Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
PLoS Pathog ; 12(9): e1005885, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27658293

RESUMEN

Currently, there are mounting data suggesting that HIV-1 acquisition in women can be affected by the use of certain hormonal contraceptives. However, in non-human primate models, endogenous or exogenous progestin-dominant states are shown to increase acquisition. To gain mechanistic insights into this increased acquisition, we studied how mucosal barrier function and CD4+ T-cell and CD68+ macrophage density and localization changed in the presence of natural progestins or after injection with high-dose DMPA. The presence of natural or injected progestins increased virus penetration of the columnar epithelium and the infiltration of susceptible cells into a thinned squamous epithelium of the vaginal vault, increasing the likelihood of potential virus interactions with target cells. These data suggest that increasing either endogenous or exogenous progestin can alter female reproductive tract barrier properties and provide plausible mechanisms for increased HIV-1 acquisition risk in the presence of increased progestin levels.


Asunto(s)
Interacciones Huésped-Patógeno/efectos de los fármacos , Macrófagos/efectos de los fármacos , Membrana Mucosa/efectos de los fármacos , Progestinas/uso terapéutico , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Vagina/efectos de los fármacos , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Cuello del Útero/efectos de los fármacos , Cuello del Útero/inmunología , Cuello del Útero/metabolismo , Cuello del Útero/virología , Preparaciones de Acción Retardada , Femenino , Inyecciones Intramusculares , Activación de Linfocitos/efectos de los fármacos , Macaca mulatta , Macaca nemestrina , Activación de Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/virología , Acetato de Medroxiprogesterona/administración & dosificación , Acetato de Medroxiprogesterona/uso terapéutico , Ciclo Menstrual , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Membrana Mucosa/virología , Progestinas/administración & dosificación , Progestinas/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Virus de la Inmunodeficiencia de los Simios/fisiología , Vagina/inmunología , Vagina/metabolismo , Vagina/virología , Internalización del Virus/efectos de los fármacos
2.
J Med Primatol ; 46(5): 218-227, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28488731

RESUMEN

BACKGROUND: Rectal STI coinfection models enhance the understanding of rectal HIV transmission risk factors. MATERIALS AND METHODS: Rhesus macaques (n=9) were exposed to one of three rectal Chlamydia trachomatis (CT) challenges: C. trachomatis L2 (CT-L2 ); C. trachomatis serovar E (CT-E), followed by CT-L2 ; or CT-E, treatment/clearance, then CT-L2 . Infections were monitored by PCR. Weekly blood and rectal secretion/lavage samples were collected for cytokine analyzes and/or epithelial sloughing, occult, and overt blood determinations. RESULTS: Chlamydial infections were successfully established in each animal, with varying degrees of persistence. Mucosal IL-1beta was upregulated in animals consecutively infected with CT-E then CT-L2 (P=.05). Epithelial sloughing was also significantly increased post-infection in this group (P=.0003). CONCLUSIONS: This study demonstrates successful rectal infection of rhesus macaques with CT-E and CT-L2 and describes measures of assessing rectal inflammation and pathology. Different infection strategies yield varying inflammatory and pathologic outcomes, providing well-described models for future SIV/SHIV susceptibility studies.


Asunto(s)
Infecciones por Chlamydia/complicaciones , Chlamydia trachomatis/fisiología , Modelos Animales de Enfermedad , Infecciones por VIH/complicaciones , Macaca mulatta , Enfermedades de Transmisión Sexual/complicaciones , Animales , Infecciones por Chlamydia/sangre , Infecciones por Chlamydia/patología , Coinfección , Femenino , Infecciones por VIH/sangre , Infecciones por VIH/virología , Recto , Serogrupo , Enfermedades de Transmisión Sexual/sangre , Enfermedades de Transmisión Sexual/microbiología , Enfermedades de Transmisión Sexual/virología , Virus de la Inmunodeficiencia de los Simios/fisiología
3.
Virol J ; 12: 90, 2015 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-26070461

RESUMEN

There are few nonhuman primate models of enhanced HIV susceptibility. Such models can improve comprehension of HIV acquisition risk factors and provide rigorous testing platforms for preclinical prevention strategies. This paper reviews past, current, and proposed research on macaque HIV acquisition risk models and identifies areas where modeling is significantly lacking. We compare different experimental approaches and provide practical considerations for designing macaque susceptibility studies. Modifiable (mucosal and systemic coinfections, hormonal contraception, and rectal lubricants) and non-modifiable (hormonal fluctuations) risk factors are highlighted. Risk acquisition models via vaginal, rectal, and penile challenge routes are discussed. There is no consensus on the best statistical model for evaluating increased susceptibility, and additional research is required. The use of enhanced susceptibility macaque models would benefit multiple facets of the HIV research field, including basic acquisition and pathogenesis studies as well as the vaccine and other biomedical preventions pipeline.


Asunto(s)
Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Infecciones por VIH/inmunología , Macaca , Animales , Modelos Estadísticos
4.
J Med Primatol ; 44(5): 286-95, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26238265

RESUMEN

BACKGROUND: Depot-medroxyprogesterone acetate (DMPA) has been associated in some studies with increased HIV susceptibility in women. We used a pigtail macaque model to document the effects of repeated DMPA treatments and their potential contribution to increased SHIV susceptibility. METHODS: Nine pigtails were administered 2.5, 1.5, or 0.5 mg/kg DMPA in study weeks one and four. Menstrual cycling, vaginal epithelial thickness, and other SHIV susceptibility factors were monitored for a mean of 24 study weeks. RESULTS: All DMPA treatments suppressed menstrual cycling and increased vaginal pH. The vaginal epithelium thinned naturally during baseline menstrual cycles (from mean of 351 to 161 µm in late-luteal phase). Following DMPA, the non-nucleated layer was temporarily absent. Two weeks post-second DMPA injection, mean epithelial thickness was 53, 45, and 167 µm for the descending doses, respectively. CONCLUSIONS: All animals showed temporal vaginal epithelial thinning with loss of the non-nucleated layer, and vaginal pH changes post-DMPA injections.


Asunto(s)
Anticonceptivos Femeninos/administración & dosificación , Macaca nemestrina , Acetato de Medroxiprogesterona/administración & dosificación , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/fisiología , Vagina/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/virología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Concentración de Iones de Hidrógeno , Ciclo Menstrual/efectos de los fármacos , Membrana Mucosa/efectos de los fármacos , Progesterona/metabolismo , Vagina/anatomía & histología
5.
J Infect Dis ; 210(8): 1239-47, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24755433

RESUMEN

BACKGROUND: Sexually transmitted infections (STIs) are associated with an increased risk of human immunodeficiency virus (HIV) infection, but their biological effect on HIV susceptibility is not fully understood. METHODS: Female pig-tailed macaques inoculated with Chlamydia trachomatis and Trichomonas vaginalis (n = 9) or medium (controls; n = 7) were repeatedly challenged intravaginally with SHIVSF162p3. Virus levels were evaluated by real-time polymerase chain reaction, plasma and genital cytokine levels by Luminex assays, and STI clinical signs by colposcopy. RESULTS: Simian/HIV (SHIV) susceptibility was enhanced in STI-positive macaques (P = .04, by the log-rank test; relative risk, 2.5 [95% confidence interval, 1.1-5.6]). All STI-positive macaques were SHIV infected, whereas 3 controls (43%) remained uninfected. Moreover, relative to STI-negative animals, SHIV infections occurred earlier in the menstrual cycle in STI-positive macaques (P = .01, by the Wilcoxon test). Levels of inflammatory cytokines (interferon γ, interleukin 6, and granulocyte colony-stimulating factor [G-CSF]) were higher in STI-positive macaques during STI inoculation and SHIV exposure periods (P ≤ .05, by the Wilcoxon test). CONCLUSIONS: C. trachomatis and T. vaginalis infection increase the susceptibility to SHIV, likely because of prolonged genital tract inflammation. These novel data demonstrate a biological link between these nonulcerative STIs and the risk of SHIV infection, supporting epidemiological associations of HIV and STIs. This study establishes a macaque model for studies of high-risk HIV transmission and prevention.


Asunto(s)
Infecciones por Chlamydia/complicaciones , Chlamydia trachomatis , Coinfección/inmunología , Virus de la Inmunodeficiencia de los Simios/fisiología , Vaginitis por Trichomonas/complicaciones , Trichomonas vaginalis , Animales , Cuello del Útero/microbiología , Cuello del Útero/parasitología , Cuello del Útero/patología , Colposcopía , Femenino , Macaca nemestrina , Factores de Riesgo , Enfermedades de Transmisión Sexual/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Simio/transmisión , Síndrome de Inmunodeficiencia Adquirida del Simio/virología
6.
AIDS Res Hum Retroviruses ; 31(11): 1166-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26165400

RESUMEN

Vaginal SHIVSF162P3 acquisition in pigtail macaques (Macaca nemestrina) is dependent on time point during the menstrual cycle. Susceptibility is higher around menstruation and lower at ovulation in mid cycle. This complicates the design of repeat low-dose (RLD) SHIV exposure studies because virus challenges given during low susceptibility periods have lower chances to infect. To account for fluctuating susceptibility, we analyzed menstrual cycles rather than exposures until infection following virus challenges. We first reanalyzed infection data of 41 macaques receiving placebo or no treatment during once (n=18) or twice (n=23) weekly virus exposures. The same number of cycles was required for infection with either challenge frequency, while it took a median four or six challenges for once or twice weekly exposures, respectively. More virus challenges to infection likely reflect frequent unsuccessful exposures in frequently exposed animals. When reanalyzing two previously reported biomedical HIV intervention studies, we found 1% tenofovir gel was 74% or 86% efficacious based on cycles or exposures (p=0.019 or p=0.003, respectively, Fisher's exact test), while 1% raltegravir gel was 84% or 89 % efficacious, respectively (p=0.047 or p=0.031). Evaluating the number of menstrual cycles rather than exposures until infection can account for varying susceptibility during the menstrual cycle. Our observations have implications for future study designs such as planning the frequency of virus exposures. Menstrual cycle analysis may also avoid potential overestimation of efficacy against vaginal challenges during low susceptibility periods in the cycle that are unlikely to cause infection.


Asunto(s)
Antiinfecciosos Locales/administración & dosificación , Antirretrovirales/administración & dosificación , Transmisión de Enfermedad Infecciosa/prevención & control , Ciclo Menstrual/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/transmisión , Virus de la Inmunodeficiencia de los Simios/crecimiento & desarrollo , Administración Tópica , Animales , Susceptibilidad a Enfermedades , Femenino , Macaca nemestrina , Placebos/administración & dosificación , Resultado del Tratamiento
7.
AIDS Res Hum Retroviruses ; 30(11): 1125-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25313448

RESUMEN

The repeat low-dose virus challenge model is commonly used in nonhuman primate studies of HIV transmission and biomedical preventions. For some viruses or challenge routes, it is uncertain whether the repeated exposure design might induce virus-directed innate or adaptive immunity that could affect infection or viremic outcomes. Retrospective cohorts of male Indian rhesus (n=40) and female pigtail (n=46) macaques enrolled in repeat low-dose rectal or vaginal SHIV(SF162p3) challenge studies, respectively, were studied to compare the relationship between the number of previous exposures and peak plasma SHIV RNA levels or viral load area under the curve (AUC), surrogate markers of viral control. Repeated mucosal exposures of 10 or 50 TCID50 of virus for rectal and vaginal exposures, respectively, were performed. Virus levels were measured by quantitative reverse-transcriptase real-time PCR. The cumulative number of SHIV(SF162p3) exposures did not correlate with observed peak virus levels or with AUC in rectally challenged rhesus macaques [peak: rho (ρ)=0.04, p=0.8; AUC: ρ=0.33, p=0.06] or vaginally challenged pigtail macaques (peak: ρ=-0.09, p=0.7; AUC: ρ=0.11, p=0.6). Infections in these models occur independently of exposure history and provide assurance that neither inoculation route nor number of exposures required for infection correlates with postinfection viremia. These data also indicate that both the vaginal and rectal repeated low-dose virus exposure models using SHIV(SF162p3) provide a reliable system for nonhuman primate studies.


Asunto(s)
VIH/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Carga Viral , Animales , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Macaca mulatta , Macaca nemestrina , Masculino , ARN Viral/sangre , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA