidney disease (CKD) leading to poor regeneration of endothelium and renal perfusion. EPCs have been shown to be a robust cardiovascular disease (CVD) risk indicator. Effect of sodium glucose channel inhibitors (SGLT2i) such as Canagliflozin (CG) on a cellular biomarker such as CD34+ve progenitor cells, which may help predict CVD risk, in patients with T2DM with established CKD has not been explored. METHODS: This is a pilot study where 29 subjects taking metformin and/or Insulin were enrolled in a 16 week, double blind, randomized placebo matched trial, with a low dose 100 mg CG as the intervention group compared to matched placebo. Type 2 diabetes subjects (30-70 years old), with hemoglobin A1c (HbA1c) of 7-10%, were enrolled. CD34+ve cell number, migratory function, gene expression along with vascular parameters such as arterial stiffness, serum biochemistry pertaining to cardio-metabolic health, resting energy expenditure and body composition were measured. Data were collected at week 0, 8 and 16. A mixed model regression analysis was done and p value less than 0.05 was considered statistically significant. RESULTS: A significant expression of CXCR4 receptor with a concomittant increase in migratory function of CD34+ve cells was observed in CG treated group as compared to placebo group. Gene expression analysis of CD34+ve cells showed an increase in expression of antioxidants (superoxide dismutase 2 or SOD2, Catalase and Glutathione Peroxidase or GPX) and notable endothelial markers (PECAM1, VEGF-A, and NOS3). A significant reduction in glucose and HbA1c levels were observed along with improved systolic and diastolic blood pressure in the CG group. A significant increase in adiponectin (p = 0.006) was also noted in treatment group. Urinary exosomal protein leak in urine, examining podocyte health (podocalyxin, Wilm's tumor and nephrin) showed reduction with CG CONCLUSION: Low dose Canagliflozin has a beneficial effect on CD34+ cell function, serum biochemistry and urinary podocyte specific exosomes in type 2 diabetes.


Antigens, CD34/metabolism , Blood Glucose/drug effects , Canagliflozin/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Endothelial Progenitor Cells/drug effects , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Adult , Aged , Biomarkers/blood , Blood Glucose/metabolism , Canagliflozin/adverse effects , Cells, Cultured , Chemotaxis/drug effects , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Double-Blind Method , Drug Therapy, Combination , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Exosomes/drug effects , Exosomes/metabolism , Female , Glycated Hemoglobin/metabolism , Humans , Insulin/therapeutic use , Male , Metformin/therapeutic use , Middle Aged , Phenotype , Pilot Projects , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Time Factors , Treatment Outcome
4.
Cardiovasc Diabetol ; 19(1): 72, 2020 06 03.
Article En | MEDLINE | ID: mdl-32493344

BACKGROUND: Endothelial Progenitor cells (EPCs) has been shown to be dysfunctional in both type 2 diabetes mellitus (T2DM) and chronic kidney disease (CKD) leading to poor regeneration of endothelium and renal perfusion. EPCs have been shown to be a robust cardiovascular disease (CVD) risk indicator. Cellular mechanisms of DPP4 inhibitors such as linagliptin (LG) on CVD risk, in patients with T2DM with established CKD has not been established. Linagliptin, a DPP4 inhibitor when added to insulin, metformin or both may improve endothelial dysfunction in a diabetic kidney disease (DKD) population. METHODS: 31 subjects taking metformin and/or Insulin were enrolled in this 12 weeks, double blind, randomized placebo matched trial, with 5 mg LG compared to placebo. Type 2 diabetes subjects (30-70 years old), HbA1c of 6.5-10%, CKD Stage 1-3 were included. CD34+ cell number, migratory function, gene expression along with vascular parameters such as arterial stiffness, biochemistry, resting energy expenditure and body composition were measured. Data were collected at week 0, 6 and 12. A mixed model regression analysis was done with p value < 0.05 considered significant. RESULTS: A double positive CD34/CD184 cell count had a statistically significant increase (p < 0.02) as determined by flow cytometry in LG group where CD184 is SDF1a cell surface receptor. Though mRNA differences in CD34+ve was more pronounced CD34- cell mRNA analysis showed increase in antioxidants (superoxide dismutase 2 or SOD2, Catalase and Glutathione Peroxidase or GPX) and prominent endothelial markers (PECAM1, VEGF-A, vWF and NOS3). Arterial stiffness measures such as augmentation Index (AI) (p < 0.04) and pulse wave analysis (PWV) were improved (reduced in stiffness) in LG group. A reduction in LDL: HDL ratio was noted in treatment group (p < 0.04). Urinary exosome protein examining podocyte health (podocalyxin, Wilms tumor and nephrin) showed reduction or improvement. CONCLUSIONS: In DKD subjects, Linagliptin promotes an increase in CXCR4 expression on CD34 + progenitor cells with a concomitant improvement in vascular and renal parameters at 12 weeks. Trial Registration Number NCT02467478 Date of Registration: 06/08/2015.


Antigens, CD34/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetic Nephropathies/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Endothelial Progenitor Cells/drug effects , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Linagliptin/therapeutic use , Metformin/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Adult , Aged , Biomarkers/blood , Cells, Cultured , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetic Nephropathies/blood , Diabetic Nephropathies/diagnosis , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , District of Columbia , Double-Blind Method , Drug Therapy, Combination , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Female , Humans , Hypoglycemic Agents/adverse effects , Insulin/adverse effects , Linagliptin/adverse effects , Male , Metformin/adverse effects , Middle Aged , Pilot Projects , Receptors, CXCR4/blood , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/diagnosis , Time Factors , Treatment Outcome
6.
mBio ; 11(1)2020 01 21.
Article En | MEDLINE | ID: mdl-31964734

Apolipoprotein A-I binding protein (AIBP) is a protein involved in regulation of lipid rafts and cholesterol efflux. AIBP has been suggested to function as a protective factor under several sets of pathological conditions associated with increased abundance of lipid rafts, such as atherosclerosis and acute lung injury. Here, we show that exogenously added AIBP reduced the abundance of lipid rafts and inhibited HIV replication in vitro as well as in HIV-infected humanized mice, whereas knockdown of endogenous AIBP increased HIV replication. Endogenous AIBP was much more abundant in activated T cells than in monocyte-derived macrophages (MDMs), and exogenous AIBP was much less effective in T cells than in MDMs. AIBP inhibited virus-cell fusion, specifically targeting cells with lipid rafts mobilized by cell activation or Nef-containing exosomes. MDM-HIV fusion was sensitive to AIBP only in the presence of Nef provided by the virus or exosomes. Peripheral blood mononuclear cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, bound less AIBP than cells from donors with other HLA genotypes and were not protected by AIBP from rapid HIV-1 replication. These results provide the first evidence for the role of Nef exosomes in regulating HIV-cell fusion by modifying lipid rafts and suggest that AIBP is an innate factor that restricts HIV replication by targeting lipid rafts.IMPORTANCE Apolipoprotein A-I binding protein (AIBP) is a recently identified innate anti-inflammatory factor. Here, we show that AIBP inhibited HIV replication by targeting lipid rafts and reducing virus-cell fusion. Importantly, AIBP selectively reduced levels of rafts on cells stimulated by an inflammatory stimulus or treated with extracellular vesicles containing HIV-1 protein Nef without affecting rafts on nonactivated cells. Accordingly, fusion of monocyte-derived macrophages with HIV was sensitive to AIBP only in the presence of Nef. Silencing of endogenous AIBP significantly upregulated HIV-1 replication. Interestingly, HIV-1 replication in cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, was not inhibited by AIBP. These results suggest that AIBP is an innate anti-HIV factor that targets virus-cell fusion.

7.
PLoS Pathog ; 15(7): e1007907, 2019 07.
Article En | MEDLINE | ID: mdl-31344124

HIV infection has a profound effect on "bystander" cells causing metabolic co-morbidities. This may be mediated by exosomes secreted by HIV-infected cells and containing viral factors. Here we show that exosomes containing HIV-1 protein Nef (exNef) are rapidly taken up by macrophages releasing Nef into the cell interior. This caused down-regulation of ABCA1, reduction of cholesterol efflux and sharp elevation of the abundance of lipid rafts through reduced activation of small GTPase Cdc42 and decreased actin polymerization. Changes in rafts led to re-localization of TLR4 and TREM-1 to rafts, phosphorylation of ERK1/2, activation of NLRP3 inflammasome, and increased secretion of pro-inflammatory cytokines. The effects of exNef on lipid rafts and on inflammation were reversed by overexpression of a constitutively active mutant of Cdc42. Similar effects were observed in macrophages treated with exosomes produced by HIV-infected cells or isolated from plasma of HIV-infected subjects, but not with exosomes from cells and subjects infected with ΔNef-HIV or uninfected subjects. Mice injected with exNef exhibited monocytosis, reduced ABCA1 in macrophages, increased raft abundance in monocytes and augmented inflammation. Thus, Nef-containing exosomes potentiated pro-inflammatory response by inducing changes in cholesterol metabolism and reorganizing lipid rafts. These mechanisms may contribute to HIV-associated metabolic co-morbidities.


HIV Infections/metabolism , HIV Infections/virology , nef Gene Products, Human Immunodeficiency Virus/metabolism , ATP Binding Cassette Transporter 1/metabolism , Animals , Bystander Effect , Cholesterol/metabolism , Exosomes/metabolism , Exosomes/virology , HEK293 Cells , HIV-1 , Humans , Inflammation/metabolism , Inflammation/virology , Membrane Microdomains/metabolism , Membrane Microdomains/virology , Mice , Mice, Inbred C57BL , Models, Biological , RAW 264.7 Cells , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Toll-Like Receptor 4/metabolism , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , cdc42 GTP-Binding Protein/metabolism , nef Gene Products, Human Immunodeficiency Virus/genetics
8.
AIDS Res Hum Retroviruses ; 33(1): 57-60, 2017 01.
Article En | MEDLINE | ID: mdl-27649790

HIV-infected individuals are at high risk of developing atherosclerosis and cardiovascular disease, in part, due to HIV-induced impairment of cholesterol metabolism. In vitro studies demonstrated that HIV-1 protein Nef inhibits activity of ABCA1, the main cellular cholesterol transporter, leading to cholesterol accumulation in macrophages and conversion of these cells into foam cells, characteristic for atherosclerosis. However, the mechanisms of Nef-mediated effects on cholesterol metabolism in vivo are not well characterized. In this study, we generated Nef-transgenic mice and evaluated the accumulation of neutral lipids in liver and aorta of these animals. Nef expression was low in all transgenic mice, with some mice carrying the Nef transgene, but not expressing the Nef RNA. Using Oil Red O staining, we demonstrated increased levels of neutral lipids in liver and aorta of mice expressing Nef relative to transgenic animals, with no detectable Nef expression or control wild-type mice. These results provide direct evidence that Nef promotes cholesterol deposition in tissues.


Aorta/pathology , Lipids/analysis , Liver/pathology , nef Gene Products, Human Immunodeficiency Virus/biosynthesis , Animals , Histocytochemistry , Mice, Inbred C57BL , Mice, Transgenic , Staining and Labeling , nef Gene Products, Human Immunodeficiency Virus/genetics
9.
Virology ; 497: 11-22, 2016 10.
Article En | MEDLINE | ID: mdl-27414250

The post-entry events of HIV-1 infection occur within reverse transcription complexes derived from the viral cores entering the target cell. HIV-1 cores contain host proteins incorporated from virus-producing cells. In this report, we show that MCM5, a subunit of the hexameric minichromosome maintenance (MCM) DNA helicase complex, associates with Gag polyprotein and is incorporated into HIV-1 virions. The progeny virions depleted of MCM5 demonstrated reduced reverse transcription in newly infected cells, but integration and subsequent replication steps were not affected. Interestingly, increased packaging of MCM5 into the virions also led to reduced reverse transcription, but here viral replication was impaired. Our data suggest that incorporation of physiological amounts of MCM5 promotes aberrant reverse transcription, leading to partial incapacitation of cDNA, whereas increased MCM5 abundance leads to reduced reverse transcription and infection. Therefore, MCM5 has the properties of an inhibitory factor that interferes with production of an integration-competent cDNA product.


Cell Cycle Proteins/metabolism , HIV-1/physiology , Virion , Virus Replication , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line , Cells, Cultured , HIV Infections/virology , Humans , Macrophages/metabolism , Macrophages/virology , Multiprotein Complexes/metabolism , Protein Binding , Protein Transport , gag Gene Products, Human Immunodeficiency Virus/metabolism
10.
Arterioscler Thromb Vasc Biol ; 36(9): 1758-71, 2016 09.
Article En | MEDLINE | ID: mdl-27470515

OBJECTIVE: HIV-infected patients are at an increased risk of developing atherosclerosis, in part because of downmodulation and functional impairment of ATP-binding cassette A1 (ABCA1) cholesterol transporter by the HIV-1 protein Nef. The mechanism of this effect involves Nef interacting with an ER chaperone calnexin and disrupting calnexin binding to ABCA1, leading to ABCA1 retention in ER, its degradation and resulting suppression of cholesterol efflux. However, molecular details of Nef-calnexin interaction remained unknown, limiting the translational impact of this finding. APPROACH AND RESULTS: Here, we used molecular modeling and mutagenesis to characterize Nef-calnexin interaction and to identify small molecule compounds that could block it. We demonstrated that the interaction between Nef and calnexin is direct and can be reconstituted using recombinant proteins in vitro with a binding affinity of 89.1 nmol/L measured by surface plasmon resonance. The cytoplasmic tail of calnexin is essential and sufficient for interaction with Nef, and binds Nef with an affinity of 9.4 nmol/L. Replacing lysine residues in positions 4 and 7 of Nef with alanines abrogates Nef-calnexin interaction, prevents ABCA1 downregulation by Nef, and preserves cholesterol efflux from HIV-infected cells. Through virtual screening of the National Cancer Institute library of compounds, we identified a compound, 1[(7-oxo-7H-benz[de]anthracene-3-yl)amino]anthraquinone, which blocked Nef-calnexin interaction, partially restored ABCA1 activity in HIV-infected cells, and reduced foam cell formation in a culture of HIV-infected macrophages. CONCLUSION: This study identifies potential targets that can be exploited to block the pathogenic effect of HIV infection on cholesterol metabolism and prevent atherosclerosis in HIV-infected subjects.


Anthraquinones/pharmacology , Atherosclerosis/prevention & control , Calnexin/metabolism , Cholesterol/metabolism , Drug Design , HIV Infections/drug therapy , Hypolipidemic Agents/pharmacology , Molecular Docking Simulation , nef Gene Products, Human Immunodeficiency Virus/metabolism , ATP Binding Cassette Transporter 1/metabolism , Anthraquinones/chemistry , Atherosclerosis/metabolism , Atherosclerosis/virology , Biological Transport , Calnexin/chemistry , Calnexin/genetics , Computer-Aided Design , Foam Cells/drug effects , Foam Cells/metabolism , HEK293 Cells , HIV Infections/metabolism , HIV Infections/virology , Humans , Hypolipidemic Agents/chemistry , Lysine , Mutation , Protein Binding , Protein Interaction Domains and Motifs , Structure-Activity Relationship , Transfection , nef Gene Products, Human Immunodeficiency Virus/chemistry , nef Gene Products, Human Immunodeficiency Virus/genetics
12.
Methods Mol Biol ; 1354: 281-92, 2016.
Article En | MEDLINE | ID: mdl-26714719

Cholesterol is an essential component of the cellular membranes and, by extension, of the HIV envelope membrane, which is derived from the host cell plasma membrane. Depletion of the cellular cholesterol has an inhibitory effect on HIV assembly, reduces infectivity of the produced virions, and makes the cell less susceptible to HIV infection. It is not surprising that the virus has evolved to gain access to cellular proteins regulating cholesterol metabolism. One of the key mechanisms used by HIV to maintain high levels of cholesterol in infected cells is Nef-mediated inhibition of cholesterol efflux and the cholesterol transporter responsible for this process, ABCA1. In this chapter, we describe methods to investigate these effects of HIV-1 infection.


ATP Binding Cassette Transporter 1/metabolism , Cholesterol/metabolism , HIV Infections/metabolism , HIV-1/physiology , Microscopy, Confocal/methods , nef Gene Products, Human Immunodeficiency Virus/metabolism , Biological Transport , Cell Line , Humans , Image Processing, Computer-Assisted/methods
14.
J Biol Chem ; 289(42): 28870-84, 2014 Oct 17.
Article En | MEDLINE | ID: mdl-25170080

HIV-infected patients are at increased risk of developing atherosclerosis, in part due to an altered high density lipoprotein profile exacerbated by down-modulation and impairment of ATP-binding cassette transporter A1 (ABCA1) activity by the HIV-1 protein Nef. However, the mechanisms of this Nef effect remain unknown. Here, we show that Nef interacts with an endoplasmic reticulum chaperone calnexin, which regulates folding and maturation of glycosylated proteins. Nef disrupted interaction between calnexin and ABCA1 but increased affinity and enhanced interaction of calnexin with HIV-1 gp160. The Nef mutant that did not bind to calnexin did not affect the calnexin-ABCA1 interaction. Interaction with calnexin was essential for functionality of ABCA1, as knockdown of calnexin blocked the ABCA1 exit from the endoplasmic reticulum, reduced ABCA1 abundance, and inhibited cholesterol efflux; the same effects were observed after Nef overexpression. However, the effects of calnexin knockdown and Nef on cholesterol efflux were not additive; in fact, the combined effect of these two factors together did not differ significantly from the effect of calnexin knockdown alone. Interestingly, gp160 and ABCA1 interacted with calnexin differently; although gp160 binding to calnexin was dependent on glycosylation, glycosylation was of little importance for the interaction between ABCA1 and calnexin. Thus, Nef regulates the activity of calnexin to stimulate its interaction with gp160 at the expense of ABCA1. This study identifies a mechanism for Nef-dependent inactivation of ABCA1 and dysregulation of cholesterol metabolism.


ATP Binding Cassette Transporter 1/metabolism , Calnexin/metabolism , Endoplasmic Reticulum/metabolism , HIV Envelope Protein gp160/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Atherosclerosis/metabolism , Cholesterol/metabolism , Glycosylation , HEK293 Cells , HIV-1/metabolism , HeLa Cells , Humans , Protein Binding , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
15.
PLoS One ; 8(11): e78817, 2013.
Article En | MEDLINE | ID: mdl-24265721

Sexual transmission of human immunodeficiency virus type 1 (HIV-1) across the cervicovaginal mucosa in women is influenced by many factors including the microbiota and the presence of underlying inflammation. It is important that potential HIV preventative agents do not alter the mucosal environment in a way that enhances HIV acquisition. We examined the impact of a "live" microbicide on the vaginal mucosal environment in a rhesus macaque repeated vaginal simian-HIV (SHIVSF162P3) challenge model. The microbicide contained a human vaginal Lactobacillus jensenii expressing the HIV-1 entry inhibitor, modified Cyanovirin-N (mCV-N), and henceforth called LB-mCV-N. Macaques were colonized vaginally each week with LB-mCV-N and sampled six days after colonization for culturable bacteria, pH and cervical-vaginal cytokines during the duration of the six-week study. We show that macaques that retained the engineered LB-mCV-N strain in their vaginal microbiota, during SHIV challenge, had lower pH, when colonization levels were higher, and had no evidence of inflammatory cytokines. Indeed, Interleukin-13, a mediator of inflammation, was detected less often in LB-mCV-N colonized macaques than in controls and we found higher levels of Interleukin 1 receptor antagonist (IL-1RA) in LB-mCV-N colonized macaques during the SHIV challenge period. We noted an inverse correlation between levels of mucosal IL-1RA and peak plasma viral load, thus higher IL-1RA correlated with lower viral load in LB-mCV-N treated macaques. These data support the use of LB-mCV-N as a safe "live" microbicide and suggest that lactobacilli themselves may positively impact the mucosal environment.


Bacterial Proteins/biosynthesis , Carrier Proteins/biosynthesis , Lactobacillus/metabolism , Vagina/microbiology , Animals , Anti-Infective Agents, Local , Biomarkers/metabolism , Coinfection , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility/immunology , Female , Hydrogen-Ion Concentration , Inflammation Mediators/metabolism , Macaca mulatta , Menstrual Cycle , Microbiota , Mucous Membrane/metabolism , Mucous Membrane/microbiology , Mucous Membrane/virology , Risk Factors , Simian Acquired Immunodeficiency Syndrome/microbiology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Vagina/virology , Vaginitis/immunology , Vaginitis/metabolism , Vaginitis/microbiology , Vaginitis/virology
16.
BMC Res Notes ; 4: 396, 2011 Oct 11.
Article En | MEDLINE | ID: mdl-21989251

BACKGROUND: Previous studies identified CD147 as the chemotactic receptor on inflammatory leukocytes for extracellular cyclophilins (eCyp). However, CD147 is not known to associate with signal transducing molecules, so other transmembrane proteins, such as proteoglycans, integrins, and CD98, were suggested as receptor or co-receptor for eCyp. CD147 is ubiquitously expressed on many cell types, but relationship between the level of CD147 expression and cellular responses to eCyp has never been analyzed. Given the role of eCyp in pathogenesis of many diseases, it is important to know whether cellular responses to eCyp are regulated at the level of CD147 expression. RESULTS: Here, we manipulated CD147 expression levels on HeLa cells using RNAi and investigated the signalling and chemotactic responses to eCypA. Both Erk activation and chemotaxis correlated with the level of CD147 expression, with cells exhibiting low level expression being practically unresponsive to eCypA. CONCLUSIONS: Our results provide the first demonstration of a chemotactic response of HeLa cells to eCypA, establish a correlation between the level of CD147 expression and the magnitude of cellular responses to eCypA, and indicate that CD147 may be a limiting factor in the receptor complex determining cyclophilin-induced Erk activation and cell migration.

17.
PLoS One ; 5(9)2010 Sep 20.
Article En | MEDLINE | ID: mdl-20862220

The first line of a host's response to various pathogens is triggered by their engagement of cellular pattern recognition receptors (PRRs). Binding of microbial ligands to these receptors leads to the induction of a variety of cellular factors that alter intracellular and extracellular environment and interfere directly or indirectly with the life cycle of the triggering pathogen. Such changes may also affect any coinfecting microbe. Using ligands to Toll-like receptors (TLRs) 5 and 9, we examined their effect on human immunodeficiency virus (HIV)-1 replication in lymphoid tissue ex vivo. We found marked differences in the outcomes of such treatment. While flagellin (TLR5 agonist) treatment enhanced replication of CC chemokine receptor 5 (CCR 5)-tropic and CXC chemokine receptor 4 (CXCR4)-tropic HIV-1, treatment with oligodeoxynucleotide (ODN) M362 (TLR9 agonist) suppressed both viral variants. The differential effects of these TLR ligands on HIV-1 replication correlated with changes in production of CC chemokines CCL3, CCL4, CCL5, and of CXC chemokines CXCL10, and CXCL12 in the ligand-treated HIV-1-infected tissues. The nature and/or magnitude of these changes were dependent on the ligand as well as on the HIV-1 viral strain. Moreover, the tested ligands differed in their ability to induce cellular activation as evaluated by the expression of the cluster of differentiation markers (CD) 25, CD38, CD39, CD69, CD154, and human leukocyte antigen D related (HLA)-DR as well as of a cell proliferation marker, Ki67, and of CCR5. No significant effect of the ligand treatment was observed on apoptosis and cell death/loss in the treated lymphoid tissue ex vivo. Our results suggest that binding of microbial ligands to TLRs is one of the mechanisms that mediate interactions between coinfected microbes and HIV-1 in human tissues. Thus, the engagement of appropriate TLRs by microbial molecules or their mimetic might become a new strategy for HIV therapy or prevention.


Flagellin/pharmacology , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Oligodeoxyribonucleotides/pharmacology , Toll-Like Receptors/agonists , Anti-HIV Agents/immunology , Anti-HIV Agents/pharmacology , Chemokines/immunology , Flagellin/immunology , HIV Infections/drug therapy , HIV-1/drug effects , HIV-1/pathogenicity , HIV-1/physiology , Humans , In Vitro Techniques , Ligands , Oligodeoxyribonucleotides/immunology , Palatine Tonsil/drug effects , Palatine Tonsil/immunology , Palatine Tonsil/virology , Toll-Like Receptors/immunology , Virus Replication/drug effects
18.
BMC Immunol ; 11: 23, 2010 May 18.
Article En | MEDLINE | ID: mdl-20482754

BACKGROUND: At present, the relatively sudden appearance and explosive spread of HIV throughout Africa and around the world beginning in the 1950s has never been adequately explained. Theorizing that this phenomenon may be somehow related to the eradication of smallpox followed by the cessation of vaccinia immunization, we undertook a comparison of HIV-1 susceptibility in the peripheral blood mononuclear cells from subjects immunized with the vaccinia virus to those from vaccinia naive donors. RESULTS: Vaccinia immunization in the preceding 3-6 months resulted in an up to 5-fold reduction in CCR5-tropic but not in CXCR4-tropic HIV-1 replication in the cells from vaccinated subjects. The addition of autologous serum to the cell cultures resulted in enhanced R5 HIV-1 replication in the cells from unvaccinated, but not vaccinated subjects. There were no significant differences in the concentrations of MIP-1alpha, MIP-1beta and RANTES between the cell cultures derived from vaccinated and unvaccinated subjects when measured in culture medium on days 2 and 5 following R5 HIV-1 challenge. DISCUSSION: Since primary HIV-1 infections are caused almost exclusively by the CCR5-tropic HIV-1 strains, our results suggest that prior immunization with vaccinia virus might provide an individual with some degree of protection to subsequent HIV infection and/or progression. The duration of such protection remains to be determined. A differential elaboration of MIP-1alpha, MIP-1beta and RANTES between vaccinated and unvaccinated subjects, following infection, does not appear to be a mechanism in the noted protection.


HIV Infections/immunology , HIV-1/physiology , Receptors, CCR5/immunology , Vaccinia virus/immunology , Viral Vaccines/immunology , Adult , Chemokine CCL3/biosynthesis , Chemokine CCL3/immunology , Chemokine CCL4/biosynthesis , Chemokine CCL4/immunology , Chemokine CCL5/biosynthesis , Chemokine CCL5/immunology , Enzyme-Linked Immunosorbent Assay , Female , HIV Infections/metabolism , Humans , Male , Receptors, CCR5/metabolism , Receptors, CXCR4/immunology , Reverse Transcriptase Polymerase Chain Reaction , Virus Replication/immunology , Young Adult
19.
J Infect Dis ; 201(4): 635-43, 2010 Feb 15.
Article En | MEDLINE | ID: mdl-20085496

BACKGROUND: Human immunodeficiency virus type 1 (HIV-1) and herpes simplex virus type 2 (HSV-2) are responsible for 2 intersecting epidemics in which the disease caused by 1 virus facilitates the transmission of and pathogenesis by the other. Therefore, suppression of one virus infection will affect the other. Acyclovir, a common antiherpetic drug, was shown to directly suppress both viruses in coinfected tissues. However, both antiviral activities of acyclovir are dependent on phosphorylation by the nucleoside kinase activity of coinfecting human herpesviruses. METHODS: We developed acyclovir ProTides, monophosphorylated acyclovir with the phosphate group masked by lipophilic groups to allow efficient cellular uptake, and investigated their antiviral potential in cell lines and in human tissues ex vivo. RESULTS: Acyclovir ProTides suppressed both HIV-1 and HSV-2 at median effective concentrations in the submicromolar range in ex vivo lymphoid and cervicovaginal human tissues and at 3-12 micromol/L in CD4(+) T cells. Acyclovir ProTides retained activity against acyclovir-resistant HSV-2. CONCLUSIONS: Acyclovir ProTides represent a new class of antivirals that suppress both HIV-1 and HSV-2 by directly and independently blocking the key replicative enzymes of both viruses. Further optimization of such compounds may lead to double-targeted antivirals that can prevent viral transmission and treat the 2 synergistic diseases caused by HIV-1 and HSV-2. To our knowledge, the acyclovir ProTides described here represent the first example of acyclic nucleoside monophosphate prodrugs being active against HIV-1.


Acyclovir/analogs & derivatives , Antiviral Agents/pharmacology , HIV-1/drug effects , Herpesvirus 2, Human/drug effects , Prodrugs/pharmacology , Acyclovir/pharmacology , Cell Line , Cervix Uteri/virology , Female , HIV Infections/drug therapy , Herpes Genitalis/drug therapy , Histocytochemistry , Humans , Palatine Tonsil/virology , T-Lymphocytes/virology , Virus Replication/drug effects
20.
Microbes Infect ; 11(14-15): 1106-13, 2009 Dec.
Article En | MEDLINE | ID: mdl-19671446

Critical events of HIV-1 pathogenesis occur in lymphoid tissues where HIV-1 is typically accompanied by infections with other pathogens (HIV co-pathogens). Co-pathogens greatly affect the clinical course of the disease and the transmission of HIV. The apicomplexan parasite Toxoplasma gondii is a common HIV co-pathogen associated with AIDS development. Here, we examined the interaction of T. gondii and HIV in coinfected human lymphoid tissue ex vivo. Both pathogens readily replicate in ex vivo infected blocks of human tonsillar tissue. Surprisingly, we found that live T. gondii preferentially inhibits R5 HIV-1 replication in coinfected tissues. This effect is reproduced by treatment of the tissue blocks with recombinant C-18, a T. gondii-encoded cyclophilin that binds to CCR5. These ex vivo findings raise the possibility that, in addition to being a co-factor in HIV disease, T. gondii may influence the outcome of viral infection by preferentially suppressing R5 variants.


HIV-1/physiology , Palatine Tonsil , Receptors, CCR5/metabolism , Toxoplasma/pathogenicity , Virus Replication/drug effects , Animals , Cyclophilins/genetics , Cyclophilins/metabolism , Cyclophilins/pharmacology , HIV Infections/complications , HIV Infections/virology , HIV-1/metabolism , HIV-1/pathogenicity , Humans , Lymphoid Tissue/parasitology , Lymphoid Tissue/virology , Palatine Tonsil/parasitology , Palatine Tonsil/virology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Toxoplasma/classification , Toxoplasma/metabolism , Toxoplasma/physiology , Toxoplasmosis/complications , Toxoplasmosis/virology
...