Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
JCI Insight ; 4(11)2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31167974

RESUMEN

Reduction/elimination of HIV-1 reservoirs that persist despite combination antiretroviral therapy (cART) will likely require induction of viral expression by residual infected cells and enhanced clearance of these cells. TLR7 agonists have potential to mediate these activities. We evaluated immunologic and virologic effects of repeated doses of the TLR7 agonist GS-9620 in SIV-infected rhesus macaques receiving cART, which was initiated at 13 days after infection and was continued for 75 weeks prior to GS-9620 administration. During cART, GS-9620 induced transient upregulation of IFN-stimulated genes in blood and tissues, increases in plasma cytokines, and changes in immune cell population activation and phenotypes but did not result in measurable increases in plasma viremia or viral RNA-to-viral DNA ratio in PBMCs or tissues nor decreases in viral DNA in PBMC or tissues. SIV-specific CD8+ T cell responses, negligible prior to GS-9620 treatment, were not measurably boosted by treatment; a second course of GS-9620 administration overlapping with later cART discontinuation was associated with increased CD8+ T cell responses during viral recrudescence. These results confirm and extend evidence for GS-9620-mediated enhancement of antiviral immune responses in SIV-infected macaques but suggest that GS-9620-mediated viral induction may depend critically on the timing of initiation and duration of cART and resulting characteristics of viral reservoirs.


Asunto(s)
Antirretrovirales , Pteridinas , Síndrome de Inmunodeficiencia Adquirida del Simio , Receptor Toll-Like 7/agonistas , Viremia , Animales , Antirretrovirales/administración & dosificación , Antirretrovirales/farmacología , Antirretrovirales/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Citocinas/metabolismo , Quimioterapia Combinada , Macaca mulatta , Masculino , Pteridinas/administración & dosificación , Pteridinas/farmacología , Pteridinas/uso terapéutico , ARN Viral/genética , ARN Viral/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Regulación hacia Arriba/efectos de los fármacos , Carga Viral/efectos de los fármacos , Viremia/tratamiento farmacológico , Viremia/inmunología , Viremia/virología
2.
Viruses ; 10(5)2018 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-29710792

RESUMEN

CD8⁺ T cells are critical for controlling viremia during human immunodeficiency virus (HIV) infection. These cells produce cytolytic factors and antiviral cytokines that eliminate virally- infected cells. During the chronic phase of HIV infection, CD8⁺ T cells progressively lose their proliferative capacity and antiviral functions. These dysfunctional cells are unable to clear the productively infected and reactivated cells, representing a roadblock in HIV cure. Therefore, mechanisms to understand CD8⁺ T cell dysfunction and strategies to boost CD8⁺ T cell function need to be investigated. Using the feline immunodeficiency virus (FIV) model for lentiviral persistence, we have demonstrated that CD8⁺ T cells exhibit epigenetic changes such as DNA demethylation during the course of infection as compared to uninfected cats. We have also demonstrated that lentivirus-activated CD4⁺CD25⁺ T regulatory cells induce forkhead box P3 (Foxp3) expression in virus-specific CD8⁺ T cell targets, which binds the interleukin (IL)-2, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ promoters in these CD8⁺ T cells. Finally, we have reported that epigenetic modulation reduces Foxp3 binding to these promoter regions. This review compares and contrasts our current understanding of CD8⁺ T cell epigenetics and mechanisms of lymphocyte suppression during the course of lentiviral infection for two animal models, FIV and simian immunodeficiency virus (SIV).


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epigénesis Genética , Virus de la Inmunodeficiencia Felina , Infecciones por Lentivirus/inmunología , Infecciones por Lentivirus/veterinaria , Virus de la Inmunodeficiencia de los Simios , Animales , Gatos , Citocinas/inmunología , Infecciones por VIH/inmunología , Humanos , Activación de Linfocitos , Linfocitos T Reguladores/inmunología , Viremia/veterinaria
3.
AIDS Res Hum Retroviruses ; 34(3): 269-276, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29037051

RESUMEN

Polyfunctional CD8+ T cells play a critical role in controlling viremia during AIDS lentiviral infections. However, for most HIV-infected individuals, virus-specific CD8+ T cells exhibit loss of polyfunctionality, including loss of IL2, TNFα, and IFNγ. Using the feline immunodeficiency virus (FIV) model for AIDS lentiviral persistence, our laboratory has demonstrated that FIV-activated Treg cells target CD8+ T cells, leading to a reduction in IL2 and IFNγ production. Furthermore, we have demonstrated that Treg cells induce expression of the repressive transcription factor, Foxp3, in CD8+ T cells. Based upon these findings, we asked if Treg-induced Foxp3 could bind to the IL2, TNFα, and IFNγ promoter regions in virus-specific CD8+ T cells. Following coculture with autologous Treg cells, we demonstrated decreased mRNA levels of IL2 and IFNγ at weeks 4 and 8 postinfection and decreased TNFα at week 4 postinfection in virus-specific CD8+ T cells. We also clearly demonstrated Treg cell-induced Foxp3 expression in virus-specific CD8+ T cells at weeks 1, 4, and 8 postinfection. Finally, we documented Foxp3 binding to the IL2, TNFα, and IFNγ promoters at 8 weeks and 6 months postinfection in virus-specific CD8+ T cells following Treg cell coculture. In summary, the results here clearly demonstrate that Foxp3 inhibits IL2, TNFα, and IFNγ transcription by binding to their promoter regions in lentivirus-specific CD8+ T cells. We believe this is the first description of this process during the course of AIDS lentiviral infection.


Asunto(s)
Linfocitos T CD8-positivos/virología , Síndrome de Inmunodeficiencia Adquirida del Felino/inmunología , Factores de Transcripción Forkhead/metabolismo , Virus de la Inmunodeficiencia Felina , Interferón gamma/genética , Interleucina-2/genética , Linfocitos T Reguladores/inmunología , Factor de Necrosis Tumoral alfa/genética , Animales , Linfocitos T CD8-positivos/inmunología , Gatos , Modelos Animales de Enfermedad , Síndrome de Inmunodeficiencia Adquirida del Felino/virología , Factores de Transcripción Forkhead/biosíntesis , Activación de Linfocitos , Regiones Promotoras Genéticas , Viremia/inmunología , Viremia/virología
4.
Viruses ; 10(6)2018 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-29861472

RESUMEN

CD8⁺ T cells are critical for controlling HIV infection. During the chronic phase of lentiviral infection, CD8⁺ T cells lose their proliferative capacity and exhibit impaired antiviral function. This loss of CD8⁺ T cell function is due, in part, to CD4⁺CD25⁺ T regulatory (Treg) cell-mediated suppression. Our research group has demonstrated that lentivirus-activated CD4⁺CD25⁺ Treg cells induce the repressive transcription factor forkhead box P3 (Foxp3) in autologous CD8⁺ T cells following co-culture. We have recently reported that Treg-induced Foxp3 binds the interleukin-2 (IL-2), interferon-γ (IFN- γ), and tumor necrosis factor-α (TNF-α) promoters in virus-specific CD8⁺ T cells. These data suggest an important role of Foxp3-mediated CD8⁺ T cell dysfunction in lentiviral infection. To elucidate the mechanism of this suppression, we previously reported that decreased methylation facilitates Foxp3 binding in mitogen-activated CD8⁺ T cells from feline immunodeficiency virus (FIV)-infected cats. We demonstrated the reduced binding of Foxp3 to the IL-2 promoter by increasing methylation of CD8⁺ T cells. In the studies presented here, we ask if another form of epigenetic modulation might alleviate Foxp3-mediated suppression in CD8⁺ T cells. We hypothesized that decreasing histone acetylation in virus-specific CD8⁺ T cells would decrease Treg-induced Foxp3 binding to the IL-2 promoter. Indeed, using anacardic acid (AA), a known histone acetyl transferase (HAT) inhibitor, we demonstrate a reduction in Foxp3 binding to the IL-2 promoter in virus-specific CD8⁺ T cells co-cultured with autologous Treg cells. These data identify a novel mechanism of Foxp3-mediated CD8⁺ T cell dysfunction during lentiviral infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Síndrome de Inmunodeficiencia Adquirida del Felino/inmunología , Factores de Transcripción Forkhead/metabolismo , Histonas/metabolismo , Interleucina-2/genética , Linfocitos T Reguladores/inmunología , Acetilación , Ácidos Anacárdicos/farmacología , Animales , Linfocitos T CD8-positivos/virología , Gatos , Técnicas de Cocultivo , Epigénesis Genética , Virus de la Inmunodeficiencia Felina , Interferón gamma/inmunología , Regiones Promotoras Genéticas
5.
Vet Sci ; 4(1)2017 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-29056671

RESUMEN

CD4⁺CD25⁺Foxp3⁺ T regulatory (Treg) cells are activated during the course of lentiviral infection and exhibit heightened suppressor function when compared to Treg cells from uninfected controls. Foxp3 is essential to Treg cell function and multiple studies have documented that lentivirus-activated Treg cells exhibit heightened Foxp3 expression when compared to Treg cells from uninfected controls. Our hypothesis was that lentivirus-induced micro-RNAs (miRNAs) contribute to heightened Treg cell suppressor function by stabilizing Foxp3 expression. We demonstrated that CD4⁺CD25⁺ T cells from both feline immunodeficiency virus infected (FIV⁺) cats and uninfected control cats exhibit increased miRNA 10a and 21 levels compared to autologous CD4⁺CD25- T cells but there was no difference in the levels of these miRNAs when Treg cells from FIV⁺ cats were compared to Treg cells from uninfected controls. Further, there was no increase in Foxp3 mRNA following transfection of miRNA 10a or 21 into a feline cell line. However, transfection with miRNA 10a resulted in increased Foxp3 protein expression.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA