Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Int J Mol Sci ; 25(10)2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38791357

RESUMEN

The lung is prone to infections from respiratory viruses such as Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). A challenge in combating these infections is the difficulty in targeting antiviral activity directly at the lung mucosal tract. Boosting the capability of the respiratory mucosa to trigger a potent immune response at the onset of infection could serve as a potential strategy for managing respiratory infections. This study focused on screening immunomodulators to enhance innate immune response in lung epithelial and immune cell models. Through testing various subfamilies and pathways of pattern recognition receptors (PRRs), the nucleotide-binding and oligomerization domain (NOD)-like receptor (NLR) family was found to selectively activate innate immunity in lung epithelial cells. Activation of NOD1 and dual NOD1/2 by the agonists TriDAP and M-TriDAP, respectively, increased the number of IL-8+ cells by engaging the NF-κB and interferon response pathways. Lung epithelial cells showed a stronger response to NOD1 and dual NOD1/2 agonists compared to control. Interestingly, a less-pronounced response to NOD1 agonists was noted in PBMCs, indicating a tissue-specific effect of NOD1 in lung epithelial cells without inducing widespread systemic activation. The specificity of the NOD agonist pathway was confirmed through gene silencing of NOD1 (siRNA) and selective NOD1 and dual NOD1/2 inhibitors in lung epithelial cells. Ultimately, activation induced by NOD1 and dual NOD1/2 agonists created an antiviral environment that hindered SARS-CoV-2 replication in vitro in lung epithelial cells.


Asunto(s)
COVID-19 , Células Epiteliales , Pulmón , Proteína Adaptadora de Señalización NOD1 , SARS-CoV-2 , Humanos , Células A549 , Antivirales/farmacología , COVID-19/inmunología , COVID-19/virología , Tratamiento Farmacológico de COVID-19 , Ácido Diaminopimélico/análogos & derivados , Ácido Diaminopimélico/farmacología , Células Epiteliales/virología , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Inmunidad Innata/efectos de los fármacos , Interleucina-8/metabolismo , Pulmón/inmunología , Pulmón/virología , Pulmón/metabolismo , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Adaptadora de Señalización NOD1/agonistas , Proteína Adaptadora de Señalización NOD2/agonistas , Proteína Adaptadora de Señalización NOD2/metabolismo , SARS-CoV-2/fisiología , SARS-CoV-2/inmunología , Transducción de Señal/efectos de los fármacos
2.
Int J Mol Sci ; 23(23)2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36499329

RESUMEN

HIV latent infection may be associated with disrupted viral RNA sensing, interferon (IFN) signaling, and/or IFN stimulating genes (ISG) activation. Here, we evaluated the use of compounds selectively targeting at the inhibitor of nuclear factor-κB (IκB) kinase (IKK) complex subunits and related kinases (TBK1) as a novel pathway to reverse HIV-1 latency in latently infected non-clonal lymphoid and myeloid cell in vitro models. IKK inhibitors (IKKis) triggered up to a 1.8-fold increase in HIV reactivation in both, myeloid and lymphoid cell models. The best-in-class IKKis, targeting TBK-1 (MRT67307) and IKKß (TCPA-1) respectively, were also able to significantly induce viral reactivation in CD4+ T cells from people living with HIV (PLWH) ex vivo. More importantly, although none of the compounds tested showed antiviral activity, the combination of the distinct IKKis with ART did not affect the latency reactivation nor blockade of HIV infection by ART. Finally, as expected, IKKis did not upregulate cell activation markers in primary lymphocytes and innate immune signaling was blocked, resulting in downregulation of inflammatory cytokines. Overall, our results support a dual role of IKKis as immune modulators being able to tackle the HIV latent reservoir in lymphoid and myeloid cellular models and putatively control the hyperinflammatory responses in chronic HIV-1 infection.


Asunto(s)
Infecciones por VIH , VIH-1 , Humanos , VIH-1/fisiología , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Latencia del Virus , Activación Viral , Linfocitos T CD4-Positivos
3.
Front Immunol ; 13: 1001068, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36131914

RESUMEN

The persistence of latent HIV reservoirs allows for viral rebound upon antiretroviral therapy interruption, hindering effective HIV-1 cure. Emerging evidence suggests that modulation of innate immune stimulation could impact viral latency and contribute to the clearing of HIV reservoir. Here, the latency reactivation capacity of a subclass of selective JAK2 inhibitors was characterized as a potential novel therapeutic strategy for HIV-1 cure. Notably, JAK2 inhibitors reversed HIV-1 latency in non-clonal lymphoid and myeloid in vitro models of HIV-1 latency and also ex vivo in CD4+ T cells from ART+ PWH, albeit its function was not dependent on JAK2 expression. Immunophenotypic characterization and whole transcriptomic profiling supported reactivation data, showing common gene expression signatures between latency reactivating agents (LRA; JAK2i fedratinib and PMA) in contrast to other JAK inhibitors, but with significantly fewer affected gene sets in the pathway analysis. In depth evaluation of differentially expressed genes, identified a significant upregulation of IRF7 expression despite the blockade of the JAK-STAT pathway and downregulation of proinflammatory cytokines and chemokines. Moreover, IRF7 expression levels positively correlated with HIV latency reactivation capacity of JAK2 inhibitors and also other common LRAs. Collectively, these results represent a promising step towards HIV eradication by demonstrating the potential of innate immune modulation for reducing the viral reservoir through a novel pathway driven by IRF7.


Asunto(s)
Infecciones por VIH , VIH-1 , Inhibidores de las Cinasas Janus , Citocinas/farmacología , Infecciones por VIH/tratamiento farmacológico , Humanos , Inhibidores de las Cinasas Janus/uso terapéutico , Quinasas Janus , Factores de Transcripción STAT , Transducción de Señal , Activación Viral , Latencia del Virus
4.
Eur J Med Chem ; 208: 112696, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-32883642

RESUMEN

We designed and synthesized 21 new indolylarylsulfones (IASs) as new HIV-1 NNRTIs. Among these, IAS 12 exhibited a remarkable antiviral activity against single and double mutants (K103N EC50 = <0.7 nM; Y181C EC50 = <0.7 nM; Y188L EC50 = 21.3 nM; K103N-Y181C EC50 = 6.2 nM), resulting equally or more active than previuosly reported IAS 6 and some approved anti-HIV-1 drugs. Docking and molecular dynamics simulations of compound 12 in complex with WT, Y181C, Y188L, K103N and K103N-Y181C RTs clarified a general binding mode that was consistent with biological results. Kinetic experiments disclosed that derivative 12 preferentially binds WT and K103N-Y181C RTs to binary and ternary complexes, respectively.


Asunto(s)
Fármacos Anti-VIH/farmacología , VIH-1/efectos de los fármacos , Indoles/farmacología , Inhibidores de la Transcriptasa Inversa/farmacología , Sulfonas/farmacología , Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/metabolismo , Línea Celular Tumoral , Diseño de Fármacos , Sinergismo Farmacológico , Transcriptasa Inversa del VIH/metabolismo , VIH-1/genética , Humanos , Indoles/síntesis química , Indoles/metabolismo , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Mutación , Unión Proteica , Inhibidores de la Transcriptasa Inversa/síntesis química , Inhibidores de la Transcriptasa Inversa/metabolismo , Relación Estructura-Actividad , Sulfonas/síntesis química , Sulfonas/metabolismo , Zidovudina/análogos & derivados , Zidovudina/farmacología
5.
Viruses ; 12(7)2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32640577

RESUMEN

HIV-1 infection requires life-long treatment and with 2.1 million new infections/year, faces the challenge of an increased rate of transmitted drug-resistant mutations. Therefore, a constant and timely effort is needed to identify new HIV-1 inhibitors active against drug-resistant variants. The ribonuclease H (RNase H) activity of HIV-1 reverse transcriptase (RT) is a very promising target, but to date, still lacks an efficient inhibitor. Here, we characterize the mode of action of N'-(2-hydroxy-benzylidene)-3,4,5-trihydroxybenzoylhydrazone (compound 13), an N-acylhydrazone derivative that inhibited viral replication (EC50 = 10 µM), while retaining full potency against the NNRTI-resistant double mutant K103N-Y181C virus. Time-of-addition and biochemical assays showed that compound 13 targeted the reverse-transcription step in cell-based assays and inhibited the RT-associated RNase H function, being >20-fold less potent against the RT polymerase activity. Docking calculations revealed that compound 13 binds within the RNase H domain in a position different from other selective RNase H inhibitors; site-directed mutagenesis studies revealed interactions with conserved amino acid within the RNase H domain, suggesting that compound 13 can be taken as starting point to generate a new series of more potent RNase H selective inhibitors active against circulating drug-resistant variants.


Asunto(s)
Fármacos Anti-VIH/uso terapéutico , VIH-1/efectos de los fármacos , Ribonucleasa H del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Fármacos Anti-VIH/farmacología , Sitios de Unión , Farmacorresistencia Viral , VIH-1/enzimología , Humanos , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Ribonucleasa H
7.
Cancers (Basel) ; 12(3)2020 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-32197329

RESUMEN

Sterile alpha motif and histidine-aspartic acid domain-containing protein 1 (SAMHD1) is a dNTP triphosphohydrolase involved in the regulation of the intracellular dNTP pool, linked to viral restriction, cancer development and autoimmune disorders. SAMHD1 function is regulated by phosphorylation through a mechanism controlled by cyclin-dependent kinases and tightly linked to cell cycle progression. Recently, SAMHD1 has been shown to decrease the efficacy of nucleotide analogs used as chemotherapeutic drugs. Here, we demonstrate that SAMHD1 can enhance or decrease the efficacy of various classes of anticancer drug, including nucleotide analogues, but also anti-folate drugs and CDK inhibitors. Importantly, we show that selective CDK4/6 inhibitors are pharmacological activators of SAMHD1 that act by inhibiting its inactivation by phosphorylation. Combinations of a CDK4/6 inhibitor with nucleoside or folate antimetabolites potently enhanced drug efficacy, resulting in highly synergic drug combinations (CI < 0.04). Mechanistic analyses reveal that cell cycle-controlled modulation of SAMHD1 function is the central process explaining changes in anticancer drug efficacy, therefore providing functional proof of the potential of CDK4/6 inhibitors as a new class of adjuvants to boost chemotherapeutic regimens. The evaluation of SAMHD1 expression in cancer tissues allowed for the identification of cancer types that would benefit from the pharmacological modulation of SAMHD1 function. In conclusion, these results indicate that the modulation of SAMHD1 function may represent a promising strategy for the improvement of current antimetabolite-based treatments.

8.
Sci Rep ; 9(1): 19848, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31882741

RESUMEN

Infection by human papillomavirus (HPV) alters the microenvironment of keratinocytes as a mechanism to evade the immune system. A-to-I editing by ADAR1 has been reported to regulate innate immunity in response to viral infections. Here, we evaluated the role of ADAR1 in HPV infection in vitro and in vivo. Innate immune activation was characterized in human keratinocyte cell lines constitutively infected or not with HPV. ADAR1 knockdown induced an innate immune response through enhanced expression of RIG-I-like receptors (RLR) signaling cascade, over-production of type-I IFNs and pro-inflammatory cytokines. ADAR1 knockdown enhanced expression of HPV proteins, a process dependent on innate immune function as no A-to-I editing could be identified in HPV transcripts. A genetic association study was performed in a cohort of HPV/HIV infected individuals followed for a median of 6 years (range 0.1-24). We identified the low frequency haplotype AACCAT significantly associated with recurrent HPV dysplasia, suggesting a role of ADAR1 in the outcome of HPV infection in HIV+ individuals. In summary, our results suggest that ADAR1-mediated innate immune activation may influence HPV disease outcome, therefore indicating that modification of innate immune effectors regulated by ADAR1 could be a therapeutic strategy against HPV infection.


Asunto(s)
Adenosina Desaminasa/genética , Coinfección/fisiopatología , Infecciones por VIH/fisiopatología , Infecciones por Papillomavirus/fisiopatología , Proteínas de Unión al ARN/genética , Adenosina Desaminasa/metabolismo , Adulto , Anciano , Línea Celular Tumoral , Coinfección/genética , Coinfección/virología , Femenino , Infecciones por VIH/genética , Infecciones por VIH/virología , Humanos , Sistema Inmunológico/metabolismo , Sistema Inmunológico/virología , Queratinocitos/metabolismo , Queratinocitos/virología , Masculino , Persona de Mediana Edad , Papillomaviridae/genética , Papillomaviridae/fisiología , Infecciones por Papillomavirus/virología , Polimorfismo de Nucleótido Simple , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Lesiones Precancerosas/fisiopatología , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/genética , Adulto Joven
9.
Antiviral Res ; 168: 18-27, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31077767

RESUMEN

Midostaurin is a multi-kinase inhibitor with antineoplastic activity. We assessed the capacity of midostaurin to affect early and late steps of HIV-1 infection and to reactivate HIV-1 latently infected cells, alone or in combination with histone deacetylase inhibitors (HDACi) known to act as latency-reversing agents (LRA). Acute HIV-1 infection was assessed by flow cytometry in three cell types treated with midostaurin in the presence or absence of SAMHD1. Non-infected cells were treated with midostaurin and harvested for Western blot analysis. Macrophage infections were also measured by quantitative RT-PCR. HIV-1 latency reactivation was assessed in several latency models. Midostaurin induced G2/M arrest and inhibited CDK2, preventing the phosphorylation of SAMHD1 associated to inhibition of its dNTPase activity. In the presence of SAMHD1, midostaurin blocked HIV-1 DNA formation and viral replication. However, following Vpx-mediated SAMHD1 degradation, midostaurin increased viral transcripts and virus replication. In three out of four HIV-1 latency models, including primary CD4+ T cells, midostaurin effectively reversed HIV-1 latency and was synergistic in combination with LRA vorinostat and panobinostat. Our study describes a dual effect for midostaurin in HIV-1 infection, antiviral or proviral depending on SAMHD1 activation, and highlights a role for active SAMHD1 in regulating the activity of potential HIV-1 latency reversal agents.


Asunto(s)
Infecciones por VIH/virología , VIH-1/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Estaurosporina/análogos & derivados , Latencia del Virus/efectos de los fármacos , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/virología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Células Cultivadas , VIH-1/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/virología , Fosforilación/efectos de los fármacos , Proteína 1 que Contiene Dominios SAM y HD/antagonistas & inhibidores , Proteína 1 que Contiene Dominios SAM y HD/genética , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Estaurosporina/farmacología , Activación Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
10.
Int J STD AIDS ; 30(1): 56-63, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30170532

RESUMEN

Currently, Papanicolaou smears are proposed at three-year intervals for cervical screening to all women living with HIV. The aim of this retrospective cohort study was to provide data on the incidence of cervical high-grade squamous intraepithelial lesions (HSIL) in cervical smear confirmed by histology in HIV-1-infected women (two consecutive normal Papanicolaou smears at baseline) after a long-term follow-up. Sixty-seven women (recruited between March 1999 and January 2003) were analyzed. The median period of follow-up was 13.2 years (range: 7.4-17.1 years) with a total of 583 Papanicolaou smears. Twenty-seven percent of these HIV-1-infected women had poorly-controlled HIV. Cumulative incidence of HSIL was 18% (12/67; 95%CI: 11-29%) of which one was an invasive squamous cell carcinoma and two were carcinoma in situ. These women had not been well-engaged with the annual Papanicolaou smear screening program and had poor adherence to antiretroviral therapy. Development of HSIL was associated with high-risk-HPV infection (OR: 14.9; 95%CI: 3.0, 75.1). At last Papanicolaou smear, prevalence of high-risk-HPV infection was 30% (20/66, 95%CI: 21-42%). In conclusion, the incidence of cervical HSIL in HIV-1-infected women with poor antiretroviral therapy adherence or poor immunological status reinforces the need to identify those HIV-1-infected women at risk of developing cervical cancer.


Asunto(s)
Infecciones por VIH/complicaciones , Infecciones por VIH/epidemiología , Lesiones Intraepiteliales Escamosas de Cuello Uterino/epidemiología , Displasia del Cuello del Útero/epidemiología , Neoplasias del Cuello Uterino/epidemiología , Adulto , Fármacos Anti-VIH/uso terapéutico , Terapia Antirretroviral Altamente Activa/métodos , Femenino , Estudios de Seguimiento , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Humanos , Incidencia , Cumplimiento de la Medicación , Persona de Mediana Edad , Infecciones por Papillomavirus/epidemiología , Estudios Retrospectivos , Asunción de Riesgos , España/epidemiología , Lesiones Intraepiteliales Escamosas de Cuello Uterino/virología , Neoplasias del Cuello Uterino/virología , Displasia del Cuello del Útero/virología
11.
J Med Chem ; 62(2): 604-621, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30525601

RESUMEN

Conformational restriction applied to dihydrobenzylpyrimidin-4-(3 H)-ones (DABOs) by the intoduction of a methyl group at the α-benzylic position is known to massively improve the anti-HIV-1 activity of these compounds. Here, we report the effects of methoxy substitution at the α-benzylic position in S-, NH-, and N, N-DABOs carrying 2,6-difluoro, 2-chloro-6-fluoro, or 2,6-dichloro substituted benzyl moieties. The various α-methoxy DABO series (12-14) present different SAR at the dihalo benzyl substitution, with the most potent compounds (12d,e and 13c) showing similar (picomolar/nanomolar) anti-HIV-1 potency as the corresponding α-methyl analogues against wt HIV-1, and 10-100-fold increased potency (up to low nanomolar) against clinically relevant K103N, Y181C, Y188L, IRLL98, and K103N+Y181C HIV-1 mutant strains, highlighting the importance of the α-methoxy substitution to provide highly efficient DABOs as "second generation" NNRTIs. HPLC enantioseparation of three of the most potent derivatives (12d, 13c, and 14c) provided single enantiomers with significant enantioselectivity in HIV-1 inhibition. Computational studies allowed to correlate the best antiviral activity with the ( R) absolute configuration at the α-methoxy stereogenic center.


Asunto(s)
Fármacos Anti-VIH/química , Pirimidinonas/química , Fármacos Anti-VIH/metabolismo , Fármacos Anti-VIH/farmacología , Sitios de Unión , Línea Celular , Farmacorresistencia Viral/efectos de los fármacos , Transcriptasa Inversa del VIH/antagonistas & inhibidores , Transcriptasa Inversa del VIH/genética , Transcriptasa Inversa del VIH/metabolismo , VIH-1/efectos de los fármacos , VIH-1/genética , Humanos , Simulación del Acoplamiento Molecular , Mutación , Estructura Terciaria de Proteína , Pirimidinonas/metabolismo , Pirimidinonas/farmacología , Estereoisomerismo , Relación Estructura-Actividad
12.
Nat Commun ; 9(1): 2739, 2018 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-30013105

RESUMEN

CD32 has been shown to be preferentially expressed in latently HIV-1-infected cells in an in vitro model of quiescent CD4 T cells. Here we show that stimulation of CD4+ T cells with IL-2, IL-7, PHA, and anti-CD3/CD28 antibodies induces T-cell proliferation, co-expression of CD32 and the activation of the markers HLA-DR and CD69. HIV-1 infection increases CD32 expression. 79.2% of the CD32+/CD4+ T cells from HIV+ individuals under antiretroviral treatment were HLA-DR+. Resting CD4+ T cells infected in vitro generally results in higher integration of provirus. We observe no difference in provirus integration or replication-competent inducible latent HIV-1 in CD32+ or CD32- CD4+ T cells from HIV+ individuals. Our results demonstrate that CD32 expression is a marker of CD4+ T cell activation in HIV+ individuals and raises questions regarding the immune resting status of CD32+ cells harboring HIV-1 proviruses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/genética , VIH-1/inmunología , Interacciones Huésped-Patógeno/inmunología , Activación de Linfocitos/genética , Receptores de IgG/genética , Adulto , Anticuerpos Monoclonales/farmacología , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/inmunología , Antígenos CD28/antagonistas & inhibidores , Antígenos CD28/genética , Antígenos CD28/inmunología , Complejo CD3/antagonistas & inhibidores , Complejo CD3/genética , Complejo CD3/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/virología , Expresión Génica , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/genética , Antígenos HLA-DR/genética , Antígenos HLA-DR/inmunología , Humanos , Interleucina-2/farmacología , Interleucina-7/farmacología , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Activación de Linfocitos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Fitohemaglutininas/farmacología , Cultivo Primario de Células , Provirus/genética , Provirus/inmunología , Receptores de IgG/inmunología , Integración Viral
13.
Antiviral Res ; 156: 116-127, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29906476

RESUMEN

The hepatitis C virus (HCV) is a globally prevalent infectious pathogen. As many as 80% of people infected with HCV do not control the virus and develop a chronic infection. Response to interferon (IFN) therapy is widely variable in chronic HCV infected patients, suggesting that HCV has evolved mechanisms to suppress and evade innate immunity responsible for its control and elimination. Adenosine deaminase acting on RNA 1 (ADAR1) is a relevant factor in the regulation of the innate immune response. The loss of ADAR1 RNA-editing activity and the resulting loss of inosine bases in RNA are critical in producing aberrant RLR-mediated innate immune response, mediated by RNA sensors MDA5 and RIG-I. Here, we describe ADAR1 role as a regulator of innate and antiviral immune function in HCV infection, both in vitro and in patients. Polymorphisms within ADAR1 gene were found significantly associated to poor clinical outcome to HCV therapy and advanced liver fibrosis in a cohort of HCV and HIV-1 coinfected patients. Moreover, ADAR1 knockdown in primary macrophages and Huh7 hepatoma cells enhanced IFN and IFN stimulated gene expression and increased HCV replication in vitro. Overall, our results demonstrate that ADAR1 regulates innate immune signaling and is an important contributor to the outcome of the HCV virus-host interaction. ADAR1 is a potential target to boost antiviral immune response in HCV infection.


Asunto(s)
Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Predisposición Genética a la Enfermedad , Hepacivirus/inmunología , Hepatitis C/inmunología , Interacciones Huésped-Patógeno , Inmunidad Innata , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Células Cultivadas , Humanos , Polimorfismo Genético
14.
Sci Rep ; 7(1): 13339, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-29042669

RESUMEN

ADAR1-dependent A-to-I editing has recently been recognized as a key process for marking dsRNA as self, therefore, preventing innate immune activation and affecting the development and resolution of immune-mediated diseases and infections. Here, we have determined the role of ADAR1 as a regulator of innate immune activation and modifier of viral susceptibility in primary myeloid and lymphoid cells. We show that ADAR1 knockdown significantly enhanced interferon, cytokine and chemokine production in primary macrophages that function as antiviral paracrine factors, rendering them resistant to HIV-1 infection. ADAR1 knockdown induced deregulation of the RLRs-MAVS signaling pathway, by increasing MDA5, RIG-I, IRF7 and phospho-STAT1 expression, an effect that was partially rescued by pharmacological blockade of the pathway. In summary, our results demonstrate a role of ADAR1 in regulating innate immune function in primary macrophages, suggesting that macrophages may play an essential role in disease associated to ADAR1 dysfunction. We also show that viral inhibition is exclusively dependent on innate immune activation consequence of ADAR1 knockdown, pointing towards ADAR1 as a potential target to boost antiviral immune response.


Asunto(s)
Adenosina Desaminasa/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Macrófagos/inmunología , Macrófagos/metabolismo , Edición de ARN , Proteínas de Unión al ARN/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , ARN Helicasas DEAD-box/metabolismo , Técnicas de Silenciamiento del Gen , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/fisiología , Humanos , Interferón Tipo I/metabolismo , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Macrófagos/virología , Transducción de Señal , Virosis/etiología , Replicación Viral
15.
Artículo en Inglés | MEDLINE | ID: mdl-28874382

RESUMEN

The persistence of HIV despite suppressive antiretroviral therapy is a major roadblock to HIV eradication. Current strategies focused on inducing the expression of latent HIV fail to clear the persistent reservoir, prompting the development of new approaches for killing HIV-positive cells. Recently, acitretin was proposed as a pharmacological enhancer of the innate cellular defense network that led to virus reactivation and preferential death of infected cells. We evaluated the capacity of acitretin to reactivate and/or to facilitate immune-mediated clearance of HIV-positive cells. Acitretin did not induce HIV reactivation in latently infected cell lines (J-Lat and ACH-2). We could observe only modest induction of HIV reactivation by acitretin in latently green fluorescent protein-HIV-infected Jurkat cells, comparable to suboptimal concentrations of vorinostat, a known latency-reversing agent (LRA). Acitretin induction was insignificant, however, compared to optimal concentrations of LRAs. Acitretin failed to reactivate HIV in a model of latently infected primary CD4+ T cells but induced retinoic acid-inducible gene I (RIG-I) and mitochondrial antiviral signaling (MAVS) expression in infected and uninfected cells, confirming the role of acitretin as an innate immune modulator. However, this effect was not associated with selective killing of HIV-positive cells. In conclusion, acitretin-mediated stimulation of the RIG-I pathway for HIV reactivation is modest and thus may not meaningfully affect the HIV reservoir. Stimulation of the RIG-I-dependent interferon (IFN) cascade by acitretin may not significantly affect the selective destruction of latently infected HIV-positive cells.


Asunto(s)
Acitretina/farmacología , Infecciones por VIH/inmunología , Inmunidad Innata/efectos de los fármacos , Factores Inmunológicos/farmacología , Latencia del Virus/efectos de los fármacos , Proteína 58 DEAD Box/metabolismo , Infecciones por VIH/tratamiento farmacológico , VIH-1/patogenicidad , VIH-1/fisiología , Humanos , Receptores Inmunológicos , Transducción de Señal/efectos de los fármacos
16.
J Med Chem ; 60(15): 6528-6547, 2017 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-28628334

RESUMEN

We designed and synthesized a series of chiral indolyarylsulfones (IASs) as new HIV-1 NNRTIs. The new IASs 8-37 showed potent inhibition of the HIV-1 WT NL4-3 strain and of the mutant K103N, Y181C, Y188L, and K103N-Y181C HIV-1 strains. Six racemic mixtures, 8, 23-25, 31, and 33, were separated at semipreparative level into their pure enantiomers. The (R)-8 enantiomer bearing the chiral (α-methylbenzyl) was superior to the (S)-counterpart. IAS derivatives bearing the (S) alanine unit, (S)-23, (S,R)-25, (S)-31, and (S)-33, were remarkably more potent than the corresponding (R)-enantiomers. Compound 23 protected hippocampal neuronal cells from the excitotoxic insult, while efavirenz (EFV) did not contrast the neurotoxic effect of glutamate. The present results highlight the chiral IASs as new NNRTIs with improved resistance profile against the mutant HIV-1 strains and reduced neurotoxic effects.


Asunto(s)
Fármacos Anti-VIH/farmacología , VIH-1/efectos de los fármacos , Indoles/farmacología , Inhibidores de la Transcriptasa Inversa/farmacología , Sulfonas/farmacología , Animales , Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/toxicidad , Células Cultivadas , Ácido Glutámico/toxicidad , VIH-1/genética , Humanos , Indoles/síntesis química , Indoles/toxicidad , Lipopolisacáridos/farmacología , Ratones Endogámicos C57BL , Microglía/citología , Microglía/efectos de los fármacos , Simulación del Acoplamiento Molecular , Mutación , Neuronas/citología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/toxicidad , Inhibidores de la Transcriptasa Inversa/síntesis química , Inhibidores de la Transcriptasa Inversa/toxicidad , Estereoisomerismo , Relación Estructura-Actividad , Sulfonas/síntesis química , Sulfonas/toxicidad
17.
Curr Opin Virol ; 24: 46-54, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28437703

RESUMEN

The ability of some viruses to establish latently infected chronic reservoirs that escape to immune control becomes a major roadblock that impedes the cure of these infections. Therefore, new alternatives are needed to pursuit the eradication of viral persistent infections. Gene silencing technologies are in constant evolution and provide an outstanding sequence specificity that allows targeting any coding sequence of interest. Here we provide an overview of the development of gene silencing technologies ranging from initially RNA interference to the recently developed CRISPR/Cas9 and their potential as new antiviral strategies focusing on the eradication of HIV.


Asunto(s)
Antivirales/administración & dosificación , Edición Génica , Silenciador del Gen , Virosis/tratamiento farmacológico , Animales , Antivirales/uso terapéutico , Sistemas CRISPR-Cas , Desoxirribonucleasas/genética , Terapia Genética , Genoma Viral , Humanos , Ratones , Interferencia de ARN , Virosis/genética , Virosis/virología
18.
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
19.
PLoS Pathog ; 12(8): e1005829, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27541004

RESUMEN

Macrophages are a heterogeneous cell population strongly influenced by differentiation stimuli that become susceptible to HIV-1 infection after inactivation of the restriction factor SAMHD1 by cyclin-dependent kinases (CDK). Here, we have used primary human monocyte-derived macrophages differentiated through different stimuli to evaluate macrophage heterogeneity on cell activation and proliferation and susceptibility to HIV-1 infection. Stimulation of monocytes with GM-CSF induces a non-proliferating macrophage population highly restrictive to HIV-1 infection, characterized by the upregulation of the G1/S-specific cyclin D2, known to control early steps of cell cycle progression. Knockdown of cyclin D2, enhances HIV-1 replication in GM-CSF macrophages through inactivation of SAMHD1 restriction factor by phosphorylation. Co-immunoprecipitation experiments show that cyclin D2 forms a complex with CDK4 and p21, a factor known to restrict HIV-1 replication by affecting the function of the downstream cascade that leads to SAMHD1 deactivation. Thus, we demonstrate that cyclin D2 acts as regulator of cell cycle proteins affecting SAMHD1-mediated HIV-1 restriction in non-proliferating macrophages.


Asunto(s)
Ciclina D2/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Macrófagos/inmunología , Animales , Proliferación Celular , Quinasa 4 Dependiente de la Ciclina/inmunología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Macrófagos/virología , Ratones , Proteínas de Unión al GTP Monoméricas/inmunología , Proteína 1 que Contiene Dominios SAM y HD
20.
Antiviral Res ; 133: 145-55, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27510577

RESUMEN

HIV-1 infection is thought to impair type I interferon (IFN-I) production in macrophages, a cell type that is also relatively resistant to HIV-1 cytotoxic effects. Here, we show that monocyte differentiation into macrophages by M-CSF led to cell proliferation and susceptibility to HIV-1 infection that induced cell cycle arrest and increased cell death. Established HIV-1 infection of monocyte-derived macrophages induced the upregulation of the pattern recognition receptors MDA5 and Rig-I that serve as virus sensors; production of interferon-ß, and transcription of interferon-stimulated genes including CXCL10. Infected macrophages showed increased expression of p21 and subsequent inactivation of cyclin-CDK2 activity leading to a hypo-phosphorylated active retinoblastoma protein (pRb) and deactivation of E2F1-dependent transcription and CDK1 downregulation. Additionally, HIV-1 infection limited deoxynucleotide pool by downregulation of the ribonucleotide reductase subunit R2 (RNR2) and reactivation of the HIV-1 restriction factor SAMHD1 together with increased cell death. In conclusion, HIV-1 induced an innate antiviral mechanism associated to IFN-I production, interferon stimulated gene activation, and p21-mediated G2/M arrest leading to elevated levels of cell death in monocyte derived macrophages. Upregulation of MDA5 and Rig-I may serve as targets for the development of antiviral strategies leading to the elimination of HIV-1 infected cells.


Asunto(s)
Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/inmunología , Inmunidad Innata , Macrófagos/inmunología , Macrófagos/virología , Antivirales/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Citocinas/metabolismo , Regulación de la Expresión Génica , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , VIH-1/efectos de los fármacos , Humanos , Inmunidad Innata/genética , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interferón Tipo I/metabolismo , Leucocitos Mononucleares , Activación de Macrófagos/inmunología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA