Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Virol J ; 11: 92, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24886416

RESUMEN

BACKGROUND: HIV-1 gene expression is driven by the long terminal repeat (LTR), which contains many binding sites shown to interact with an array of host and viral factors. Selective pressures within the host as well as the low fidelity of reverse transcriptase lead to changes in the relative prevalence of genetic variants within the HIV-1 genome, including the LTR, resulting in viral quasispecies that can be differentially regulated and can potentially establish niches within specific cell types and tissues. METHODS: Utilizing flow cytometry and electromobility shift assays, specific single-nucleotide sequence polymorphisms (SNPs) were shown to alter both the phenotype of LTR-driven transcription and reactivation. Additional studies also demonstrated differential loading of transcription factors to probes derived from the double-variant LTR as compared to probes from the wild type. RESULTS: This study has identified specific SNPs within CCAAT/enhancer binding protein (C/EBP) site I and Sp site III (3 T, C-to-T change at position 3, and 5 T, C-to-T change at position 5 of the binding site, respectively) that alter LTR-driven gene transcription and may alter the course of viral latency and reactivation. The HIV-1 LAI LTRs containing the SNPs of interest were coupled to a plasmid encoding green fluorescent protein (GFP), and polyclonal HIV-1 LTR-GFP stable cell lines utilizing bone marrow progenitor, T, and monocytic cell lines were constructed and utilized to explore the LTR phenotype associated with these genotypic changes. CONCLUSIONS: Although the 3 T and 5 T SNPs have been shown to be low-affinity binding sites, the fact that they can still result in effective HIV-1 LTR-driven gene expression, particularly within the TF-1 cell line, has suggested that the low binding site affinities associated with the 3 T C/EBP site I and 5 T Sp site III are potentially compensated for by the interaction of nuclear factor-κB with its corresponding binding sites under selected physiological and cellular conditions. Additionally, tumor necrosis factor-α and Tat can enhance basal transcription of each SNP-specific HIV-1 LTR; however, differential regulation of the LTR is both SNP- and cell type-specific.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Duplicado del Terminal Largo de VIH/genética , VIH-1/genética , Polimorfismo de Nucleótido Simple , Factor de Transcripción Sp1/metabolismo , Línea Celular , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , VIH-1/fisiología , Humanos , Unión Proteica , Transcripción Genética , Activación Viral
2.
J Gen Virol ; 93(Pt 6): 1151-1172, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22422068

RESUMEN

Despite the success of highly active antiretroviral therapy in combating human immunodeficiency virus type 1 (HIV-1) infection, the virus still persists in viral reservoirs, often in a state of transcriptional silence. This review focuses on the HIV-1 protein and regulatory machinery and how expanding knowledge of the function of individual HIV-1-coded proteins has provided valuable insights into understanding HIV transcriptional regulation in selected susceptible cell types. Historically, Tat has been the most studied primary transactivator protein, but emerging knowledge of HIV-1 transcriptional regulation in cells of the monocyte-macrophage lineage has more recently established that a number of the HIV-1 accessory proteins like Vpr may directly or indirectly regulate the transcriptional process. The viral proteins Nef and matrix play important roles in modulating the cellular activation pathways to facilitate viral replication. These observations highlight the cross talk between the HIV-1 transcriptional machinery and cellular activation pathways. The review also discusses the proposed transcriptional regulation mechanisms that intersect with the pathways regulated by microRNAs and how development of the knowledge of chromatin biology has enhanced our understanding of key protein-protein and protein-DNA interactions that form the HIV-1 transcriptome. Finally, we discuss the potential pharmacological approaches to target viral persistence and enhance effective transcription to purge the virus in cellular reservoirs, especially within the central nervous system, and the novel therapeutics that are currently in various stages of development to achieve a much superior prognosis for the HIV-1-infected population.


Asunto(s)
Regulación Viral de la Expresión Génica , Infecciones por VIH/virología , VIH-1/metabolismo , Proteínas Virales/metabolismo , Animales , Fármacos Anti-VIH/farmacología , Regulación Viral de la Expresión Génica/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , VIH-1/genética , Humanos , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética , Replicación Viral
3.
Sci Rep ; 10(1): 424, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31949209

RESUMEN

Foxp3+ T-regulatory (Treg) cells are capable of suppressing immune responses. Lysine acetylation is a key mechanism of post-translational control of various transcription factors, and when acetylated, Foxp3 is stabilized and transcriptionally active. Therefore, understanding the roles of various histone/protein deacetylases (HDAC) are key to promoting Treg-based immunotherapy. Several of the 11 classical HDAC enzymes are necessary for optimal Treg function while others are dispensable. We investigated the effect of HDAC10 in murine Tregs. HDAC10 deletion had no adverse effect on the health of mice, which retained normal CD4+ and CD8+ T cell function. However, HDAC10-/- Treg exhibited increased suppressive function in vitro and in vivo. C57BL/6 Rag1-/- mice adoptively transferred with HDAC10-/- but not wild Treg, were protected from developing colitis. HDAC10-/- but not wild-type mice receiving fully MHC-mismatched cardiac transplants became tolerant and showed long-term allograft survival (>100 d). We conclude that targeting of HDAC10 may be of therapeutic value for inflammatory disorders including colitis and also for transplantation.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Eliminación de Gen , Linfocitos T Reguladores/citología , Animales , Colitis/genética , Colitis/inmunología , Células HEK293 , Trasplante de Corazón/efectos adversos , Humanos , Ratones , Tolerancia al Trasplante/genética
4.
Transplantation ; 102(1): 70-78, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29272255

RESUMEN

BACKGROUND: Vascularized composite allografts (VCA) are novel, life-enhancing forms of transplantation (Tx). However, host immune responses to the various VCA components, especially those involving skin, are complex and make selection of appropriate therapy challenging. Although the interplay between Foxp3+ T regulatory (Treg) cells and CD4 and CD8 effector T cells is of central importance in determining the acceptance or rejection of solid organ allografts, there is little information available concerning the contribution of Treg cells to VCA survival. In addition, the effects of therapeutic expansion in vivo of host Treg cell populations on VCA survival are unknown. METHODS: We established a fully major histocompatibility complex-disparate (BALB/c- > C57BL/6) murine orthotopic forelimb Tx model to explore the benefits of pre- and post-Tx IL-2/anti-IL-2 monoclonal antibody complex (IL-2C) administration to expand the host Treg cell population and thereby attempt to promote Treg cell-dependent VCA survival. RESULTS: Both strategies expanded the Treg cell population in vivo and prolonged VCA survival (P < 0.001), but IL-2C administration pre-Tx led to significantly longer survival compared with IL-2C administration post-Tx (P < 0.01). In addition, compared with post-Tx therapy, pre-Tx therapy resulted in an increased ratio of Treg cells to CD8+ T cells (P < 0.001), reduced proliferation of CD4 and CD8 effector T cells, and reduced production of IFN-γ. Optimal effects were seen when combined with rapamycin therapy, whereas the combination of IL-2C therapy plus calcineurin inhibitor was counterproductive. CONCLUSIONS: Our studies involving different IL-2C-mediated Treg cell expansion strategies demonstrate that pre-Tx IL-2C therapy may be a useful component for developing strategies to promote VCA survival.


Asunto(s)
Aloinjertos Compuestos , Miembro Anterior/trasplante , Supervivencia de Injerto/efectos de los fármacos , Interleucina-2/farmacología , Animales , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T Reguladores/inmunología
5.
Sci Rep ; 7(1): 8626, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28819166

RESUMEN

Current interest in Foxp3+ T-regulatory (Treg) cells as therapeutic targets in transplantation is largely focused on their harvesting pre-transplant, expansion and infusion post-transplantation. An alternate strategy of pharmacologic modulation of Treg function using histone/protein deacetylase inhibitors (HDACi) may allow more titratable and longer-term dosing. However, the effects of broadly acting HDACi vary, such that HDAC isoform-selective targeting is likely required. We report data from mice with constitutive or conditional deletion of HDAC11 within Foxp3+ Treg cells, and their use, along with small molecule HDAC11 inhibitors, in allograft models. Global HDAC11 deletion had no effect on health or development, and compared to WT controls, Foxp3+ Tregs lacking HDAC11 showed increased suppressive function, and increased expression of Foxp3 and TGF-ß. Likewise, compared to WT recipients, conditional deletion of HDAC11 within Tregs led to long-term survival of fully MHC-mismatched cardiac allografts, and prevented development of transplant arteriosclerosis in an MHC class II-mismatched allograft model. The translational significance of HDAC11 targeting was shown by the ability of an HDAC11i to promote long-term allograft allografts in fully MHC-disparate strains. These data are powerful stimuli for the further development and testing of HDAC11-selective pharmacologic inhibitors, and may ultimately provide new therapies for transplantation and autoimmune diseases.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Histona Desacetilasas/inmunología , Histonas/inmunología , Linfocitos T Reguladores/inmunología , Aloinjertos , Animales , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica/métodos , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Células HEK293 , Trasplante de Corazón/métodos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Linfocitos T Reguladores/metabolismo
6.
Cell Metab ; 25(6): 1282-1293.e7, 2017 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-28416194

RESUMEN

Immune cells function in diverse metabolic environments. Tissues with low glucose and high lactate concentrations, such as the intestinal tract or ischemic tissues, frequently require immune responses to be more pro-tolerant, avoiding unwanted reactions against self-antigens or commensal bacteria. T-regulatory cells (Tregs) maintain peripheral tolerance, but how Tregs function in low-glucose, lactate-rich environments is unknown. We report that the Treg transcription factor Foxp3 reprograms T cell metabolism by suppressing Myc and glycolysis, enhancing oxidative phosphorylation, and increasing nicotinamide adenine dinucleotide oxidation. These adaptations allow Tregs a metabolic advantage in low-glucose, lactate-rich environments; they resist lactate-mediated suppression of T cell function and proliferation. This metabolic phenotype may explain how Tregs promote peripheral immune tolerance during tissue injury but also how cancer cells evade immune destruction in the tumor microenvironment. Understanding Treg metabolism may therefore lead to novel approaches for selective immune modulation in cancer and autoimmune diseases.


Asunto(s)
Microambiente Celular/inmunología , Reprogramación Celular/inmunología , Factores de Transcripción Forkhead/inmunología , Glucosa/inmunología , Ácido Láctico/inmunología , Linfocitos T Reguladores/inmunología , Animales , Línea Celular , Microambiente Celular/genética , Reprogramación Celular/genética , Factores de Transcripción Forkhead/genética , Glucosa/genética , Glucólisis/genética , Glucólisis/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fosforilación Oxidativa , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/inmunología
7.
EBioMedicine ; 13: 99-112, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27769803

RESUMEN

Foxp3+ T-regulatory (Treg) cells are known to suppress protective host immune responses to a wide variety of solid tumors, but their therapeutic targeting is largely restricted to their transient depletion or "secondary" modulation, e.g. using anti-CTLA-4 monoclonal antibody. Our ongoing studies of the post-translational modifications that regulate Foxp3 demonstrated that the histone/protein acetyltransferase, Tip60, plays a dominant role in promoting acetylation, dimerization and function in Treg cells. We now show that the ubiquitin-specific protease, Usp7, controls Treg function largely by stabilizing the expression and promoting the multimerization of Tip60 and Foxp3. Genetic or pharmacologic targeting of Usp7 impairs Foxp3+ Treg suppressive functions, while conventional T cell responses remain intact. As a result, pharmacologic inhibitors of Usp7 can limit tumor growth in immunocompetent mice, and promote the efficacy of antitumor vaccines and immune checkpoint therapy with anti-PD1 monoclonal antibody in murine models. Hence, pharmacologic therapy with Usp7 inhibitors may have an important role in future cancer immunotherapy.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Histona Acetiltransferasas/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Transactivadores/metabolismo , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores , Animales , Autoinmunidad/genética , Autoinmunidad/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Expresión Génica , Inmunidad , Activación de Linfocitos/inmunología , Lisina Acetiltransferasa 5 , Ratones , Ratones Noqueados , Ratones Transgénicos , Neoplasias/patología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Carga Tumoral , Peptidasa Específica de Ubiquitina 7 , Proteasas Ubiquitina-Específicas/genética
8.
PLoS One ; 9(2): e88116, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24551078

RESUMEN

Transcriptional control of the human immunodeficiency virus type 1 (HIV-1) promoter, the long terminal repeat (LTR), is achieved by interactions with cis-acting elements present both upstream and downstream of the start site. In silico transcription factor binding analysis of the HIV-1 subtype B LTR sequences revealed a potential downstream CCAAT enhancer binding protein (C/EBP) binding site. This binding site (+158 to+172), designated DS3, was found to be conserved in 67% of 3,858 unique subtype B LTR sequences analyzed in terms of nucleotide sequence as well as physical location in the LTR. DS3 was found to be well represented in other subtypes as well. Interestingly, DS3 overlaps with a previously identified region that bind members of the nuclear factor of activated T cells (NFAT) family of proteins. NFATc2 exhibited a higher relative affinity for DS3 as compared with members of the C/EBP family (C/EBP α and ß). DS3 was able to compete efficiently with the low-affinity upstream C/EBP binding site I with respect to C/EBP binding, suggesting utilization of both NFAT and C/EBP. Moreover, cyclosporine A treatment, which has been shown to prevent dephosphorylation and nuclear translocation of NFAT isoforms, resulted in enhanced C/EBPα binding. The interactions at DS3 were also validated in an integrated HIV-1 LTR in chronically infected U1 cells. A binding knockout of DS3 demonstrated reduced HIV-1 LTR-directed transcription under both basal and interleukin-6-stimulated conditions only in cells of the monocyte-macrophage lineage cells and not in cells of T-cell origin. Thus, the events at DS3 positively regulate the HIV-1 promoter in cells of the monocyte-macrophage lineage.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Linaje de la Célula , Duplicado del Terminal Largo de VIH/genética , VIH-1/genética , Macrófagos/metabolismo , Monocitos/metabolismo , Factores de Transcripción NFATC/metabolismo , Secuencia de Bases , Sitios de Unión , Línea Celular , Secuencia Conservada/genética , Humanos , Macrófagos/citología , Datos de Secuencia Molecular , Monocitos/citología , Unión Proteica/genética , Isoformas de Proteínas/metabolismo , Sitio de Iniciación de la Transcripción , Transcripción Genética
9.
Adv Virus Res ; 87: 183-240, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23809924

RESUMEN

HIV-associated neurologic disease continues to be a significant complication in the era of highly active antiretroviral therapy. A substantial subset of the HIV-infected population shows impaired neuropsychological performance as a result of HIV-mediated neuroinflammation and eventual central nervous system (CNS) injury. CNS compartmentalization of HIV, coupled with the evolution of genetically isolated populations in the CNS, is responsible for poor prognosis in patients with AIDS, warranting further investigation and possible additions to the current therapeutic strategy. This chapter reviews key advances in the field of neuropathogenesis and studies that have highlighted how molecular diversity within the HIV genome may impact HIV-associated neurologic disease. We also discuss the possible functional implications of genetic variation within the viral promoter and possibly other regions of the viral genome, especially in the cells of monocyte-macrophage lineage, which are arguably key cellular players in HIV-associated CNS disease.


Asunto(s)
Complejo SIDA Demencia/virología , Enfermedades del Sistema Nervioso Central/inmunología , Enfermedades del Sistema Nervioso Central/virología , Infecciones por VIH/virología , VIH-1/patogenicidad , Complejo SIDA Demencia/diagnóstico , Complejo SIDA Demencia/inmunología , Complejo SIDA Demencia/patología , Terapia Antirretroviral Altamente Activa , Enfermedades del Sistema Nervioso Central/diagnóstico , Variación Genética , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/patología , Duplicado del Terminal Largo de VIH/genética , VIH-1/genética , Humanos , Macrófagos/inmunología , Monocitos/inmunología , Regiones Promotoras Genéticas , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/genética
10.
Adv Virol ; 2012: 123605, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22899925

RESUMEN

The human immunodeficiency virus type 1 (HIV-1) promoter or long-terminal repeat (LTR) regulates viral gene expression by interacting with multiple viral and host factors. The viral transactivator protein Tat plays an important role in transcriptional activation of HIV-1 gene expression. Functional domains of Tat and its interaction with transactivation response element RNA and cellular transcription factors have been examined. Genetic variation within tat of different HIV-1 subtypes has been shown to affect the interaction of the viral transactivator with cellular and/or viral proteins, influencing the overall level of transcriptional activation as well as its action as a neurotoxic protein. Consequently, the genetic variability within tat may impact the molecular architecture of functional domains of the Tat protein that may impact HIV pathogenesis and disease. Tat as a therapeutic target for anti-HIV drugs has also been discussed.

11.
J Virol Methods ; 167(1): 10-6, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20211204

RESUMEN

A tetravalent dengue vaccine that can protect against all four serotypes of dengue viruses is a global priority. The host-receptor binding, multiple neutralizing epitope-containing carboxy-terminal region of the dengue envelope protein, known as domain III (EDIII), has emerged as a promising subunit vaccine antigen. One strategy to develop a tetravalent dengue subunit vaccine envisages mixing recombinant EDIIIs, corresponding to the four dengue virus serotypes. Towards this objective, a recombinant clone of the methylotrophic yeast Pichia pastoris, harboring the EDIII gene of dengue virus type 2 (EDIII-2) for its intracellular expression, was created. Recombinant EDIII-2 protein, expressed by this clone was purified to near homogeneity by affinity chromatography, with final yields of >50mg/l culture. Groups of Balb/c mice were immunized with this protein, separately formulated in two adjuvants, alum and montanide ISA 720. The EDIII-2 antigen, formulated in either adjuvant, elicited high levels of neutralizing antibodies to dengue virus type 2 in mice as analyzed by Plaque Reduction Neutralization Test (PRNT). This study demonstrates the feasibility of using P. pastoris to produce EDIII antigens capable of eliciting potent virus-neutralizing antibodies.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Vacunas contra el Dengue/inmunología , Proteínas del Envoltorio Viral/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Compuestos de Alumbre/administración & dosificación , Animales , Vacunas contra el Dengue/genética , Virus del Dengue/inmunología , Manitol/administración & dosificación , Manitol/análogos & derivados , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Pruebas de Neutralización , Ácidos Oléicos/administración & dosificación , Pichia/genética , Análisis de Secuencia de ADN , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética , Ensayo de Placa Viral
12.
Am J Trop Med Hyg ; 79(3): 353-63, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18784226

RESUMEN

There is currently no vaccine to prevent dengue (DEN) virus infection, which is caused by any one of four closely related serotypes, DEN-1, DEN-2, DEN-3, or DEN-4. A DEN vaccine must be tetravalent, because immunity to a single serotype does not offer cross-protection against the other serotypes. We have developed a novel tetravalent chimeric protein by fusing the receptor-binding envelope domain III (EDIII) of the four DEN virus serotypes. This protein was expressed in the yeast, Pichia pastoris, and purified to near homogeneity in high yields. Antibodies induced in mice by the tetravalent protein, formulated in different adjuvants, neutralized the infectivity of all four serotypes. This, coupled with the high expression potential of the P. pastoris system and easy one-step purification, makes the EDIII-based recombinant protein a potentially promising candidate for the development of a safe, efficacious, and inexpensive, tetravalent DEN vaccine.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Antígenos Virales/inmunología , Virus del Dengue/inmunología , Pichia/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Línea Celular , Cricetinae , Culicidae , Dengue/inmunología , Dengue/prevención & control , Dengue/virología , Vacunas contra el Dengue/inmunología , Virus del Dengue/clasificación , Relación Dosis-Respuesta Inmunológica , Ingeniería Genética , Haplorrinos , Ratones , Ratones Endogámicos BALB C , Pichia/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Serotipificación , Bazo/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA