Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Genome Med ; 13(1): 168, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702310

RESUMEN

BACKGROUND: In spite of many years of research, our understanding of the molecular bases of Alzheimer's disease (AD) is still incomplete, and the medical treatments available mainly target the disease symptoms and are hardly effective. Indeed, the modulation of a single target (e.g., ß-secretase) has proven to be insufficient to significantly alter the physiopathology of the disease, and we should therefore move from gene-centric to systemic therapeutic strategies, where AD-related changes are modulated globally. METHODS: Here we present the complete characterization of three murine models of AD at different stages of the disease (i.e., onset, progression and advanced). We combined the cognitive assessment of these mice with histological analyses and full transcriptional and protein quantification profiling of the hippocampus. Additionally, we derived specific Aß-related molecular AD signatures and looked for drugs able to globally revert them. RESULTS: We found that AD models show accelerated aging and that factors specifically associated with Aß pathology are involved. We discovered a few proteins whose abundance increases with AD progression, while the corresponding transcript levels remain stable, and showed that at least two of them (i.e., lfit3 and Syt11) co-localize with Aß plaques in the brain. Finally, we found two NSAIDs (dexketoprofen and etodolac) and two anti-hypertensives (penbutolol and bendroflumethiazide) that overturn the cognitive impairment in AD mice while reducing Aß plaques in the hippocampus and partially restoring the physiological levels of AD signature genes to wild-type levels. CONCLUSIONS: The characterization of three AD mouse models at different disease stages provides an unprecedented view of AD pathology and how this differs from physiological aging. Moreover, our computational strategy to chemically revert AD signatures has shown that NSAID and anti-hypertensive drugs may still have an opportunity as anti-AD agents, challenging previous reports.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Proteómica/métodos , Transcriptoma , Envejecimiento , Péptidos beta-Amiloides , Animales , Encéfalo/metabolismo , Disfunción Cognitiva , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Sustitución del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide/metabolismo
2.
Nat Commun ; 12(1): 3932, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34168145

RESUMEN

Chemical descriptors encode the physicochemical and structural properties of small molecules, and they are at the core of chemoinformatics. The broad release of bioactivity data has prompted enriched representations of compounds, reaching beyond chemical structures and capturing their known biological properties. Unfortunately, bioactivity descriptors are not available for most small molecules, which limits their applicability to a few thousand well characterized compounds. Here we present a collection of deep neural networks able to infer bioactivity signatures for any compound of interest, even when little or no experimental information is available for them. Our signaturizers relate to bioactivities of 25 different types (including target profiles, cellular response and clinical outcomes) and can be used as drop-in replacements for chemical descriptors in day-to-day chemoinformatics tasks. Indeed, we illustrate how inferred bioactivity signatures are useful to navigate the chemical space in a biologically relevant manner, unveiling higher-order organization in natural product collections, and to enrich mostly uncharacterized chemical libraries for activity against the drug-orphan target Snail1. Moreover, we implement a battery of signature-activity relationship (SigAR) models and show a substantial improvement in performance, with respect to chemistry-based classifiers, across a series of biophysics and physiology activity prediction benchmarks.


Asunto(s)
Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad , Línea Celular Tumoral , Bases de Datos Farmacéuticas , Evaluación Preclínica de Medicamentos/métodos , Humanos , Factores de Transcripción de la Familia Snail/antagonistas & inhibidores , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo
3.
J Chem Inf Model ; 60(12): 5730-5734, 2020 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-32672454

RESUMEN

Until a vaccine becomes available, the current repertoire of drugs is our only therapeutic asset to fight the SARS-CoV-2 outbreak. Indeed, emergency clinical trials have been launched to assess the effectiveness of many marketed drugs, tackling the decrease of viral load through several mechanisms. Here, we present an online resource, based on small-molecule bioactivity signatures and natural language processing, to expand the portfolio of compounds with potential to treat COVID-19. By comparing the set of drugs reported to be potentially active against SARS-CoV-2 to a universe of 1 million bioactive molecules, we identify compounds that display analogous chemical and functional features to the current COVID-19 candidates. Searches can be filtered by level of evidence and mechanism of action, and results can be restricted to drug molecules or include the much broader space of bioactive compounds. Moreover, we allow users to contribute COVID-19 drug candidates, which are automatically incorporated to the pipeline once per day. The computational platform, as well as the source code, is available at https://sbnb.irbbarcelona.org/covid19.


Asunto(s)
Antivirales/química , Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos/métodos , SARS-CoV-2/efectos de los fármacos , Antivirales/farmacología , Simulación por Computador , Diseño de Fármacos , Humanos , Modelos Moleculares , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología
4.
Nat Biotechnol ; 38(9): 1098, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32440008

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Nat Biotechnol ; 38(9): 1087-1096, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32440005

RESUMEN

Small molecules are usually compared by their chemical structure, but there is no unified analytic framework for representing and comparing their biological activity. We present the Chemical Checker (CC), which provides processed, harmonized and integrated bioactivity data on ~800,000 small molecules. The CC divides data into five levels of increasing complexity, from the chemical properties of compounds to their clinical outcomes. In between, it includes targets, off-targets, networks and cell-level information, such as omics data, growth inhibition and morphology. Bioactivity data are expressed in a vector format, extending the concept of chemical similarity to similarity between bioactivity signatures. We show how CC signatures can aid drug discovery tasks, including target identification and library characterization. We also demonstrate the discovery of compounds that reverse and mimic biological signatures of disease models and genetic perturbations in cases that could not be addressed using chemical information alone. Overall, the CC signatures facilitate the conversion of bioactivity data to a format that is readily amenable to machine learning methods.


Asunto(s)
Preparaciones Farmacéuticas/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Productos Biológicos/química , Productos Biológicos/metabolismo , Productos Biológicos/uso terapéutico , Biomarcadores Farmacológicos/metabolismo , Bases de Datos Factuales , Descubrimiento de Drogas , Quimioterapia , Humanos , Preparaciones Farmacéuticas/química , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/uso terapéutico
6.
Antiviral Res ; 142: 123-135, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28359840

RESUMEN

SAMHD1 is a triphosphohydrolase that restricts HIV-1 by limiting the intracellular dNTP pool required for reverse transcription. Although SAMHD1 is expressed and active/unphosphorylated in most cell lines, its restriction activity is thought to be relevant only in non-cycling cells. However, an in depth evaluation of SAMHD1 function and relevance in cycling cells is required. Here, we show that SAMHD1-induced degradation by HIV-2 Vpx affects the dNTP pool and HIV-1 replication capacity in the presence of the 3'-azido-3'-deoxythymidine (AZT) in cycling cells. Similarly, in SAMHD1 knockout cells, HIV-1 showed increased replicative capacity in the presence of nucleoside inhibitors, especially AZT, that was reverted by re-expression of wild type SAMHD1. Sensitivity to non-nucleoside inhibitors (nevirapine and efavirenz) or the integrase inhibitor raltegravir was not affected by SAMHD1. Combination of three mutations (S18A, T21A, T25A) significantly prevented SAMHD1 phosphorylation but did not significantly affect HIV-1 replication in the presence of AZT. Our results demonstrate that SAMHD1 is active in HIV-1 permissive cells, does not modify susceptibility to HIV-1 infection but strongly affects sensitivity to nucleoside inhibitors.


Asunto(s)
VIH-1/efectos de los fármacos , Proteína 1 que Contiene Dominios SAM y HD/farmacología , Replicación Viral/efectos de los fármacos , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Replicación del ADN/efectos de los fármacos , Edición Génica , Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Infecciones por VIH/metabolismo , VIH-1/patogenicidad , VIH-2/efectos de los fármacos , Interacciones Huésped-Patógeno , Humanos , Fosforilación , Transcripción Reversa/efectos de los fármacos , Proteína 1 que Contiene Dominios SAM y HD/genética , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Proteínas Reguladoras y Accesorias Virales/efectos de los fármacos , Zidovudina/farmacología
7.
J Antimicrob Chemother ; 71(2): 387-94, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26542306

RESUMEN

OBJECTIVES: Sterile α motif and histidine-aspartate domain-containing protein 1 (SAMHD1) has been shown to restrict retroviruses and DNA viruses by decreasing the pool of intracellular deoxynucleotides. In turn, SAMHD1 is controlled by cyclin-dependent kinases (CDK) that regulate the cell cycle and cell proliferation. Here, we explore the effect of CDK6 inhibitors on the replication of herpes simplex virus type 1 (HSV-1) in primary monocyte-derived macrophages (MDM). METHODS: MDM were treated with palbociclib, a selective CDK4/6 inhibitor, and then infected with a GFP-expressing HSV-1. Intracellular deoxynucleotide triphosphate (dNTP) content was determined using a polymerase-based method. RESULTS: CDK6 inhibitor palbociclib blocked SAMHD1 phosphorylation, intracellular dNTP levels and HSV-1 replication in MDM at subtoxic concentrations. Treatment of MDM with palbociclib reduced CDK2 activation, measured as the phosphorylation of the T-loop at Thr160. The antiviral activity of palbociclib was lost when SAMHD1 was degraded by viral protein X. Similarly, palbociclib did not block HSV-1 replication in SAMHD1-negative Vero cells at subtoxic concentrations, providing further evidence for a role of SAMHD1 in mediating the antiviral effect. CONCLUSIONS: SAMHD1-mediated HSV-1 restriction is controlled by CDK and points to a preferential role for CDK6 and CDK2 as mediators of SAMHD1 activation. Similarly, the restricting activity of SAMHD1 against DNA viruses suggests that control of dNTP availability is the major determinant of its antiviral activity. This is the first study describing the anti-HSV-1 activity of palbociclib.


Asunto(s)
Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Herpesvirus Humano 1/fisiología , Macrófagos/virología , Proteínas de Unión al GTP Monoméricas/metabolismo , Piperazinas/farmacología , Piridinas/farmacología , Replicación Viral/efectos de los fármacos , Animales , Células Cultivadas , Herpesvirus Humano 1/efectos de los fármacos , Humanos , Proteína 1 que Contiene Dominios SAM y HD
8.
Cell Cycle ; 14(11): 1657-65, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25927932

RESUMEN

Cyclins control the activation of cyclin-dependent kinases (CDK), which in turn, control the cell cycle and cell division. Intracellular availability of deoxynucleotides (dNTP) plays a fundamental role in cell cycle progression. SAM domain and HD domain-containing protein 1 (SAMHD1) degrades nucleotide triphosphates and controls the size of the dNTP pool. SAMHD1 activity appears to be controlled by CDK. Here, we show that knockdown of cyclin D3 a partner of CDK6 and E2 a partner of CDK2 had a major impact in SAMHD1 phosphorylation and inactivation and led to decreased dNTP levels and inhibition of HIV-1 at the reverse transcription step in primary human macrophages. The effect of cyclin D3 RNA interference was lost after degradation of SAMHD1 by HIV-2 Vpx, demonstrating the specificity of the mechanism. Cyclin D3 inhibition correlated with decreased activation of CDK2. Our results confirm the fundamental role of the CDK6-cyclin D3 pair in controlling CDK2-dependent SAMHD1 phosphorylation and dNTP pool in primary macrophages.


Asunto(s)
Ciclina D3/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Desoxirribonucleótidos/metabolismo , VIH-1/fisiología , Macrófagos/fisiología , Modelos Biológicos , Replicación Viral/fisiología , Ciclina D3/genética , Técnicas de Silenciamiento del Gen , Humanos , Macrófagos/virología , Proteínas de Unión al GTP Monoméricas/metabolismo , Fosforilación , Interferencia de ARN , Proteína 1 que Contiene Dominios SAM y HD
9.
J Med Chem ; 58(1): 183-96, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25075558

RESUMEN

We developed a pharmacophore model for type II inhibitors that was used to guide the construction of a library of kinase inhibitors. Kinome-wide selectivity profiling of the library resulted in the identification of a series of 4-substituted 1H-pyrrolo[2,3-b]pyridines that exhibited potent inhibitory activity against two mitogen-activated protein kinases (MAPKs), TAK1 (MAP3K7) and MAP4K2, as well as pharmacologically well interrogated kinases such as p38α (MAPK14) and ABL. Further investigation of the structure-activity relationship (SAR) resulted in the identification of potent dual TAK1 and MAP4K2 inhibitors such as 1 (NG25) and 2 as well as MAP4K2 selective inhibitors such as 16 and 17. Some of these inhibitors possess good pharmacokinetic properties that will enable their use in pharmacological studies in vivo. A 2.4 Å cocrystal structure of TAK1 in complex with 1 confirms that the activation loop of TAK1 assumes the DFG-out conformation characteristic of type II inhibitors.


Asunto(s)
Quinasas Quinasa Quinasa PAM/química , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/química , Animales , Área Bajo la Curva , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Diseño de Fármacos , Descubrimiento de Drogas , Quinasas del Centro Germinal , Humanos , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Ratones , Modelos Químicos , Modelos Moleculares , Estructura Molecular , Fosforilación/efectos de los fármacos , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteoma/antagonistas & inhibidores , Proteoma/química , Proteoma/metabolismo , Piridinas/química , Piridinas/farmacocinética , Piridinas/farmacología , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacocinética , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
10.
J Biol Chem ; 289(40): 27665-76, 2014 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-25100719

RESUMEN

HIV-1 exploits multiple host proteins during infection. siRNA-based screenings have identified new proteins implicated in different pathways of the viral cycle that participate in a broad range of cellular functions. The human Mediator complex (MED) is composed of 28 elements and represents a fundamental component of the transcription machinery, interacting with the RNA polymerase II enzyme and regulating its ability to express genes. Here, we provide an evaluation of the MED activity on HIV replication. Knockdown of 9 out of 28 human MED proteins significantly impaired viral replication without affecting cell viability, including MED6, MED7, MED11, MED14, MED21, MED26, MED27, MED28, and MED30. Impairment of viral replication by MED subunits was at a post-integration step. Inhibition of early HIV transcripts was observed by siRNA-mediated knockdown of MED6, MED7, MED11, MED14, and MED28, specifically affecting the transcription of the nascent viral mRNA transactivation-responsive element. In addition, MED14 and MED30 were shown to have special relevance during the formation of unspliced viral transcripts (p < 0.0005). Knockdown of the selected MED factors compromised HIV transcription induced by Tat, with the strongest inhibitory effect shown by siMED6 and siMED14 cells. Co-immunoprecipitation experiments suggested physical interaction between MED14 and HIV-1 Tat protein. A better understanding of the mechanisms and factors controlling HIV-1 transcription is key to addressing the development of new strategies required to inhibit HIV replication or reactivate HIV-1 from the latent reservoirs.


Asunto(s)
Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/genética , Complejo Mediador/metabolismo , Transcripción Genética , Regulación Viral de la Expresión Génica , Productos del Gen tat/genética , Productos del Gen tat/metabolismo , Infecciones por VIH/genética , VIH-1/metabolismo , Humanos , Complejo Mediador/genética , Unión Proteica
11.
J Antimicrob Chemother ; 69(11): 3057-60, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25063780

RESUMEN

OBJECTIVES: SAMHD1 and the CDKN1A (p21) cyclin-dependent kinase inhibitor have been postulated to mediate HIV-1 restriction in CD4+ cells. We have shown that p21 affects HIV replication through its effect on SAMHD1. Thus, we aimed at evaluating the expression of SAMHD1 and p21 in different HIV+ phenotypic groups. PATIENTS AND METHODS: We evaluated SAMHD1 and CDKN1A mRNA expression in CD4+ T cells from HIV+ individuals including elite controllers (n = 12), individuals who control HIV without the need for antiretroviral treatment, viraemic progressors (n = 10) and HIV-1 seronegative healthy donors (n = 14). Immunological variables were measured by flow cytometry. RESULTS: We show that a subset of HIV+ elite controllers with lower T cell proliferation levels (Ki67+ cells) expressed higher SAMHD1 compared with healthy donors or viraemic progressors. Conversely, there was no difference in p21 expression before or after T cell activation with a bispecific CD3/CD8 antibody. CONCLUSIONS: Our results suggest that SAMHD1 may play a role in controlling virus replication in HIV+ individuals and slow the rate of disease progression.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , VIH-1/enzimología , Proteínas de Unión al GTP Monoméricas/biosíntesis , Fenotipo , Replicación Viral/fisiología , Humanos , Antígeno Ki-67/biosíntesis , Proteína 1 que Contiene Dominios SAM y HD
12.
AIDS ; 28(15): 2213-22, 2014 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-25036183

RESUMEN

BACKGROUND: Sterile α motif and HD domain-containing protein-1 (SAMHD1) inhibits HIV-1 reverse transcription by decreasing the pool of intracellular deoxynucleotides. SAMHD1 is controlled by cyclin-dependent kinase (CDK)-mediated phosphorylation. However, the exact mechanism of SAMHD1 regulation in primary cells is unclear. We explore the effect of palbociclib, a CDK6 inhibitor, in HIV-1 replication. METHODS: Human primary monocytes were differentiated into macrophages with monocyte-colony stimulating factor and CD4 T lymphocytes stimulated with phytohaemagglutinin (PHA)/interleukin-2. Cells were treated with palbociclib and then infected with a Green fluorescent protein-expressing HIV-1 or R5 HIV-1 BaL. Viral DNA was measured by quantitative PCR and infection assessed by flow cytometry. Deoxynucleotide triphosphate (dNTP) content was determined using a polymerase-based method. RESULTS: Pan-CDK inhibitors AT7519, roscovitine and purvalanol A reduced SAMHD1 phosphorylation. HIV-1 replication was blocked by AT7519 (66.4 ±â€Š3.8%; n = 4), roscovitine (47.3 ±â€Š3.9%; n = 4) and purvalanol A (55.7 ±â€Š15.7%; n = 4) at subtoxic concentrations. Palbociclib, a potent and selective CDK6 inhibitor, blocked SAMHD1 phosphorylation, intracellular dNTP levels, HIV-1 reverse transcription and HIV-1 replication in primary macrophages and CD4 T lymphocytes. Notably, treatment of macrophages with palbociclib led to reduced CDK2 activation, measured as the phosphorylation of the T-loop at the Thr160. The antiviral effect was lost when SAMHD1 was degraded by Vpx, providing further evidence for a role of SAMHD1 in mediating the antiretroviral effect. CONCLUSIONS: Our results indicate that SAMHD1-mediated HIV-1 restriction is controlled by CDK as previously suggested but point to a preferential role for CDK2 and CDK6 as mediators of SAMHD1 activation. Our study provides a new signaling pathway susceptible for the development of new therapeutic approaches against HIV-1 infection.


Asunto(s)
Fármacos Anti-VIH/metabolismo , VIH-1/fisiología , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Piperazinas/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Piridinas/metabolismo , Transcripción Reversa , Células Cultivadas , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , ADN Viral/análisis , Humanos , Leucocitos Mononucleares/virología , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína 1 que Contiene Dominios SAM y HD
13.
J Immunol ; 193(4): 1988-97, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25015816

RESUMEN

Proliferating cells are preferentially susceptible to infection by retroviruses. Sterile α motif and HD domain-containing protein-1 (SAMHD1) is a recently described deoxynucleotide phosphohydrolase controlling the size of the intracellular deoxynucleotide triphosphate (dNTP) pool, a limiting factor for retroviral reverse transcription in noncycling cells. Proliferating (Ki67(+)) primary CD4(+) T cells or macrophages express a phosphorylated form of SAMHD1 that corresponds with susceptibility to infection in cell culture. We identified cyclin-dependent kinase (CDK) 6 as an upstream regulator of CDK2 controlling SAMHD1 phosphorylation in primary T cells and macrophages susceptible to infection by HIV-1. In turn, CDK2 was strongly linked to cell cycle progression and coordinated SAMHD1 phosphorylation and inactivation. CDK inhibitors specifically blocked HIV-1 infection at the reverse transcription step in a SAMHD1-dependent manner, reducing the intracellular dNTP pool. Our findings identify a direct relationship between control of the cell cycle by CDK6 and SAMHD1 activity, which is important for replication of lentiviruses, as well as other viruses whose replication may be regulated by intracellular dNTP availability.


Asunto(s)
Puntos de Control del Ciclo Celular/inmunología , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Infecciones por VIH/inmunología , Proteínas de Unión al GTP Monoméricas/metabolismo , Bencilaminas , Linfocitos T CD4-Positivos/inmunología , Ciclo Celular/inmunología , Células Cultivadas , Ciclamas , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/genética , Células HEK293 , Infecciones por VIH/virología , VIH-1/inmunología , Compuestos Heterocíclicos/farmacología , Humanos , Activación de Linfocitos/inmunología , Linfocitos/inmunología , Macrófagos/inmunología , Células Mieloides/inmunología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Interferencia de ARN , ARN Interferente Pequeño , Receptores CXCR4/antagonistas & inhibidores , Proteína 1 que Contiene Dominios SAM y HD
14.
Antimicrob Agents Chemother ; 58(8): 4804-13, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24913159

RESUMEN

Sterile alpha motif and histidine-aspartic domain-containing protein 1 (SAMHD1) is a deoxynucleoside triphosphate (dNTP) triphosphohydrolase recently recognized as an antiviral factor that acts by depleting dNTP availability for viral reverse transcriptase (RT). SAMHD1 restriction is counteracted by the human immunodeficiency virus type 2 (HIV-2) accessory protein Vpx, which targets SAMHD1 for proteosomal degradation, resulting in an increased availability of dNTPs and consequently enhanced viral replication. Nucleoside reverse transcriptase inhibitors (NRTI), one of the most common agents used in antiretroviral therapy, compete with intracellular dNTPs as the substrate for viral RT. Consequently, SAMHD1 activity may be influencing NRTI efficacy in inhibiting viral replication. Here, a panel of different RT inhibitors was analyzed for their different antiviral efficacy depending on SAMHD1. Antiviral potency was measured for all the inhibitors in transformed cell lines and primary monocyte-derived macrophages and CD4(+) T cells infected with HIV-1 with or without Vpx. No changes in sensitivity to non-NRTI or the integrase inhibitor raltegravir were observed, but for NRTI, sensitivity significantly changed only in the case of the thymidine analogs (AZT and d4T). The addition of exogenous thymidine mimicked the change in viral sensitivity observed after Vpx-mediated SAMHD1 degradation, pointing toward a differential effect of SAMHD1 activity on thymidine. Accordingly, sensitivity to AZT was also reduced in CD4(+) T cells infected with HIV-2 compared to infection with the HIV-2ΔVpx strain. In conclusion, reduction of SAMHD1 levels significantly decreases HIV sensitivity to thymidine but not other nucleotide RT analog inhibitors in both macrophages and lymphocytes.


Asunto(s)
Transcriptasa Inversa del VIH/antagonistas & inhibidores , VIH-2/efectos de los fármacos , Proteínas de Unión al GTP Monoméricas/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología , Estavudina/farmacología , Proteínas Reguladoras y Accesorias Virales/metabolismo , Zidovudina/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Expresión Génica , Transcriptasa Inversa del VIH/genética , Transcriptasa Inversa del VIH/metabolismo , VIH-2/enzimología , Interacciones Huésped-Patógeno , Humanos , Células Jurkat , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/virología , Proteínas de Unión al GTP Monoméricas/genética , Cultivo Primario de Células , Proteína 1 que Contiene Dominios SAM y HD , Timidina/metabolismo , Proteínas Reguladoras y Accesorias Virales/genética , Replicación Viral/efectos de los fármacos
15.
Antimicrob Agents Chemother ; 58(8): 4318-27, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24820090

RESUMEN

Genome editing using zinc finger nucleases (ZFNs) has been successfully applied to disrupt CCR5 or CXCR4 host factors and inhibit viral entry and infection. Gene therapy using ZFNs to modify the PSIP1 gene, which encodes the lens epithelium-derived growth factor (LEDGF) protein, might restrain an early step of the viral replication cycle at the integration level. ZFNs targeting the PSIP1 gene (ZFNLEDGF) were designed to specifically recognize the sequence after the integrase binding domain (IBD) of the LEDGF/p75 protein. ZFNLEDGF successfully recognized the target region of the PSIP1 gene in TZM-bl cells by heteroduplex formation and DNA sequence analysis. Gene editing induced a frameshift of the coding region and resulted in the abolishment of LEDGF expression at the mRNA and protein levels. Functional assays revealed that infection with the HIV-1 R5 BaL or X4 NL4-3 viral strains was impaired in LEDGF/p75 knockout cells regardless of entry tropism due to a blockade in HIV-1 proviral integration into the host genome. However, residual infection was detected in the LEDGF knockout cells. Indeed, LEDGF knockout restriction was overcome at a high multiplicity of infection, suggesting alternative mechanisms for HIV-1 genome integration rather than through LEDGF/p75. However, the observed residual integration was sensitive to the integrase inhibitor raltegravir. These results demonstrate that the described ZFNLEDGF effectively targets the PSIP1 gene, which is involved in the early steps of the viral replication cycle; thus, ZFNLEDGF may become a potential antiviral agent for restricting HIV-1 integration. Moreover, LEDGF knockout cells represent a potent tool for elucidating the role of HIV integration cofactors in virus replication.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Endonucleasas/genética , VIH-1/efectos de los fármacos , Plásmidos/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Dedos de Zinc/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Fármacos Anti-VIH/farmacología , Endonucleasas/metabolismo , Regulación de la Expresión Génica , Integrasa de VIH/genética , Integrasa de VIH/metabolismo , VIH-1/genética , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Células K562 , Datos de Secuencia Molecular , Terapia Molecular Dirigida , Sistemas de Lectura Abierta , Plásmidos/química , Ingeniería de Proteínas , Pirrolidinonas/farmacología , Raltegravir Potásico , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Integración Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
17.
PLoS One ; 8(11): e79221, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244453

RESUMEN

Memory CD4+ T cells are preferentially infected by HIV-1 compared to naïve cells. HIV-1 fusion and entry is a dynamic process in which the cytoskeleton plays an important role by allowing virion internalization and uncoating. Here, we evaluate the role of the cortical actin in cell-to-cell transfer of virus antigens and infection of target CD4+ T cells. Using different actin remodeling compounds we demonstrate that efficiency of HIV-internalization was proportional to the actin polymerization of the target cell. Naïve (CD45RA+) and memory (CD45RA-) CD4+ T cells could be phenotypically differentiated by the degree of cortical actin density and their capacity to capture virus. Thus, the higher cortical actin density of memory CD4+ T cells was associated to increased efficiency of HIV-antigen internalization and the establishment of a productive infection. Conversely, the lower cortical actin density in naïve CD4+ T cells restricted viral antigen transfer and consequently HIV-1 infection. In conclusion, the cortical actin density differentially affects the susceptibility to HIV-1 infection in naïve and memory CD4+ T cells by modulating the efficiency of HIV antigen internalization.


Asunto(s)
Actinas/inmunología , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Memoria Inmunológica , Transporte Biológico/inmunología , Linfocitos T CD4-Positivos/patología , Células Cultivadas , Femenino , Infecciones por VIH/patología , Infecciones por VIH/transmisión , Humanos , Antígenos Comunes de Leucocito/inmunología , Masculino
18.
J Immunol ; 191(5): 2717-30, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23918981

RESUMEN

The roles of IL-1R-associated kinase (IRAK)2 and IRAK1 in cytokine production were investigated using immune cells from knock-in mice expressing the TNFR-associated factor 6 (TRAF6) binding-defective mutant IRAK2[E525A] or the catalytically inactive IRAK1[D359A] mutant. In bone marrow-derived macrophages (BMDMs), the IRAK2-TRAF6 interaction was required for the late (2-8 h) but not the early phase (0-2 h) of il6 and tnfa mRNA production, and hence for IL-6 and TNF-α secretion by TLR agonists that signal via MyD88. Loss of the IRAK2-TRAF6 interaction had little effect on the MyD88-dependent production of anti-inflammatory molecules produced during the early phase, such as Dual Specificity Phosphatase 1, and a modest effect on IL-10 secretion. The LPS/TLR4-stimulated production of il6 and tnfa mRNA and IL-6 and TNF-α secretion was hardly affected, because the Toll/IL-1R domain-containing adapter-inducing IFN-ß (TRIF) signaling pathway was used instead of the IRAK2-TRAF6 interaction to sustain late-phase mRNA production. IRAK1 catalytic activity was not rate limiting for il6, tnfa, or il10 mRNA production or the secretion of these cytokines by BMDMs, but IFN-ß mRNA induction by TLR7 and TLR9 agonists was greatly delayed in plasmacytoid dendritic cells (pDCs) from IRAK1[D359A] mice. In contrast, IFN-ß mRNA production was little affected in pDCs from IRAK2[E525A] mice, but subsequent IFN-α mRNA production and IFN-α secretion were reduced. IFN-ß and IFN-α production were abolished in pDCs from IRAK1[D359A] × IRAK2[E525A] double knock-in mice. Our results establish that the IRAK2-TRAF6 interaction is rate limiting for the late, but not the early phase of cytokine production in BMDM and pDCs, and that the IRAK2-TRAF6 interaction is needed to sustain IκB-inducing kinase ß activity during prolonged activation of the MyD88 signaling network. [corrected]


Asunto(s)
Citocinas/biosíntesis , Células Dendríticas/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Macrófagos/metabolismo , Transducción de Señal/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células Dendríticas/inmunología , Técnicas de Sustitución del Gen , Quinasa I-kappa B/inmunología , Quinasa I-kappa B/metabolismo , Immunoblotting , Inmunoprecipitación , Inflamación/inmunología , Inflamación/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Macrófagos/inmunología , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor 6 Asociado a Receptor de TNF/inmunología , Factor 6 Asociado a Receptor de TNF/metabolismo , Transfección
19.
J Immunol ; 190(9): 4736-41, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23526823

RESUMEN

Monocyte-derived macrophages (MDM) can polarize into different subsets depending on the environment and the activation signal to which they are submitted. Differentiation into macrophages allows HIV-1 strains to infect cells of the monocytic lineage. In this study, we show that culture of monocytes with a combination of IL-12 and IL-18 led to macrophage differentiation that was resistant to HIV-1 infection. In contrast, M-CSF-derived MDM were readily infected by HIV-1. When monocytes were differentiated in the presence of M-CSF and then further treated with IL-12/IL-18, cells became resistant to infection. The restriction on HIV-1 replication was not dependent on virus entry or coreceptor expression, as vesicular stomatitis virus-pseudotyped HIV-1 replication was also blocked by IL-12/IL-18. The HIV-1 restriction factor sterile α motif and HD domain-containing protein-1 (SAMHD1) was significantly overexpressed in IL-12/IL-18 MDM compared with M-CSF MDM, and degradation of SAMHD1 by RNA interference or viral-like particles carrying the lentiviral protein Vpx restored HIV-1 infectivity of IL-12/IL-18 MDM. SAMHD1 overexpression induced by IL-12/IL-18 was not dependent on IFN-γ. Thus, we conclude that IL-12 and IL-18 may contribute to the response against HIV-1 infection through the induction of restriction factors such as SAMHD1.


Asunto(s)
VIH-1/fisiología , Interleucina-12/genética , Interleucina-18/genética , Macrófagos/virología , Proteínas de Unión al GTP Monoméricas/genética , Replicación Viral/genética , Diferenciación Celular/genética , VIH-1/genética , VIH-1/metabolismo , Humanos , Interleucina-12/inmunología , Interleucina-12/metabolismo , Interleucina-18/inmunología , Interleucina-18/metabolismo , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/inmunología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/virología , Proteínas de Unión al GTP Monoméricas/inmunología , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteína 1 que Contiene Dominios SAM y HD , Regulación hacia Arriba
20.
Antiviral Res ; 97(2): 180-2, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23266292

RESUMEN

SAMHD1 (sterile alpha motif and histidine/aspartic acid (HD) domain-containing protein 1) has been identified as a novel HIV-1 restriction factor in myeloid cells and resting CD4+ T lymphocytes. SAMHD1 restriction is antagonized by the lentiviral protein Vpx. Here, we comment on the latest knowledge of SAMHD1 biology, focusing on how it regulates the pool of intracellular nucleotides to control HIV replication. We discuss how HIV restriction by SAMHD1 and viral counter-restriction mechanisms may suggest new strategies for therapeutic intervention.


Asunto(s)
VIH-1/patogenicidad , Interacciones Huésped-Patógeno , Proteínas de Unión al GTP Monoméricas/metabolismo , Nucleótidos/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Replicación Viral , VIH-1/inmunología , VIH-1/fisiología , Humanos , Proteína 1 que Contiene Dominios SAM y HD , Factores de Virulencia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA