Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
MAbs ; 16(1): 2297451, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38170638

RESUMEN

The development of specific, safe, and potent monoclonal antibodies (Abs) has led to novel therapeutic options for infectious disease. In addition to preventing viral infection through neutralization, Abs can clear infected cells and induce immunomodulatory functions through engagement of their crystallizable fragment (Fc) with complement proteins and Fc receptors on immune cells. Little is known about the role of Fc effector functions of neutralizing Abs in the context of encephalitic alphavirus infection. To determine the role of Fc effector function in therapeutic efficacy against Venezuelan equine encephalitis virus (VEEV), we compared the potently neutralizing anti-VEEV human IgG F5 (hF5) Ab with intact Fc function (hF5-WT) or containing the loss of function Fc mutations L234A and L235A (hF5-LALA) in the context of VEEV infection. We observed significantly reduced binding to complement and Fc receptors, as well as differential in vitro kinetics of Fc-mediated cytotoxicity for hF5-LALA compared to hF5-WT. The in vivo efficacy of hF5-LALA was comparable to hF5-WT at -24 and + 24 h post infection, with both Abs providing high levels of protection. However, when hF5-WT and hF5-LALA were administered + 48 h post infection, there was a significant decrease in the therapeutic efficacy of hF5-LALA. Together these results demonstrate that optimal therapeutic Ab treatment of VEEV, and possibly other encephalitic alphaviruses, requires neutralization paired with engagement of immune effectors via the Fc region.


Asunto(s)
Anticuerpos Antivirales , Virus de la Encefalitis Equina Venezolana , Animales , Caballos , Humanos , Virus de la Encefalitis Equina Venezolana/genética , Anticuerpos Neutralizantes/farmacología , Receptores Fc , Inmunoglobulina G
2.
MAbs ; 13(1): 1958663, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34348076

RESUMEN

The respiratory virus responsible for coronavirus disease 2019 (COVID-19), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has affected nearly every aspect of life worldwide, claiming the lives of over 3.9 million people globally, at the time of this publication. Neutralizing humanized nanobody (VHH)-based antibodies (VHH-huFc) represent a promising therapeutic intervention strategy to address the current SARS-CoV-2 pandemic and provide a powerful toolkit to address future virus outbreaks. Using a synthetic, high-diversity VHH bacteriophage library, several potent neutralizing VHH-huFc antibodies were identified and evaluated for their capacity to tightly bind to the SARS-CoV-2 receptor-binding domain, to prevent binding of SARS-CoV-2 spike (S) to the cellular receptor angiotensin-converting enzyme 2, and to neutralize viral infection. Preliminary preclinical evaluation of multiple VHH-huFc antibody candidates demonstrate that they are prophylactically and therapeutically effective in vivo against wildtype SARS-CoV-2. The identified and characterized VHH-huFc antibodies described herein represent viable candidates for further preclinical evaluation and another tool to add to our therapeutic arsenal to address the COVID-19 pandemic.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19 , SARS-CoV-2/inmunología , Anticuerpos de Dominio Único/inmunología , Humanos
3.
Front Mol Biosci ; 8: 678701, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34327214

RESUMEN

A rapid response is necessary to contain emergent biological outbreaks before they can become pandemics. The novel coronavirus (SARS-CoV-2) that causes COVID-19 was first reported in December of 2019 in Wuhan, China and reached most corners of the globe in less than two months. In just over a year since the initial infections, COVID-19 infected almost 100 million people worldwide. Although similar to SARS-CoV and MERS-CoV, SARS-CoV-2 has resisted treatments that are effective against other coronaviruses. Crystal structures of two SARS-CoV-2 proteins, spike protein and main protease, have been reported and can serve as targets for studies in neutralizing this threat. We have employed molecular docking, molecular dynamics simulations, and machine learning to identify from a library of 26 million molecules possible candidate compounds that may attenuate or neutralize the effects of this virus. The viability of selected candidate compounds against SARS-CoV-2 was determined experimentally by biolayer interferometry and FRET-based activity protein assays along with virus-based assays. In the pseudovirus assay, imatinib and lapatinib had IC50 values below 10 µM, while candesartan cilexetil had an IC50 value of approximately 67 µM against Mpro in a FRET-based activity assay. Comparatively, candesartan cilexetil had the highest selectivity index of all compounds tested as its half-maximal cytotoxicity concentration 50 (CC50) value was the only one greater than the limit of the assay (>100 µM).

4.
J Funct Morphol Kinesiol ; 5(3)2020 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-33467269

RESUMEN

The purpose of this study was to investigate balance and motor control in dancers and non-dancers with different foot positions. Physically active female dancers (n = 11) and non-dancers (n = 9) randomly completed two balance tests in a single visit: 1) Y-balance test (YBT), and 2) motor control test (MCT). Each test was completed with two different foot positions: 1) first ballet position in which heels were touching and feet were externally rotated to 140 degrees, and 2) sixth ballet position in which heels were spaced 10 cm apart and forward parallel. For the YBT, participants completed three attempts at anterior, posteromedial, and posterolateral reaches, which were averaged and standardized to limb length for a composite score. For the MCT, participants completed a multi-directional target test on a Biosway balance system, and accuracy and time to completion were analyzed. Findings revealed no differences in YBT score (p = 0.255), MCT score (p = 0.383), or MCT time (p = 0.306) between groups in the sixth position. However, dancers displayed better YBT scores (p = 0.036), MCT scores (p = 0.020), and faster MCT times (p = 0.009) in the first position. Results suggest that superior balance and motor control in dancers may be limited to less innate dance-specific foot positions.

5.
Anal Methods ; 11(5): 559-565, 2019 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-31565079

RESUMEN

The discovery of the RNA-guided DNA nuclease CRISPR-Cas9 has enabled the targeted editing of genomes from diverse organisms, but the permanent and inheritable nature of genome modification also poses immense risks. The potential for accidental exposure, malicious use, or undesirable persistence of Cas9 therapeutics and off-target genome effects highlight the need for detection assays. Here we report a centrifugal microfluidic platform for the measurement of both Cas9 protein levels and nuclease activity. Because Cas9 from many bacterial species have been adapted for biotechnology applications, we developed the capability to detect Cas9 from the widely-used S. pyogenes, as well as S. aureus, N. meningitides, and S. thermophilus using commercially-available antibodies. Further, we show that the phage-derived anti-CRISPR protein AcrIIC1, which binds to Cas9 from several species, can be used as a capture reagent to broaden the species range of detection. As genome modification generally requires Cas9 nuclease activity, a fluorescence-based sedimentation nuclease assay was also incorporated to allow the sensitive and simultaneous measurement of both Cas9 protein and activity in a single biological sample.

6.
Anal Chem ; 90(11): 6913-6921, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29756770

RESUMEN

The RNA-guided DNA nuclease Cas9 is now widely used for the targeted modification of genomes of human cells and various organisms. Despite the extensive use of Clustered Regularly Interspaced Palindromic Repeats (CRISPR) systems for genome engineering and the rapid discovery and engineering of new CRISPR-associated nucleases, there are no high-throughput assays for measuring enzymatic activity. The current laboratory and future therapeutic uses of CRISPR technology have a significant risk of accidental exposure or clinical off-target effects, underscoring the need for therapeutically effective inhibitors of Cas9. Here, we develop a fluorescence assay for monitoring Cas9 nuclease activity and demonstrate its utility with S. pyogenes (Spy), S. aureus (Sau), and C. jejuni (Cje) Cas9. The assay was validated by quantitatively profiling the species specificity of published anti-CRISPR (Acr) proteins, confirming the reported inhibition of Spy Cas9 by AcrIIA4 and Cje Cas9 by AcrIIC1 and no inhibition of Sau Cas9 by either anti-CRISPR. To identify drug-like inhibitors, we performed a screen of 189 606 small molecules for inhibition of Spy Cas9. Of 437 hits (0.2% hit rate), six were confirmed as Cas9 inhibitors in a direct gel electrophoresis secondary assay. The high-throughput nature of this assay makes it broadly applicable for the discovery of additional Cas9 inhibitors or the characterization of Cas9 enzyme variants.


Asunto(s)
Proteína 9 Asociada a CRISPR/análisis , Ensayos Analíticos de Alto Rendimiento , Espectrometría de Fluorescencia , Proteína 9 Asociada a CRISPR/metabolismo , Campylobacter jejuni/enzimología , Humanos , Staphylococcus aureus/enzimología , Streptococcus pyogenes/enzimología
7.
Sci Rep ; 7(1): 15586, 2017 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-29138425

RESUMEN

The high-affinity receptor for IgE expressed on the surface of mast cells and basophils interacts with antigens, via bound IgE antibody, and triggers secretion of inflammatory mediators that contribute to allergic reactions. To understand how past inputs (memory) influence future inflammatory responses in mast cells, a microfluidic device was used to precisely control exposure of cells to alternating stimulatory and non-stimulatory inputs. We determined that the response to subsequent stimulation depends on the interval of signaling quiescence. For shorter intervals of signaling quiescence, the second response is blunted relative to the first response, whereas longer intervals of quiescence induce an enhanced second response. Through an iterative process of computational modeling and experimental tests, we found that these memory-like phenomena arise from a confluence of rapid, short-lived positive signals driven by the protein tyrosine kinase Syk; slow, long-lived negative signals driven by the lipid phosphatase Ship1; and slower degradation of Ship1 co-factors. This work advances our understanding of mast cell signaling and represents a generalizable approach for investigating the dynamics of signaling systems.


Asunto(s)
Inflamación/inmunología , Mastocitos/inmunología , Receptores de IgE/inmunología , Transducción de Señal/inmunología , Animales , Anticuerpos/inmunología , Antígenos/inmunología , Basófilos/inmunología , Humanos , Inflamación/genética , Inflamación/metabolismo , Dispositivos Laboratorio en un Chip , Mastocitos/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/inmunología , Receptores de IgE/genética , Transducción de Señal/genética , Quinasa Syk/genética , Quinasa Syk/inmunología
8.
J Virol ; 90(16): 7084-7097, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27226375

RESUMEN

UNLABELLED: Rift Valley fever virus (RVFV) is an arbovirus within the Bunyaviridae family capable of causing serious morbidity and mortality in humans and livestock. To identify host factors involved in bunyavirus replication, we employed genome-wide RNA interference (RNAi) screening and identified 381 genes whose knockdown reduced infection. The Wnt pathway was the most represented pathway when gene hits were functionally clustered. With further investigation, we found that RVFV infection activated Wnt signaling, was enhanced when Wnt signaling was preactivated, was reduced with knockdown of ß-catenin, and was blocked using Wnt signaling inhibitors. Similar results were found using distantly related bunyaviruses La Crosse virus and California encephalitis virus, suggesting a conserved role for Wnt signaling in bunyaviral infection. We propose a model where bunyaviruses activate Wnt-responsive genes to regulate optimal cell cycle conditions needed to promote efficient viral replication. The findings in this study should aid in the design of efficacious host-directed antiviral therapeutics. IMPORTANCE: RVFV is a mosquito-borne bunyavirus that is endemic to Africa but has demonstrated a capacity for emergence in new territories (e.g., the Arabian Peninsula). As a zoonotic pathogen that primarily affects livestock, RVFV can also cause lethal hemorrhagic fever and encephalitis in humans. Currently, there are no treatments or fully licensed vaccines for this virus. Using high-throughput RNAi screening, we identified canonical Wnt signaling as an important host pathway regulating RVFV infection. The beneficial role of Wnt signaling was observed for RVFV, along with other disparate bunyaviruses, indicating a conserved bunyaviral replication mechanism involving Wnt signaling. These studies supplement our knowledge of the fundamental mechanisms of bunyavirus infection and provide new avenues for countermeasure development against pathogenic bunyaviruses.


Asunto(s)
Genoma Viral/genética , Interferencia de ARN , Fiebre del Valle del Rift/metabolismo , Virus de la Fiebre del Valle del Rift/fisiología , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Células A549 , Animales , Western Blotting , Células Cultivadas , Chlorocebus aethiops , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fiebre del Valle del Rift/genética , Fiebre del Valle del Rift/virología , Células Vero , Replicación Viral , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/genética , beta Catenina/antagonistas & inhibidores , beta Catenina/genética
9.
J Biol Chem ; 288(12): 8702-8711, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23382389

RESUMEN

The arenavirus nucleoprotein (NP) can suppress induction of type I interferon (IFN). This anti-IFN activity is thought to be shared by all arenaviruses with the exception of Tacaribe virus (TCRV). To identify the TCRV NP amino acid residues that prevent its IFN-countering ability, we created a series of NP chimeras between residues of TCRV NP and Pichinde virus (PICV) NP, an arenavirus NP with potent anti-IFN function. Chimera NP analysis revealed that a minimal four amino acid stretch derived from PICV NP could impart efficient anti-IFN activity to TCRV NP. Strikingly, the TCRV NP gene cloned and sequenced from viral stocks obtained through National Institutes of Health Biodefense and Emerging Infections (BEI) resources deviated from the reference sequence at this particular four-amino acid region, GPPT (GenBank KC329849) versus DLQL (GenBank NC004293), respectively at residues 389-392. When efficiently expressed in cells through codon-optimization, TCRV NP containing the GPPT residues rescued the antagonistic IFN function. Consistent with cell expression results, TCRV infection did not stimulate an IFNß response early in infection in multiple cells types (e.g. A549, P388D1), and IRF-3 was not translocated to the nucleus in TCRV-infected A549 cells. Collectively, these data suggest that certain TCRV strain variants contain the important NP amino acids necessary for anti-IFN activity.


Asunto(s)
Arenavirus del Nuevo Mundo/fisiología , Interferón beta/metabolismo , Nucleoproteínas/química , Proteínas Recombinantes de Fusión/química , Proteínas Virales/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Arenavirus del Nuevo Mundo/inmunología , Núcleo Celular/metabolismo , Chlorocebus aethiops , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/genética , Ratones , Datos de Secuencia Molecular , Nucleoproteínas/biosíntesis , Nucleoproteínas/inmunología , Regiones Promotoras Genéticas , Transporte de Proteínas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/inmunología , Activación Transcripcional , Células Vero , Proteínas Virales/biosíntesis , Proteínas Virales/inmunología
10.
Lab Chip ; 13(5): 811-7, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23361404

RESUMEN

RNA interference (RNAi) is a powerful tool for functional genomics with the capacity to comprehensively analyze host-pathogen interactions. High-throughput RNAi screening is used to systematically perturb cellular pathways and discover therapeutic targets, but the method can be tedious and requires extensive capital equipment and expensive reagents. To aid in the development of an inexpensive miniaturized RNAi screening platform, we have developed a two part microfluidic system for patterning and screening gene targets on-chip to examine cellular pathways involved in virus entry and infection. First, a multilayer polydimethylsiloxane (PDMS)-based spotting device was used to array siRNA molecules into 96 microwells targeting markers of endocytosis, along with siRNA controls. By using a PDMS-based spotting device, we remove the need for a microarray printer necessary to perform previously described small scale (e.g. cellular microarrays) and microchip-based RNAi screening, while still minimizing reagent usage tenfold compared to conventional screening. Second, the siRNA spotted array was transferred to a reversibly sealed PDMS-based screening platform containing microchannels designed to enable efficient cell loading and transfection of mammalian cells while preventing cross-contamination between experimental conditions. Validation of the screening platform was examined using Vesicular stomatitis virus and emerging pathogen Rift Valley fever virus, which demonstrated virus entry pathways of clathrin-mediated endocytosis and caveolae-mediated endocytosis, respectively. The techniques here are adaptable to other well-characterized infection pathways with a potential for large scale screening in high containment biosafety laboratories.


Asunto(s)
Técnicas Analíticas Microfluídicas/métodos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Virus de la Fiebre del Valle del Rift/fisiología , Vesiculovirus/fisiología , Caveolas/metabolismo , Caveolina 1/antagonistas & inhibidores , Caveolina 1/genética , Caveolina 1/metabolismo , Dimetilpolisiloxanos/química , Dinamina II/antagonistas & inhibidores , Dinamina II/genética , Dinamina II/metabolismo , Endocitosis , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Técnicas Analíticas Microfluídicas/instrumentación , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , ARN Interferente Pequeño/química , Transfección , Internalización del Virus , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/metabolismo
11.
J Virol ; 86(23): 12954-70, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22993156

RESUMEN

Rift Valley fever virus (RVFV) is a zoonotic pathogen capable of causing serious morbidity and mortality in both humans and livestock. The lack of efficient countermeasure strategies, the potential for dispersion into new regions, and the pathogenesis in humans and livestock make RVFV a serious public health concern. The receptors, cellular factors, and entry pathways used by RVFV and other members of the family Bunyaviridae remain largely uncharacterized. Here we provide evidence that RVFV strain MP-12 uses dynamin-dependent caveola-mediated endocytosis for cell entry. Caveolae are lipid raft domains composed of caveolin (the main structural component), cholesterol, and sphingolipids. Caveola-mediated endocytosis is responsible for the uptake of a wide variety of host ligands, as well as bacteria, bacterial toxins, and a number of viruses. To determine the cellular entry mechanism of RVFV, we used small-molecule inhibitors, RNA interference (RNAi), and dominant negative (DN) protein expression to inhibit the major mammalian cell endocytic pathways. Inhibitors and RNAi specific for macropinocytosis and clathrin-mediated endocytosis had no effect on RVFV infection. In contrast, inhibitors of caveola-mediated endocytosis, and RNAi targeted to caveolin-1 and dynamin, drastically reduced RVFV infection in multiple cell lines. Expression of DN caveolin-1 also reduced RVFV infection significantly, while expression of DN EPS15, a protein required for the assembly of clathrin-coated pits, and DN PAK-1, an obligate mediator of macropinocytosis, had no significant impact on RVFV infection. These results together suggest that the primary mechanism of RVFV MP-12 uptake is dynamin-dependent, caveolin-1-mediated endocytosis.


Asunto(s)
Caveolas/metabolismo , Endocitosis/fisiología , Virus de la Fiebre del Valle del Rift/fisiología , Internalización del Virus , Animales , Western Blotting , Caveolas/fisiología , Caveolinas/genética , Chlorocebus aethiops , Citometría de Flujo , Proteínas Fluorescentes Verdes , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Interferencia de ARN , ARN Interferente Pequeño/genética
12.
J Virol ; 86(12): 6632-42, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22496210

RESUMEN

Paramyxoviruses initiate entry through the concerted action of the tetrameric attachment glycoprotein (HN, H, or G) and the trimeric fusion glycoprotein (F). The ectodomains of HN/H/G contain a stalk region important for oligomeric stability and for the F triggering resulting in membrane fusion. Paramyxovirus HN, H, and G form a dimer-of-dimers consisting of disulfide-linked dimers through their stalk domain cysteines. The G attachment protein stalk domain of the highly pathogenic Nipah virus (NiV) contains a distinct but uncharacterized cluster of three cysteine residues (C146, C158, C162). On the basis of a panoply of assays, we report that C158 and C162 of NiV-G likely mediate covalent subunit dimerization, while C146 mediates the stability of higher-order oligomers. For HN or H, mutation of stalk cysteines attenuates but does not abrogate the ability to trigger fusion. In contrast, the NiV-G stalk cysteine mutants were completely deficient in triggering fusion, even though they could still bind the ephrinB2 receptor and associate with F. Interestingly, all cysteine stalk mutants exhibited constitutive exposure of the Mab45 receptor binding-enhanced epitope, previously implicated in F triggering. The enhanced binding of Mab45 to the cysteine mutants relative to wild-type NiV-G, without the addition of the receptor, implicates the stalk cysteines in the stabilization of a pre-receptor-bound conformation and the regulation of F triggering. Sequence alignments revealed that the stalk cysteines were adjacent to a proline-rich microdomain unique to the Henipavirus genus. Our data propose that the cysteine cluster in the NiV-G stalk functions to maintain oligomeric stability but is more importantly involved in stabilizing a unique microdomain critical for triggering fusion.


Asunto(s)
Cisteína/metabolismo , Infecciones por Henipavirus/virología , Virus Nipah/fisiología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Línea Celular , Cisteína/química , Cisteína/genética , Dimerización , Humanos , Datos de Secuencia Molecular , Virus Nipah/química , Virus Nipah/genética , Estructura Terciaria de Proteína , Alineación de Secuencia , Proteínas del Envoltorio Viral/genética
13.
PLoS Pathog ; 6(6): e1000956, 2010 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-20585556

RESUMEN

Entry of human immunodeficiency virus type 1 (HIV-1) commences with binding of the envelope glycoprotein (Env) to the receptor CD4, and one of two coreceptors, CXCR4 or CCR5. Env-mediated signaling through coreceptor results in Galphaq-mediated Rac activation and actin cytoskeleton rearrangements necessary for fusion. Guanine nucleotide exchange factors (GEFs) activate Rac and regulate its downstream protein effectors. In this study we show that Env-induced Rac activation is mediated by the Rac GEF Tiam-1, which associates with the adaptor protein IRSp53 to link Rac to the Wave2 complex. Rac and the tyrosine kinase Abl then activate the Wave2 complex and promote Arp2/3-dependent actin polymerization. Env-mediated cell-cell fusion, virus-cell fusion and HIV-1 infection are dependent on Tiam-1, Abl, IRSp53, Wave2, and Arp3 as shown by attenuation of fusion and infection in cells expressing siRNA targeted to these signaling components. HIV-1 Env-dependent cell-cell fusion, virus-cell fusion and infection were also inhibited by Abl kinase inhibitors, imatinib, nilotinib, and dasatinib. Treatment of cells with Abl kinase inhibitors did not affect cell viability or surface expression of CD4 and CCR5. Similar results with inhibitors and siRNAs were obtained when Env-dependent cell-cell fusion, virus-cell fusion or infection was measured, and when cell lines or primary cells were the target. Using membrane curving agents and fluorescence microscopy, we showed that inhibition of Abl kinase activity arrests fusion at the hemifusion (lipid mixing) step, suggesting a role for Abl-mediated actin remodeling in pore formation and expansion. These results suggest a potential utility of Abl kinase inhibitors to treat HIV-1 infected patients.


Asunto(s)
Fusión Celular , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/fisiología , Proteínas Proto-Oncogénicas c-abl/metabolismo , Virión/metabolismo , Internalización del Virus , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Actinas/metabolismo , Astrocitoma/genética , Astrocitoma/metabolismo , Astrocitoma/patología , Western Blotting , Antígenos CD4/metabolismo , Células Cultivadas , Citoesqueleto , Citometría de Flujo , Genes env , Humanos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-abl/genética , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Linfocitos T , Replicación Viral , Familia de Proteínas del Síndrome de Wiskott-Aldrich/genética
14.
J Virol ; 82(18): 9191-205, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18632858

RESUMEN

Binding of human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein (Env) with the primary receptor CD4 and one of two coreceptors, CXCR4 or CCR5, activates a signaling cascade resulting in Rac-1 GTPase activation and stimulation of actin cytoskeletal reorganizations critical for HIV-1-mediated membrane fusion. The mechanism by which HIV-1 Env induces Rac-1 activation and subsequent actin cytoskeleton rearrangement is unknown. In this study, we show that Env-mediated Rac-1 activation is dependent on the activation of Galpha(q) and its downstream targets. Fusion and Rac-1 activation are mediated by Galpha(q) and phospholipase C (PLC), as shown by attenuation of fusion and Rac-1 activation in cells either expressing small interfering RNA (siRNA) targeting Galpha(q) or treated with the PLC inhibitor U73122. Rac-1 activation and fusion were also blocked by multiple protein kinase C inhibitors, by inhibitors of intracellular Ca2+ release, by Pyk2-targeted siRNA, and by the Ras inhibitor S-trans,trans-farnesylthiosalicylic acid (FTS). Fusion was blocked without altering cell viability or cell surface localization of CD4 and CCR5. Similar results were obtained when cell fusion was induced by Env expressed on viral and cellular membranes and when cell lines or primary cells were the target. Treatment with inhibitors and siRNA specific for Galpha(i) or Galpha(s) signaling mediators had no effect on Env-mediated Rac-1 activation or cell fusion, indicating that the Galpha(q) pathway alone is responsible. These results could provide a new focus for therapeutic intervention with drugs targeting host signaling mediators rather than viral molecules, a strategy which is less likely to result in resistance.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , VIH-1/patogenicidad , Fusión de Membrana/efectos de los fármacos , Transducción de Señal , Internalización del Virus , Productos del Gen env del Virus de la Inmunodeficiencia Humana/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Línea Celular , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , VIH-1/genética , VIH-1/metabolismo , Células HeLa , Humanos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores CCR5/metabolismo , Fosfolipasas de Tipo C/metabolismo , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética , Proteína de Unión al GTP rac1/genética
15.
Virology ; 368(1): 1-6, 2007 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-17640696

RESUMEN

A virus-dependent fusion assay was utilized to examine the activity of a panel of HIV-1, -2, and SIV isolates of distinct coreceptor phenotypes. This assay allowed identification of entry inhibitors, and characterization of an antagonist of a Rac guanine nucleotide exchange factor, as an inhibitor of HIV-mediated fusion.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Inhibidores de Fusión de VIH/farmacología , VIH-1/efectos de los fármacos , VIH-2/efectos de los fármacos , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Línea Celular , Humanos , Internalización del Virus/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...