Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
2.
JCI Insight ; 7(21)2022 11 08.
Article in English | MEDLINE | ID: mdl-36125890

ABSTRACT

TGF-ß plays a critical role in maintaining immune cells in a resting state by inhibiting cell activation and proliferation. Resting HIV-1 target cells represent the main cellular reservoir after long-term antiretroviral therapy (ART). We hypothesized that releasing cells from TGF-ß-driven signaling would promote latency reversal. To test our hypothesis, we compared HIV-1 latency models with and without TGF-ß and a TGF-ß type 1 receptor inhibitor, galunisertib. We tested the effect of galunisertib in SIV-infected, ART-treated macaques by monitoring SIV-env expression via PET/CT using the 64Cu-DOTA-F(ab')2 p7D3 probe, along with plasma and tissue viral loads (VLs). Exogenous TGF-ß reduced HIV-1 reactivation in U1 and ACH-2 models. Galunisertib increased HIV-1 latency reversal ex vivo and in PBMCs from HIV-1-infected, ART-treated, aviremic donors. In vivo, oral galunisertib promoted increased total standardized uptake values in PET/CT images in gut and lymph nodes of 5 out of 7 aviremic, long-term ART-treated, SIV-infected macaques. This increase correlated with an increase in SIV RNA in the gut. Two of the 7 animals also exhibited increases in plasma VLs. Higher anti-SIV T cell responses and antibody titers were detected after galunisertib treatment. In summary, our data suggest that blocking TGF-ß signaling simultaneously increases retroviral reactivation events and enhances anti-SIV immune responses.


Subject(s)
HIV Infections , HIV-1 , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Simian Acquired Immunodeficiency Syndrome/drug therapy , Copper Radioisotopes/pharmacology , Copper Radioisotopes/therapeutic use , Anti-Retroviral Agents/therapeutic use , Positron Emission Tomography Computed Tomography , Macaca mulatta , Virus Replication , Transforming Growth Factor beta , Immunity
3.
Endocrinology ; 162(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34343260

ABSTRACT

Studies suggest that HIV-1 invades the testis through initial permeation of the blood-testis barrier (BTB). The selectivity of the BTB to antiretroviral drugs makes this site a sanctuary for the virus. Little is known about how HIV-1 crosses the BTB and invades the testis. Herein, we used 2 approaches to examine the underlying mechanism(s) by which HIV-1 permeates the BTB and gains entry into the seminiferous epithelium. First, we examined if recombinant Tat protein was capable of perturbing the BTB and making the barrier leaky, using the primary rat Sertoli cell in vitro model that mimics the BTB in vivo. Second, we used HIV-1-infected Sup-T1 cells to investigate the activity of HIV-1 infection on cocultured Sertoli cells. Using both approaches, we found that the Sertoli cell tight junction permeability barrier was considerably perturbed and that HIV-1 effectively permeates the BTB by inducing actin-, microtubule-, vimentin-, and septin-based cytoskeletal changes in Sertoli cells. These studies suggest that HIV-1 directly perturbs BTB function, potentially through the activity of the Tat protein.


Subject(s)
Blood-Testis Barrier/physiology , Cytoskeleton/metabolism , HIV-1/physiology , Immune Evasion/physiology , Testis/virology , Animals , Animals, Newborn , Blood-Testis Barrier/ultrastructure , Cell Membrane Permeability , Cells, Cultured , Cytoskeleton/ultrastructure , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/pathology , Male , Rats , Rats, Sprague-Dawley , Testis/immunology , Testis/metabolism , Testis/ultrastructure
4.
Sci Transl Med ; 13(607)2021 08 18.
Article in English | MEDLINE | ID: mdl-34408080

ABSTRACT

Anti-HIV broadly neutralizing antibodies (bNAbs) may favor development of antiviral immunity by engaging the immune system during immunotherapy. Targeting integrin α4ß7 with an anti-α4ß7 monoclonal antibody (Rh-α4ß7) affects immune responses in SIV/SHIV-infected macaques. To explore the therapeutic potential of combining bNAbs with α4ß7 integrin blockade, SHIVSF162P3-infected, viremic rhesus macaques were treated with bNAbs only (VRC07-523LS and PGT128 anti-HIV antibodies) or a combination of bNAbs and Rh-α4ß7 or were left untreated as a control. Treatment with bNAbs alone decreased viremia below 200 copies/ml in all macaques, but seven of eight macaques (87.5%) in the bNAbs-only group rebounded within a median of 3 weeks (95% CI: 2 to 9). In contrast, three of six macaques treated with a combination of Rh-α4ß7 and bNAbs (50%) maintained a viremia below 200 copies/ml until the end of the follow-up period; viremia in the other three macaques rebounded within a median of 6 weeks (95% CI: 5 to 11). Thus, there was a modest delay in viral rebound in the macaques treated with the combination antibody therapy compared to bNAbs alone. Our study suggests that α4ß7 integrin blockade may prolong virologic control by bNAbs in SHIVSF162P3-infected macaques.


Subject(s)
HIV Infections , HIV-1 , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Antibodies, Neutralizing , Antibodies, Viral , Broadly Neutralizing Antibodies , HIV Antibodies , HIV Infections/drug therapy , Integrins , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/drug therapy
5.
Sci Adv ; 6(34)2020 08.
Article in English | MEDLINE | ID: mdl-32937372

ABSTRACT

Intravenous administration of anti-α4ß7 monoclonal antibody in macaques decreases simian immunodeficiency virus (SIV) vaginal infection and reduces gut SIV loads. Because of potential side effects of systemic administration, a prophylactic strategy based on mucosal administration of anti-α4ß7 antibody may be safer and more effective. With this in mind, we developed a novel intravaginal formulation consisting of anti-α4ß7 monoclonal antibody-conjugated nanoparticles (NPs) loaded in a 1% hydroxyethylcellulose (HEC) gel (NP-α4ß7 gel). When intravaginally administered as a single dose in a rhesus macaque model, the formulation preferentially bound to CD4+ or CD3+ T cells expressing high levels of α4ß7, and occupied ~40% of α4ß7 expressed by these subsets and ~25% of all cells expressing α4ß7 Blocking of the α4ß7 was restricted to the vaginal tract without any changes detected systemically.


Subject(s)
Nanoparticles , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes , Female , Integrins/metabolism , Macaca mulatta
6.
Sci Rep ; 9(1): 12078, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31427605

ABSTRACT

The establishment of latent infection and poorly characterized viral reservoirs in tissues represent major obstacles to a definitive cure for HIV. Non-human primate (NHP) models of HIV infection are critical to elucidate pathogenic processes and an essential tool to test novel therapeutic strategies. Thus, the availability of novel assays to measure residual viral replication and reservoirs in NHP models may increase their utility in the search for an HIV cure. We developed a tat/rev induced limiting dilution assay to measure the frequency of CD4+ T cells that express multiply-spliced(ms)_SIV RNA in presence and absence of stimulation. We validated the assay using cell lines and cells from blood and lymph nodes of SIV infected macaques. In vitro, SIV/SHIV TILDA detects only cells expressing viral proteins. In SIV/SHIV-infected macaques, CD4+ T cells that express msSIV/SHIV RNA (TILDA data) were detected also in the setting of very low/undetectable viremia. TILDA data were significantly higher after stimulation and correlated with plasma viral load (pVL). Interestingly, TILDA data from early cART initiation correlated with peak and AUC pVL post-cART interruption. In summary, we developed an assay that may be useful in characterizing viral reservoirs and determining the effect of HIV interventions in NHP models.


Subject(s)
HIV Infections/genetics , HIV-1/genetics , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Immunodeficiency Virus/genetics , Animals , CD4-Positive T-Lymphocytes/virology , Disease Models, Animal , HIV Infections/pathology , HIV Infections/virology , HIV-1/pathogenicity , Humans , Macaca mulatta/virology , Macrophages/metabolism , Macrophages/virology , Primates/virology , RNA, Viral/genetics , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , Viral Load/genetics , Virus Latency/genetics , Virus Replication/genetics , rev Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics
7.
PLoS Pathog ; 15(5): e1007776, 2019 05.
Article in English | MEDLINE | ID: mdl-31083697

ABSTRACT

VRC01 protects macaques from vaginal SHIV infection after a single high-dose challenge. Infusion of a simianized anti-α4ß7 mAb (Rh-α4ß7) just prior to, and during repeated vaginal exposures to SIVmac251 partially protected macaques from vaginal SIV infection and rescued CD4+ T cells. To investigate the impact of combining VRC01 and Rh-α4ß7 on SHIV infection, 3 groups of macaques were treated with a suboptimal dosing of VRC01 alone or in combination with Rh-α4ß7 or with control antibodies prior to the initiation of weekly vaginal exposures to a high dose (1000 TCID50) of SHIVAD8-EO. The combination Rh-α4ß7-VRC01 significantly delayed SHIVAD8-EO vaginal infection. Following infection, VRC01-Rh-α4ß7-treated macaques maintained higher CD4+ T cell counts and exhibited lower rectal SIV-DNA loads compared to controls. Interestingly, VRC01-Rh-α4ß7-treated macaques had fewer IL-17-producing cells in the blood and the gut during the acute phase of infection. Moreover, higher T cell responses to the V2-loop of the SHIVAD8-EO envelope in the VRC01-Rh-α4ß7 group inversely correlated with set point viremia. The combination of suboptimal amounts of VRC01 and Rh-α4ß7 delayed infection, altered antiviral immune responses and minimized CD4+ T cell loss. Further exploration of the effect of combining bNAbs with Rh-α4ß7 on SIV/HIV infection and antiviral immune responses is warranted and may lead to novel preventive and therapeutic strategies.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/pharmacology , Integrins/antagonists & inhibitors , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/drug effects , Vagina/drug effects , Viremia/prevention & control , Animals , Antibodies, Viral/immunology , Broadly Neutralizing Antibodies , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Drug Therapy, Combination , Female , HIV Antibodies , Integrins/immunology , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Vagina/immunology , Vagina/virology , Viremia/immunology , Viremia/virology
8.
J Immunol ; 200(2): 810-820, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29196458

ABSTRACT

Infusion of a simianized anti-α4ß7 mAb (Rh-α4ß7) just before and following SIV infection protected rhesus macaques from developing AIDS and partially from vaginal SIV acquisition. Recently, short-term treatment with Rh-α4ß7 in combination with cART was found to lead to prolonged viral suppression after withdrawal of all therapeutic interventions. The humanized form of Rh-α4ß7, vedolizumab, is a highly effective treatment for inflammatory bowel disease. To clarify the mechanism of action of Rh-α4ß7, naive macaques were infused with Rh-α4ß7 and sampled in blood and tissues before and after treatment to monitor several immune cell subsets. In blood, Rh-α4ß7 increased the CD4+ and CD8+ T cell counts, but not B cell counts, and preferentially increased CCR6+ subsets while decreasing CD103+ and CD69+ lymphocytes. In mucosal tissues, surprisingly, Rh-α4ß7 did not impact integrin α4+ cells, but decreased the frequencies of CCR6+ and CD69+ CD4+ T cells and, in the gut, Rh-α4ß7 transiently decreased the frequency of memory and IgA+ B cells. In summary, even in the absence of inflammation, Rh-α4ß7 impacted selected immune cell subsets in different tissues. These data provide new insights into the mechanisms by which Rh-α4ß7 may mediate its effect in SIV-infected macaques with implications for understanding the effect of treatment with vedolizumab in patients with inflammatory bowel disease.


Subject(s)
Integrins/antagonists & inhibitors , Lymphocyte Count , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Mucous Membrane/immunology , Mucous Membrane/metabolism , Receptors, CCR6/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Macaca mulatta , Organ Specificity/immunology
9.
Front Microbiol ; 8: 2342, 2017.
Article in English | MEDLINE | ID: mdl-29259582

ABSTRACT

Herpes simplex virus 1 and 2 (HSV-1/2) similarly initiate infection in mucosal epithelia and establish lifelong neuronal latency. Anogenital HSV-2 infection augments the risk for sexual human immunodeficiency virus (HIV) transmission and is associated with higher HIV viral loads. However, whether oral HSV-1 infection contributes to oral HIV susceptibility, viremia, or oral complications of HIV infection is unknown. Appropriate non-human primate (NHP) models would facilitate this investigation, yet there are no published studies of HSV-1/SIV co-infection in NHPs. Thus, we performed a pilot study for an oral HSV-1 infection model in SIV-infected rhesus macaques to describe the feasibility of the modeling and resultant immunological changes. Three SIV-infected, clinically healthy macaques became HSV-1-infected by inoculation with 4 × 108 pfu HSV-1 McKrae on buccal, tongue, gingiva, and tonsils after gentle abrasion. HSV-1 DNA was shed in oral swabs for up to 21 days, and shedding recurred in association with intra-oral lesions after periods of no shedding during 56 days of follow up. HSV-1 DNA was detected in explant cultures of trigeminal ganglia collected at euthanasia on day 56. In the macaque with lowest baseline SIV viremia, SIV plasma RNA increased following HSV-1 infection. One macaque exhibited an acute pro-inflammatory response, and all three animals experienced T cell activation and mobilization in blood. However, T cell and antibody responses to HSV-1 were low and atypical. Through rigorous assessesments, this study finds that the virulent HSV-1 strain McKrae resulted in a low level HSV-1 infection that elicited modest immune responses and transiently modulated SIV infection.

10.
J Med Primatol ; 46(4): 121-128, 2017 08.
Article in English | MEDLINE | ID: mdl-28748667

ABSTRACT

BACKGROUND: Although HSV-2 is the major cause of genital lesions, HSV-1 accounts for half of new cases in developed countries. METHODS: Three healthy SHIV-SF162P3-infected Indian rhesus macaques were inoculated with 4×108 pfu of HSV-1 twice, with the second inoculation performed after the vaginal mucosa was gently abraded with a cytobrush. RESULTS: HSV-1 DNA was detected in vaginal swabs 5 days after the second but not the first inoculation in all three macaques. An increase in inflammatory cytokines was detected in the vaginal fluids of the animals with no or intermittent shedding. Higher frequency of blood α4 ß7high CD4+ T cells was measured in the animals with consistent and intermitted shedding, while a decrease in the frequency of CD69+ CD4+ T cells was present in all animals. CONCLUSIONS: This macaque model of genital HSV-1 could be useful to study the impact of the growing epidemic of genital HSV-1 on HIV infection.


Subject(s)
Disease Models, Animal , HIV Infections/virology , Herpes Genitalis/virology , Herpesvirus 1, Human/physiology , Macaca mulatta , Animals , Female , Viral Load
11.
PLoS One ; 11(9): e0161730, 2016.
Article in English | MEDLINE | ID: mdl-27603520

ABSTRACT

Myeloid dendritic cells (mDCs) contribute to both HIV pathogenesis and elicitation of antiviral immunity. Understanding how mDC responses to stimuli shape HIV infection outcomes will inform HIV prevention and treatment strategies. The long double-stranded RNA (dsRNA) viral mimic, polyinosinic polycytidylic acid (polyIC, PIC) potently stimulates DCs to focus Th1 responses, triggers direct antiviral activity in vitro, and boosts anti-HIV responses in vivo. Stabilized polyICLC (PICLC) is being developed for vaccine adjuvant applications in humans, making it critical to understand how mDC sensing of PICLC influences HIV infection. Using the monocyte-derived DC (moDC) model, we sought to describe how PICLC (vs. other dsRNAs) impacts HIV infection within DCs and DC-T cell mixtures. We extended this work to in vivo macaque rectal transmission studies by administering PICLC with or before rectal SIVmac239 (SIVwt) or SIVmac239ΔNef (SIVΔNef) challenge. Like PIC, PICLC activated DCs and T cells, increased expression of α4ß7 and CD169, and induced type I IFN responses in vitro. The type of dsRNA and timing of dsRNA exposure differentially impacted in vitro DC-driven HIV infection. Rectal PICLC treatment similarly induced DC and T cell activation and pro- and anti-HIV factors locally and systemically. Importantly, this did not enhance SIV transmission in vivo. Instead, SIV acquisition was marginally reduced after a single high dose challenge. Interestingly, in the PICLC-treated, SIVΔNef-infected animals, SIVΔNef viremia was higher, in line with the importance of DC and T cell activation in SIVΔNef replication. In the right combination anti-HIV strategy, PICLC has the potential to limit HIV infection and boost HIV immunity.


Subject(s)
Carboxymethylcellulose Sodium/analogs & derivatives , HIV Infections/therapy , Lymphocyte Activation/immunology , Poly I-C/genetics , Polylysine/analogs & derivatives , RNA, Double-Stranded/genetics , Animals , Carboxymethylcellulose Sodium/administration & dosage , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/virology , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/transmission , HIV-1/genetics , Humans , Interferon Type I/genetics , Lymphocyte Activation/genetics , Macaca/immunology , Macaca/virology , Monocytes/drug effects , Monocytes/immunology , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/virology , Poly I-C/administration & dosage , Polylysine/administration & dosage , Polylysine/genetics , RNA, Double-Stranded/administration & dosage , Simian Immunodeficiency Virus/genetics , Th1 Cells/drug effects , Th1 Cells/immunology
13.
PLoS Pathog ; 12(6): e1005720, 2016 06.
Article in English | MEDLINE | ID: mdl-27348748

ABSTRACT

Mucosal HIV-1 transmission is inefficient. However, certain viral and host characteristics may play a role in facilitating HIV acquisition and systemic expansion. Cells expressing high levels of integrin α4ß7 have been implicated in favoring the transmission process and the infusion of an anti-α4ß7 mAb (RM-Act-1) prior to, and during a repeated low-dose vaginal challenge (RLDC) regimen with SIVmac251 reduced SIV acquisition and protected the gut-associated lymphoid tissues (GALT) in the macaques that acquired SIV. α4ß7 expression is required for lymphocyte trafficking to the gut lamina propria and gut inductive sites. Several therapeutic strategies that target α4ß7 have been shown to be effective in treating inflammatory conditions of the intestine, such as inflammatory bowel disease (IBD). To determine if blocking α4ß7 with ELN, an orally available anti-α4 small molecule, would inhibit SHIV-SF162P3 acquisition, we tested its ability to block MAdCAM-1 (α4ß7 natural ligand) and HIV-gp120 binding in vitro. We studied the pharmacokinetic profile of ELN after oral and vaginal delivery in macaques. Twenty-six macaques were divided into 3 groups: 9 animals were treated with ELN orally, 9 orally and vaginally and 8 were used as controls. All animals were challenged intra-vaginally with SHIV-SF162P3 using the RLDC regimen. We found that ELN did not protect macaques from SHIV acquisition although it reduced the SHIV-induced inflammatory status during the acute phase of infection. Notably, integrins can exist in different activation states and, comparing the effect of ELN and the anti-α4ß7 mAb RM-Act-1 that reduced susceptibility to SIV infection, we determined that ELN induces the active conformation of α4ß7, while RM-Act-1 inhibits its activation through an allosteric mechanism. These results suggest that inhibition of α4ß7 activation may be necessary to reduce susceptibility to SIV/SHIV infection and highlight the complexity of anti-integrins therapeutic approach in HIV as well as in IBD and other autoimmune diseases.


Subject(s)
Antiviral Agents/pharmacology , Immunoglobulins/metabolism , Integrins/antagonists & inhibitors , Mucoproteins/metabolism , Simian Acquired Immunodeficiency Syndrome/transmission , Animals , Antibodies, Monoclonal, Humanized/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Cell Adhesion Molecules , Cells, Cultured , Female , Flow Cytometry , Humans , Macaca mulatta , Mucous Membrane/virology , Simian Immunodeficiency Virus , Vagina/virology , Viral Load
14.
J Immunol ; 194(5): 2415-23, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25624458

ABSTRACT

The tissue microenvironment shapes the characteristics and functions of dendritic cells (DCs), which are important players in HIV infection and dissemination. Notably, DCs in the gut have the daunting task of orchestrating the balance between immune response and tolerance. They produce retinoic acid (RA), which imprints a gut-homing phenotype and influences surrounding DCs. To investigate how the gut microenvironment impacts the ability of DCs to drive HIV infection, we conditioned human immature monocyte-derived DCs (moDCs) with RA (RA-DCs), before pulsing them with HIV and mixing them with autologous T cells. RA-DCs showed a semimature, mucosal-like phenotype and released higher amounts of TGF-ß1 and CCL2. Using flow cytometry, Western blot, and microscopy, we determined that moDCs express the cell adhesion molecule mucosal vascular addressin cell adhesion molecule-1 (MAdCAM-1) and that RA increases its expression. MAdCAM-1 was also detected on a small population of DCs in rhesus macaque (Macaca mulata) mesenteric lymph node. RA-DCs formed more DC-T cell conjugates and promoted significantly higher HIV replication in DC-T cell mixtures compared with moDCs. This correlated with the increase in MAdCAM-1 expression. Blocking MAdCAM-1 partially inhibited the enhanced HIV replication. In summary, RA influences DC phenotype, increasing their ability to exacerbate HIV infection. We describe a previously unknown mechanism that may contribute to rapid HIV spread in the gut, a major site of HIV replication after mucosal exposure.


Subject(s)
Dendritic Cells/drug effects , HIV Infections/immunology , HIV-1/immunology , Intestinal Mucosa/drug effects , T-Lymphocytes/drug effects , Tretinoin/pharmacology , Animals , Cell Adhesion Molecules , Chemokine CCL2/biosynthesis , Chemokine CCL2/metabolism , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Female , Gene Expression , HIV Infections/genetics , HIV Infections/pathology , HIV Infections/virology , Humans , Immunoglobulins/genetics , Immunoglobulins/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymph Nodes/virology , Macaca mulatta , Mucoproteins/genetics , Mucoproteins/immunology , Phenotype , Protein Isoforms/genetics , Protein Isoforms/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/virology , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/metabolism , Tretinoin/immunology , Virus Replication
15.
AIDS Res Hum Retroviruses ; 30(11): 1117-24, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25354024

ABSTRACT

Epidemiological studies suggest that prevalent herpes simplex virus type 2 (HSV-2) infection increases the risk of HIV acquisition, underscoring the need to develop coinfection models to evaluate promising prevention strategies. We previously established a single high-dose vaginal coinfection model of simian human immunodeficiency virus (SHIV)/HSV-2 in Depo-Provera (DP)-treated macaques. However, this model does not appropriately mimic women's exposure. Repeated limiting dose SHIV challenge models are now used routinely to test prevention strategies, yet, at present, there are no reports of a repeated limiting dose cochallenge model in which to evaluate products targeting HIV and HSV-2. Herein, we show that 20 weekly cochallenges with 2-50 TCID50 simian human immunodeficiency virus reverse transcriptase (SHIV-RT) and 10(7) pfu HSV-2 results in infection with both viruses (4/6 SHIV-RT, 6/6 HSV-2). The frequency and level of vaginal HSV-2 shedding were significantly greater in the repeated exposure model compared to the single high-dose model (p<0.0001). We used this new model to test the Council's on-demand microbicide gel, MZC, which is active against SHIV-RT in DP-treated macaques and HSV-2 and human papillomavirus (HPV) in mice. While MZC reduced SHIV and HSV-2 infections in our repeated limiting dose model when cochallenging 8 h after each gel application, a barrier effect of carrageenan (CG) that was not seen in DP-treated animals precluded evaluation of the significance of the antiviral activity of MZC. Both MZC and CG significantly (p<0.0001) reduced the frequency and level of vaginal HSV-2 shedding compared to no gel treatment. This validates the use of this repeated limiting dose cochallenge model for testing products targeting HIV and HSV-2.


Subject(s)
Anti-Infective Agents/administration & dosage , Coinfection/virology , HIV Reverse Transcriptase/metabolism , Herpes Genitalis/virology , Herpesvirus 2, Human/growth & development , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/growth & development , Animals , Contraceptive Agents, Female/administration & dosage , Disease Models, Animal , Female , Herpes Genitalis/complications , Macaca mulatta , Medroxyprogesterone Acetate/administration & dosage , Simian Acquired Immunodeficiency Syndrome/complications , Simian Immunodeficiency Virus/enzymology , Treatment Outcome , Vagina/virology , Vaginal Creams, Foams, and Jellies/administration & dosage , Virus Shedding
16.
J Acquir Immune Defic Syndr ; 61(4): 425-35, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22820802

ABSTRACT

BACKGROUND: The goal of antiretroviral therapy (ART) is to suppress virus replication to limit immune system damage. Some have proposed combining ART with immune therapies to boost antiviral immunity. For this to be successful, ART must not impair physiological immune function. METHODS: We studied the impact of ART (tenofovir and emtricitabine) on systemic and mucosal immunity in uninfected and simian immunodeficiency (SIV)-infected Chinese rhesus macaques. Subcutaneous ART was initiated 2 weeks after tonsillar inoculation with SIVmac239. RESULTS: There was no evidence of immune dysregulation as a result of ART in either infected or uninfected animals. Early virus-induced alterations in circulating immune cell populations (decreased central memory T cells and myeloid dendritic cells) were detected, but normalized shortly after ART initiation. ART-treated animals showed marginal SIV-specific T-cell responses during treatment, which increased after ART discontinuation. Elevated expression of CXCL10 in oral, rectal, and blood samples and APOBEC3G mRNA in oral and rectal tissues was observed during acute infection and was down regulated after starting ART. ART did not impact the ability of the animals to respond to tonsillar application of polyICLC with increased CXCL10 expression in oral fluids and CD80 expression on blood myeloid dendritic cells. CONCLUSION: Early initiation of ART prevented virus-induced damage and did not impede mucosal or systemic immune functions.


Subject(s)
Adenine/analogs & derivatives , Anti-Retroviral Agents/administration & dosage , Antiretroviral Therapy, Highly Active/methods , Deoxycytidine/analogs & derivatives , Immunity, Mucosal , Organophosphonates/administration & dosage , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Adenine/administration & dosage , Animals , Deoxycytidine/administration & dosage , Emtricitabine , Immunophenotyping , Injections, Subcutaneous , Macaca mulatta , Male , T-Lymphocyte Subsets/immunology , Tenofovir
17.
PLoS Pathog ; 7(6): e1002109, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21738472

ABSTRACT

Herpes simplex virus type 2 (HSV-2) increases the risk of HIV-1 infection and, although several reports describe the interaction between these two viruses, the exact mechanism for this increased susceptibility remains unclear. Dendritic cells (DCs) at the site of entry of HSV-2 and HIV-1 contribute to viral spread in the mucosa. Specialized DCs present in the gut-associated lymphoid tissues produce retinoic acid (RA), an important immunomodulator, able to influence HIV-1 replication and a key mediator of integrin α4ß7 on lymphocytes. α4ß7 can be engaged by HIV-1 on the cell-surface and CD4⁺ T cells expressing high levels of this integrin (α4ß7 (high)) are particularly susceptible to HIV-1 infection. Herein we provide in-vivo data in macaques showing an increased percentage of α4ß7 (high) CD4⁺ T cells in rectal mucosa, iliac lymph nodes and blood within 6 days of rectal exposure to live (n = 11), but not UV-treated (n = 8), HSV-2. We found that CD11c⁺ DCs are a major target of HSV-2 infection in in-vitro exposed PBMCs. We determined that immature monocyte-derived DCs (moDCs) express aldehyde dehydrogenase ALDH1A1, an enzyme essential for RA production, which increases upon HSV-2 infection. Moreover, HSV-2-infected moDCs significantly increase α4ß7 expression on CD4⁺ T lymphocytes and HIV-1 infection in DC-T cell mixtures in a RA-dependent manner. Thus, we propose that HSV-2 modulates its microenviroment, influencing DC function, increasing RA production capability and amplifying a α4ß7 (high)CD4⁺ T cells. These factors may play a role in increasing the susceptibility to HIV-1.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Coinfection/virology , Dendritic Cells/pathology , HIV Infections/virology , HIV-1 , Herpes Genitalis/virology , Herpesvirus 2, Human , Animals , CD11c Antigen/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Dendritic Cells/virology , Disease Susceptibility/pathology , HIV Infections/complications , Herpes Genitalis/complications , Integrins/metabolism , Lymph Nodes , Macaca , Mucous Membrane/pathology , Mucous Membrane/virology , Rectum , Tretinoin/metabolism
18.
J Acquir Immune Defic Syndr ; 56(3): 204-12, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21084994

ABSTRACT

Dendritic cells (DCs) efficiently transfer captured (trans) or de novo-produced (cis) virus to CD4 T cells. Using monocyte-derived DCs, we evaluated entry inhibitors targeting HIV envelope (BMS-C, T-1249) or CCR5 (CMPD167) for their potency to prevent DC infection, DC-driven infection in T cells in trans and cis, and direct infection of DC-T-cell mixtures. Immature DC-T-cell cultures with distinct mechanisms of viral transfer yielded similar levels of infection and produced more proviral DNA compared with matched mature DC-T-cell cultures or infected immature DCs. Although all compounds completely blocked HIV replication, 16 times more of each inhibitor (250 vs 15.6 nM) was required to prevent low-level infection of DCs compared with the productive DC-T-cell cocultures. Across all cell systems tested, BMS-C blocked infection most potently. BMS-C was significantly more effective than CMPD167 at preventing DC infection. In fact, low doses of CMPD167 significantly enhanced DC infection. Elevated levels of CCL4 were observed when immature DCs were cultured with CMPD167. Viral entry inhibitors did not interfere with Candida albicans-specific DC cytokine/chemokine responses. These findings indicate that an envelope-binding small molecule is a promising tool for topical microbicide design to prevent the infection of early targets needed to establish and disseminate HIV infection.


Subject(s)
Anti-HIV Agents/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/virology , HIV Fusion Inhibitors/pharmacology , HIV Infections/virology , HIV/drug effects , HIV/growth & development , Cells, Cultured , Coculture Techniques , HIV Envelope Protein gp41/pharmacology , Humans , Peptide Fragments/pharmacology , Pyrazoles/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/virology , Valine/analogs & derivatives , Valine/pharmacology , Virus Replication/drug effects
19.
J Virol ; 79(3): 1533-42, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15650179

ABSTRACT

The zinc finger motifs in retroviral nucleocapsid (NC) proteins are essential for viral replication. Disruption of these Cys-X2-Cys-X4-His-X4-Cys zinc-binding structures eliminates infectivity. To determine if N-ethylmaleimide (NEM) can inactivate human immunodeficiency virus type 1 (HIV-1) or simian immunodeficiency virus (SIV) preparations by alkylating cysteines of NC zinc fingers, we treated infectious virus with NEM and evaluated inactivation of infectivity in cell-based assays. Inactivation was rapid and proportional to the NEM concentration. NEM treatment of HIV-1 or SIV resulted in extensive covalent modification of NC and other internal virion proteins. In contrast, viral envelope glycoproteins, in which the cysteines are disulfide bonded, remained intact and functional, as assayed by high-performance liquid chromatography, fusion-from-without analyses, and dendritic cell capture. Quantitative PCR assays for reverse transcription intermediates showed that NEM and 2,2'-dipyridyl disulfide (aldrithiol-2), a reagent which inactivates retroviruses through oxidation of cysteines in internal virion proteins such as NC, blocked HIV-1 reverse transcription prior to the formation of minus-strand strong-stop products. However, the reverse transcriptase from NEM-treated virions remained active in exogenous template assays, consistent with a role for NC in reverse transcription. Since disruption of NC zinc finger structures by NEM blocks early postentry steps in the retroviral infection cycle, virus preparations with modified NC proteins may be useful as vaccine immunogens and probes of the role of NC in viral replication.


Subject(s)
Ethylmaleimide/pharmacology , Gene Products, env/metabolism , HIV-1/drug effects , HIV-1/pathogenicity , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/pathogenicity , Cell Line , Dendritic Cells/cytology , Dendritic Cells/virology , HeLa Cells , Humans , Kinetics , Nucleocapsid/metabolism , Zinc Fingers
20.
Aviat Space Environ Med ; 75(9): 760-3, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15460626

ABSTRACT

INTRODUCTION: Spaceflight is associated with a shift of interstitial fluid toward the skin of the head and a fluid loss that can decrease superficial tissue thickness (TT). To collect the physiological data needed to develop techniques for monitoring the hydration status of astronauts during spaceflight, we investigated the changes in TT induced by a 12-h nil-by-mouth period and a 30-min period of +2 Gz hypergravity measured at forehead and tibia. METHODS: There were 16 male volunteers who were twice subjected to 30 min of +2 Gz acceleration in a human centrifuge, once following an oral fluid load of 250 ml x h(-1) (procedure A), and once following a nil-by-mouth period of 12 h (procedure B). The TT at the forehead (TT-f) and tibia (TT-t) was measured before (t0), after 10 min (t10), and after 30 min (t30) of hypergravity using a miniature 10 MHz A-mode ultrasound device. The bodyweight, hematocrit (HCT), and plasma viscosity (PVS) were measured at tO and t0. RESULTS: The nil-by-mouth period induced a significant weight loss and increase in HCT and PVS. A significant increase was noted in TT-t, but not in TT-f (p < 0.05). Following both procedures, exposure to hypergravity produced declines in TT-f at t10 and t30 compared with baseline (p < 0.05), whereas TT-t was not influenced. CONCLUSIONS: Changes in interstitial fluid load of superficial tissues can be tracked by A-mode ultrasonography. Fluid loss-induced changes are best detected at the tibia, hypergravity-induced changes at the forehead.


Subject(s)
Dehydration/diagnostic imaging , Extracellular Fluid/diagnostic imaging , Hypergravity/adverse effects , Skin Physiological Phenomena , Adult , Dehydration/etiology , Elasticity , Extracellular Fluid/physiology , Fluid Shifts/physiology , Humans , Male , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL