Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Emerg Infect Dis ; 24(1)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29261093

RESUMO

Ebola virus (EBOV) in body fluids poses risk for virus transmission. However, there are limited experimental data for such matrices on the disinfectant efficacy against EBOV. We evaluated the effectiveness of disinfectants against EBOV in blood on surfaces. Only 5% peracetic acid consistently reduced EBOV titers in dried blood to the assay limit of quantification.


Assuntos
Desinfetantes/farmacologia , Ebolavirus/efeitos dos fármacos , Clareadores/farmacologia , Células Cultivadas/virologia , Teste em Amostras de Sangue Seco , Humanos , Laboratórios , Ácido Peracético/farmacologia
2.
PLoS One ; 11(2): e0148476, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26849135

RESUMO

In support of the response to the 2013-2016 Ebola virus disease (EVD) outbreak in Western Africa, we investigated the persistence of Ebola virus/H.sapiens-tc/GIN/2014/Makona-C05 (EBOV/Mak-C05) on non-porous surfaces that are representative of hospitals, airplanes, and personal protective equipment. We performed persistence studies in three clinically-relevant human fluid matrices (blood, simulated vomit, and feces), and at environments representative of in-flight airline passenger cabins, environmentally-controlled hospital rooms, and open-air Ebola treatment centers in Western Africa. We also compared the surface stability of EBOV/Mak-C05 to that of the prototype Ebola virus/H.sapiens-tc/COD/1976/Yambuku-Mayinga (EBOV/Yam-May), in a subset of these conditions. We show that on inert, non-porous surfaces, EBOV decay rates are matrix- and environment-dependent. Among the clinically-relevant matrices tested, EBOV persisted longest in dried human blood, had limited viability in dried simulated vomit, and did not persist in feces. EBOV/Mak-C05 and EBOV/Yam-May decay rates in dried matrices were not significantly different. However, during the drying process in human blood, EBOV/Yam-May showed significantly greater loss in viability than EBOV/Mak-C05 under environmental conditions relevant to the outbreak region, and to a lesser extent in conditions relevant to an environmentally-controlled hospital room. This factor may contribute to increased communicability of EBOV/Mak-C05 when surfaces contaminated with dried human blood are the vector and may partially explain the magnitude of the most recent outbreak, compared to prior outbreaks. These EBOV persistence data will improve public health efforts by informing risk assessments, structure remediation decisions, and response procedures for future EVD outbreaks.


Assuntos
Ebolavirus/fisiologia , Equipamento de Proteção Individual/virologia , Animais , Sangue/virologia , Chlorocebus aethiops , Ebolavirus/patogenicidade , Fezes/virologia , Humanos , Umidade , Especificidade da Espécie , Células Vero/virologia , Vômito/virologia
3.
Antiviral Res ; 121: 31-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26116756

RESUMO

Human respiratory syncytial virus (hRSV) is a highly contagious Paramyxovirus that infects most children by age two, generating an estimated 75,000-125,000 hospitalizations in the U.S. annually. hRSV is the most common cause of bronchiolitis and pneumonia among infants and children under 1year of age, with significant mortality among high-risk groups. A regulatory agency-approved vaccine is not available, and existing prophylaxis and therapies are limited to use in high-risk pediatric patients; thus additional therapies are sorely needed. Here, we identify a series of benzimidazole analogs that inhibit hRSV infection in vitro with high potency, using a previously-reported high-throughput screening assay. The lead compound, SRI 29365 (1-[6-(2-furyl)[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-3-yl]methyl-1H-benzimidazole), has an EC50 of 66µM and a selectivity >50. We identified additional compounds with varying potencies by testing commercially-available chemical analogs. Time-of-addition experiments indicated that SRI 29365 effectively inhibits viral replication only if present during the early stages of viral infection. We isolated a virus with resistance to SRI 29365 and identified mutations in the transmembrane domain of the viral G protein genomic sequence that suggested that the compound inhibits G-protein mediated attachment of hRSV to cells. Additional experiments with multiple cell types indicated that SRI 29365 antiviral activity correlates with the binding of cell surface heparin by full-length G protein. Lastly, SRI 29365 did not reduce hRSV titers or morbidity/mortality in efficacy studies using a cotton rat model. Although SRI 29365 and analogs inhibit hRSV replication in vitro, this work suggests that the G-protein may not be a valid drug target in vivo.


Assuntos
Antivirais/farmacologia , Benzimidazóis/farmacologia , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Vírus Sincicial Respiratório Humano/fisiologia , Proteínas do Envelope Viral/antagonistas & inibidores , Ligação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Modelos Animais de Doenças , Farmacorresistência Viral , Ensaios de Triagem em Larga Escala , Humanos , Testes de Sensibilidade Microbiana , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/virologia , Sigmodontinae , Análise de Sobrevida , Proteínas do Envelope Viral/genética
4.
Cell ; 161(6): 1252-65, 2015 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-26046436

RESUMO

Small-molecule probes can illuminate biological processes and aid in the assessment of emerging therapeutic targets by perturbing biological systems in a manner distinct from other experimental approaches. Despite the tremendous promise of chemical tools for investigating biology and disease, small-molecule probes were unavailable for most targets and pathways as recently as a decade ago. In 2005, the NIH launched the decade-long Molecular Libraries Program with the intent of innovating in and broadening access to small-molecule science. This Perspective describes how novel small-molecule probes identified through the program are enabling the exploration of biological pathways and therapeutic hypotheses not otherwise testable. These experiences illustrate how small-molecule probes can help bridge the chasm between biological research and the development of medicines but also highlight the need to innovate the science of therapeutic discovery.


Assuntos
Descoberta de Drogas , Bibliotecas de Moléculas Pequenas , Animais , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Ensaios de Triagem em Larga Escala , Humanos , National Institutes of Health (U.S.) , Estados Unidos
5.
FASEB J ; 29(7): 2712-25, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25795456

RESUMO

We sought to determine the mechanisms by which influenza infection of human epithelial cells decreases cystic fibrosis transmembrane conductance regulator (CFTR) expression and function. We infected human bronchial epithelial (NHBE) cells and murine nasal epithelial (MNE) cells with various strains of influenza A virus. Influenza infection significantly reduced CFTR short circuit currents (Isc) and protein levels at 8 hours postinfection. We then infected CFTR expressing human embryonic kidney (HEK)-293 cells (HEK-293 CFTRwt) with influenza virus encoding a green fluorescent protein (GFP) tag and performed whole-cell and cell-attached patch clamp recordings. Forskolin-stimulated, GlyH-101-sensitive CFTR conductances, and CFTR open probabilities were reduced by 80% in GFP-positive cells; Western blots also showed significant reduction in total and plasma membrane CFTR levels. Knockdown of the influenza matrix protein 2 (M2) with siRNA, or inhibition of its activity by amantadine, prevented the decrease in CFTR expression and function. Lysosome inhibition (bafilomycin-A1), but not proteasome inhibition (lactacystin), prevented the reduction in CFTR levels. Western blots of immunoprecipitated CFTR from influenza-infected cells, treated with BafA1, and probed with antibodies against lysine 63-linked (K-63) or lysine 48-linked (K-48) polyubiquitin chains supported lysosomal targeting. These results highlight CFTR damage, leading to early degradation as an important contributing factor to influenza infection-associated ion transport defects.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Vírus da Influenza A/fisiologia , Vírus da Influenza A/patogenicidade , Proteínas da Matriz Viral/fisiologia , Animais , Apoptose , Células Cultivadas , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Epiteliais/virologia , Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Vírus da Influenza A/genética , Influenza Humana/metabolismo , Influenza Humana/patologia , Influenza Humana/virologia , Transporte de Íons , Lisossomos/metabolismo , Camundongos , Necrose , Técnicas de Patch-Clamp , Proteólise , Transfecção , Proteínas da Matriz Viral/antagonistas & inibidores , Proteínas da Matriz Viral/genética
6.
Assay Drug Dev Technol ; 13(1): 44-54, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25710545

RESUMO

High-throughput screening (HTS) has been integrated into the drug discovery process, and multiple assay formats have been widely used in many different disease areas but with limited focus on infectious agents. In recent years, there has been an increase in the number of HTS campaigns using infectious wild-type pathogens rather than surrogates or biochemical pathogen-derived targets. Concurrently, enhanced emerging pathogen surveillance and increased human mobility have resulted in an increase in the emergence and dissemination of infectious human pathogens with serious public health, economic, and social implications at global levels. Adapting the HTS drug discovery process to biocontainment laboratories to develop new drugs for these previously uncharacterized and highly pathogenic agents is now feasible, but HTS at higher biosafety levels (BSL) presents a number of unique challenges. HTS has been conducted with multiple bacterial and viral pathogens at both BSL-2 and BSL-3, and pilot screens have recently been extended to BSL-4 environments for both Nipah and Ebola viruses. These recent successful efforts demonstrate that HTS can be safely conducted at the highest levels of biological containment. This review outlines the specific issues that must be considered in the execution of an HTS drug discovery program for high-containment pathogens. We present an overview of the requirements for HTS in high-level biocontainment laboratories.


Assuntos
Bioensaio/instrumentação , Contenção de Riscos Biológicos/instrumentação , Avaliação Pré-Clínica de Medicamentos/instrumentação , Ensaios de Triagem em Larga Escala/instrumentação , Laboratórios , Tecnologia Farmacêutica/instrumentação , Desenho de Fármacos , Desenho de Equipamento , Análise de Falha de Equipamento , Robótica/instrumentação , Manejo de Espécimes/instrumentação
7.
J Med Chem ; 57(24): 10314-28, 2014 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-25399509

RESUMO

A quinazolinedione-derived screening hit 2 was discovered with cellular antiviral activity against respiratory syncytial virus (CPE EC50 = 2.1 µM), moderate efficacy in reducing viral progeny (4.2 log at 10 µM), and marginal cytotoxic liability (selectivity index, SI ∼ 24). Scaffold optimization delivered analogs with improved potency and selectivity profiles. Most notable were compounds 15 and 19 (EC50 = 300-500 nM, CC50 > 50 µM, SI > 100), which significantly reduced viral titer (>400,000-fold), and several analogs were shown to block the activity of the RNA-dependent RNA-polymerase complex of RSV.


Assuntos
Antivirais/farmacologia , Benzamidas/farmacologia , Proliferação de Células/efeitos dos fármacos , Quinazolinonas/farmacologia , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Antivirais/síntese química , Benzamidas/síntese química , Desenho de Fármacos , Células Hep G2 , Humanos , Concentração Inibidora 50 , Modelos Moleculares , Estrutura Molecular , Quinazolinonas/síntese química , Quinazolinonas/química , RNA Polimerase Dependente de RNA/metabolismo , Infecções por Vírus Respiratório Sincicial/virologia , Relação Estrutura-Atividade
8.
J Med Chem ; 57(20): 8608-21, 2014 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-25244572

RESUMO

Venezuelan equine encephalitis virus (VEEV) is an emerging pathogenic alphavirus that can cause significant disease in humans. Given the absence of therapeutic options available and the significance of VEEV as a weaponized agent, an optimization effort was initiated around a quinazolinone screening hit 1 with promising cellular antiviral activity (EC50 = 0.8 µM), limited cytotoxic liability (CC50 > 50 µM), and modest in vitro efficacy in reducing viral progeny (63-fold at 5 µM). Scaffold optimization revealed a novel rearrangement affording amidines, specifically compound 45, which was found to potently inhibit several VEEV strains in the low nanomolar range without cytotoxicity (EC50 = 0.02-0.04 µM, CC50 > 50 µM) while limiting in vitro viral replication (EC90 = 0.17 µM). Brain exposure was observed in mice with 45. Significant protection was observed in VEEV-infected mice at 5 mg kg(-1) day(-1) and viral replication appeared to be inhibited through interference of viral nonstructural proteins.


Assuntos
Antivirais/química , Antivirais/farmacologia , Benzamidas/farmacologia , Vírus da Encefalite Equina Venezuelana/efeitos dos fármacos , Piperazinas/farmacologia , Animais , Benzamidas/química , Avaliação Pré-Clínica de Medicamentos/métodos , Encefalomielite Equina Venezuelana/tratamento farmacológico , Compostos Heterocíclicos com 2 Anéis/química , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Piperazinas/química , Quinazolinonas/química , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade , Replicação Viral/efeitos dos fármacos
9.
PLoS One ; 9(7): e96054, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24983234

RESUMO

Uropathogenic Escherichia coli (UPEC) is the leading cause of community-acquired urinary tract infections (UTIs), with over 100 million UTIs occurring annually throughout the world. Increasing antimicrobial resistance among UPEC limits ambulatory care options, delays effective treatment, and may increase overall morbidity and mortality from complications such as urosepsis. The polysaccharide capsules of UPEC are an attractive target a therapeutic, based on their importance in defense against the host immune responses; however, the large number of antigenic types has limited their incorporation into vaccine development. The objective of this study was to identify small-molecule inhibitors of UPEC capsule biogenesis. A large-scale screening effort entailing 338,740 compounds was conducted in a cell-based, phenotypic screen for inhibition of capsule biogenesis in UPEC. The primary and concentration-response assays yielded 29 putative inhibitors of capsule biogenesis, of which 6 were selected for further studies. Secondary confirmatory assays identified two highly active agents, named DU003 and DU011, with 50% inhibitory concentrations of 1.0 µM and 0.69 µM, respectively. Confirmatory assays for capsular antigen and biochemical measurement of capsular sugars verified the inhibitory action of both compounds and demonstrated minimal toxicity and off-target effects. Serum sensitivity assays demonstrated that both compounds produced significant bacterial death upon exposure to active human serum. DU011 administration in mice provided near complete protection against a lethal systemic infection with the prototypic UPEC K1 isolate UTI89. This work has provided a conceptually new class of molecules to combat UPEC infection, and future studies will establish the molecular basis for their action along with efficacy in UTI and other UPEC infections.


Assuntos
Antibacterianos , Cápsulas Bacterianas/metabolismo , Infecções por Escherichia coli/tratamento farmacológico , Infecções Urinárias/tratamento farmacológico , Escherichia coli Uropatogênica/metabolismo , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Infecções por Escherichia coli/patologia , Feminino , Humanos , Camundongos , Infecções Urinárias/patologia
10.
PLoS Pathog ; 10(6): e1004213, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24967809

RESUMO

Alphaviruses present serious health threats as emerging and re-emerging viruses. Venezuelan equine encephalitis virus (VEEV), a New World alphavirus, can cause encephalitis in humans and horses, but there are no therapeutics for treatment. To date, compounds reported as anti-VEEV or anti-alphavirus inhibitors have shown moderate activity. To discover new classes of anti-VEEV inhibitors with novel viral targets, we used a high-throughput screen based on the measurement of cell protection from live VEEV TC-83-induced cytopathic effect to screen a 340,000 compound library. Of those, we identified five novel anti-VEEV compounds and chose a quinazolinone compound, CID15997213 (IC50 = 0.84 µM), for further characterization. The antiviral effect of CID15997213 was alphavirus-specific, inhibiting VEEV and Western equine encephalitis virus, but not Eastern equine encephalitis virus. In vitro assays confirmed inhibition of viral RNA, protein, and progeny synthesis. No antiviral activity was detected against a select group of RNA viruses. We found mutations conferring the resistance to the compound in the N-terminal domain of nsP2 and confirmed the target residues using a reverse genetic approach. Time of addition studies showed that the compound inhibits the middle stage of replication when viral genome replication is most active. In mice, the compound showed complete protection from lethal VEEV disease at 50 mg/kg/day. Collectively, these results reveal a potent anti-VEEV compound that uniquely targets the viral nsP2 N-terminal domain. While the function of nsP2 has yet to be characterized, our studies suggest that the protein might play a critical role in viral replication, and further, may represent an innovative opportunity to develop therapeutic interventions for alphavirus infection.


Assuntos
Antivirais/farmacologia , Vírus da Encefalite Equina Venezuelana/efeitos dos fármacos , Encefalomielite Equina Venezuelana/tratamento farmacológico , Quinazolinonas/farmacologia , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Modelos Animais de Doenças , Farmacorresistência Viral/genética , Vírus da Encefalite Equina Venezuelana/genética , Encefalomielite Equina Venezuelana/virologia , Ensaios de Triagem em Larga Escala , Camundongos , Camundongos Endogâmicos C3H , Especificidade da Espécie , Relação Estrutura-Atividade , Células Vero , Ensaio de Placa Viral , Replicação Viral/efeitos dos fármacos
11.
Assay Drug Dev Technol ; 12(3): 155-61, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24735442

RESUMO

Nipah virus is a biosafety level 4 (BSL-4) pathogen that causes severe respiratory illness and encephalitis in humans. To identify novel small molecules that target Nipah virus replication as potential therapeutics, Southern Research Institute and Galveston National Laboratory jointly developed an automated high-throughput screening platform that is capable of testing 10,000 compounds per day within BSL-4 biocontainment. Using this platform, we screened a 10,080-compound library using a cell-based, high-throughput screen for compounds that inhibited the virus-induced cytopathic effect. From this pilot effort, 23 compounds were identified with EC50 values ranging from 3.9 to 20.0 µM and selectivities >10. Three sulfonamide compounds with EC50 values <12 µM were further characterized for their point of intervention in the viral replication cycle and for broad antiviral efficacy. Development of HTS capability under BSL-4 containment changes the paradigm for drug discovery for highly pathogenic agents because this platform can be readily modified to identify prophylactic and postexposure therapeutic candidates against other BSL-4 pathogens, particularly Ebola, Marburg, and Lassa viruses.


Assuntos
Antivirais/administração & dosagem , Antivirais/química , Avaliação Pré-Clínica de Medicamentos/instrumentação , Ensaios de Triagem em Larga Escala/instrumentação , Vírus Nipah/efeitos dos fármacos , Vírus Nipah/fisiologia , Sulfonamidas/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Animais , Chlorocebus aethiops , Contenção de Riscos Biológicos/instrumentação , Relação Dose-Resposta a Droga , Desenho de Equipamento , Análise de Falha de Equipamento , Robótica/instrumentação , Células Vero , Replicação Viral/fisiologia
12.
J Biomol Screen ; 19(6): 936-46, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24464431

RESUMO

During viral infection of human cells, host kinases mediate signaling activities that are used by all viruses for replication; therefore, targeting of host kinases is of broad therapeutic interest. Here, host kinases were globally screened during human influenza virus (H1N1) infection to determine the time-dependent effects of virus infection and replication on kinase function. Desthiobiotin-labeled analogs of adenosine triphosphate and adenosine diphosphate were used to probe and covalently label host kinases in infected cell lysates, and probe affinity was determined. Using infected human A549 cells, we screened for time-dependent signal changes and identified host kinases whose probe affinities differed significantly when compared to uninfected cells. Our screen identified 10 novel host kinases that have not been previously shown to be involved with influenza virus replication, and we validated the functional importance of these novel kinases during infection using targeted small interfering RNAs (siRNAs). The effects of kinase-targeted siRNA knockdowns on replicating virus levels were measured by quantitative reverse-transcription PCR and cytoprotection assays. We identified several novel host kinases that, when knocked down, enhanced or reduced the viral load in cell culture. This preliminary work represents the first screen of the changing host kinome in influenza virus-infected human cells.


Assuntos
Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/enzimologia , Proteínas Serina-Treonina Quinases/química , RNA Interferente Pequeno/genética , Replicação Viral , Células A549 , Difosfato de Adenosina/química , Trifosfato de Adenosina/química , Apoptose , Biotina/análogos & derivados , Biotina/química , Sobrevivência Celular , Cromatografia Líquida , Descoberta de Drogas , Humanos , Espectrometria de Massas , Quinase 1 Relacionada a NIMA/química , Peptídeos/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina-Treonina Quinase 3 , Carga Viral
13.
Am J Physiol Lung Cell Mol Physiol ; 305(2): L108-17, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23709619

RESUMO

Death by respiratory complications from influenza infections continues to be a major global health concern. Antiviral drugs are widely available for therapy and prophylaxis, but viral mutations have resulted in resistance that threatens to reduce the long-term utility of approved antivirals. Vaccination is the best method for controlling influenza, but vaccine strategies are blunted by virus antigenic drift and shift. Genetic shift in particular has led to four pandemics in the last century, which have prompted the development of efficient global surveillance and vaccination programs. Although the influenza pandemic of 2009 emphasized the need for the rapid standardization of global surveillance methods and the preparation and dissemination of global assay standards for improved reporting and diagnostic tools, outbreaks of novel influenza strains continue to occur, and current efforts must be enhanced by aggressive public education programs to promote increased vaccination rates in the global population. Recently, a novel H7N9 avian influenza virus with potential to become a pandemic strain emerged in China and was transmitted from animals to humans with a demonstrated >20% mortality rate. Sporadic outbreaks of highly lethal avian virus strains have already increased public awareness and altered annual vaccine production strategies to prevent the natural adaption of this virus to human-to-human transmission. Additional strategies for combating influenza include advancement of new antivirals for unexploited viral or host cellular targets; novel adjuvants and alternate vaccine delivery systems; and development of universal protein, DNA, or multivalent vaccines designed to increase immune responsiveness and enhance public health response times.


Assuntos
Antivirais/uso terapêutico , Controle de Doenças Transmissíveis/métodos , Deriva Genética , Vacinas contra Influenza , Influenza Humana , Pandemias , Controle de Doenças Transmissíveis/organização & administração , Humanos , Vacinas contra Influenza/genética , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/uso terapêutico , Influenza Humana/genética , Influenza Humana/imunologia , Influenza Humana/mortalidade , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Educação de Pacientes como Assunto/métodos
14.
Am J Physiol Lung Cell Mol Physiol ; 304(9): L582-92, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23457187

RESUMO

The human cystic fibrosis transmembrane conductance regulator (CFTR) is a cyclic AMP-activated chloride (Cl(-)) channel in the lung epithelium that helps regulate the thickness and composition of the lung epithelial lining fluid. We investigated whether influenza M2 protein, a pH-activated proton (H(+)) channel that traffics to the plasma membrane of infected cells, altered CFTR expression and function. M2 decreased CFTR activity in 1) Xenopus oocytes injected with human CFTR, 2) epithelial cells (HEK-293) stably transfected with CFTR, and 3) human bronchial epithelial cells (16HBE14o-) expressing native CFTR. This inhibition was partially reversed by an inhibitor of the ubiquitin-activating enzyme E1. Next we investigated whether the M2 inhibition of CFTR activity was due to an increase of secretory organelle pH by M2. Incubation of Xenopus oocytes expressing CFTR with ammonium chloride or concanamycin A, two agents that alkalinize the secretory pathway, inhibited CFTR activity in a dose-dependent manner. Treatment of M2- and CFTR-expressing oocytes with the M2 ion channel inhibitor amantadine prevented the loss in CFTR expression and activity; in addition, M2 mutants, lacking the ability to transport H(+), did not alter CFTR activity in Xenopus oocytes and HEK cells. Expression of an M2 mutant retained in the endoplasmic reticulum also failed to alter CFTR activity. In summary, our data show that M2 decreases CFTR activity by increasing secretory organelle pH, which targets CFTR for destruction by the ubiquitin system. Alteration of CFTR activity has important consequences for fluid regulation and may potentially modify the immune response to viral infection.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Canais Iônicos/fisiologia , Proteínas da Matriz Viral/farmacologia , Amantadina/farmacologia , Animais , Benzoatos/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/biossíntese , Furanos/farmacologia , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Canais Iônicos/efeitos dos fármacos , Oócitos/metabolismo , Técnicas de Patch-Clamp , Pirazóis/farmacologia , Via Secretória/efeitos dos fármacos , Enzimas Ativadoras de Ubiquitina/antagonistas & inibidores , Xenopus
15.
J Med Chem ; 55(20): 8582-7, 2012 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-23043370

RESUMO

A high-throughput, cell-based screen was used to identify chemotypes as inhibitors for human respiratory syncytial virus (hRSV). Optimization of a sulfonylpyrrolidine scaffold resulted in compound 5o that inhibited a virus-induced cytopathic effect in the entry stage of infection (EC50 = 2.3 ± 0.8 µM) with marginal cytotoxicity (CC50 = 30.9 ± 1.1 µM) and reduced viral titer by 100-fold. Compared to ribavirin, sulfonylpyrrolidine 5o demonstrated an improved in vitro potency and selectivity index.


Assuntos
Antivirais/síntese química , Pirrolidinas/síntese química , Quinolinas/síntese química , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Sulfonamidas/síntese química , Sulfonas/síntese química , Antivirais/química , Antivirais/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Efeito Citopatogênico Viral/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Humanos , Pirrolidinas/química , Pirrolidinas/farmacologia , Quinolinas/química , Quinolinas/farmacologia , Vírus Sinciciais Respiratórios/fisiologia , Ribavirina/farmacologia , Estereoisomerismo , Relação Estrutura-Atividade , Sulfonamidas/química , Sulfonamidas/farmacologia , Sulfonas/química , Sulfonas/farmacologia , Carga Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
16.
Expert Opin Drug Discov ; 7(5): 429-38, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22435452

RESUMO

INTRODUCTION: Influenza antiviral high-throughput screens have been extensive, and yet no approved influenza antivirals have been identified through high-throughput screening. This underscores the idea that development of successful screens should focus on the exploitation of the underrepresented viral targets and novel, therapeutic host targets. AREAS COVERED: The authors review conventional screening applications and emerging technologies with the potential to enhance influenza antiviral discovery. Real-world examples from the authors' work in biocontained environments are also provided. Future innovations are discussed, including the use of targeted libraries, multiplexed assays, proximity-based endpoint methods, non-laboratory-adapted virus strains, and primary cells, for immediate physiological relevance and translational applications. EXPERT OPINION: The lack of successful anti-influenza drug discovery using high-throughput screening should not deter future efforts. Increased understanding of the functions of viral targets and host-pathogen interactions has broadened the target reservoir. Future screening efforts should focus on identifying new drugs against unexploited viral and host targets using currently developed assays, and on the development of novel, innovative assays to discover new drugs with novel mechanisms. Innovative screens must be designed to identify compounds that specifically inhibit protein-protein or protein-RNA interactions or other virus/host factor interactions that are crucial for viral replication. Finally, the use of recent viral isolates, increased biocontainment (for highly-pathogenic strains), primary cell lines, and targeted compound libraries must converge in efficient high-throughput primary screens to generate high-content, physiologically-relevant data on compounds with robust antiviral activity.


Assuntos
Antivirais/química , Descoberta de Drogas/métodos , Ensaios de Triagem em Larga Escala/métodos , Influenza Humana/tratamento farmacológico , Animais , Antivirais/farmacologia , Cães , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Ensaios de Triagem em Larga Escala/tendências , Humanos , Vírus da Influenza A/efeitos dos fármacos , Vacinas contra Influenza , Proteínas Virais/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos
17.
Methods Mol Biol ; 857: 399-414, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22323232

RESUMO

The number of known three-dimensional protein sequences is orders of magnitude higher than the number of known protein structures. This is a result of an increase in large-scale genomic sequencing projects, the inability of proteins to crystallize or crystals to diffract well, or a simple lack of resources. An alternative is to use one of a variety of available homology modeling programs to produce a computational model of a protein. Protein models are produced using information from known protein structures found to be similar. Here, we compare the ability of a number of popular homology modeling programs to produce quality models from user-defined target-template sequence alignments over a range of circumstances including low sequence identity, variable sequence length, and when interfaced with a protein or small molecule. Programs evaluated include Prime, SWISS-MODEL, MOE, MODELLER, ROSETTA, Composer, ORCHESTRAR, and I-TASSER. Proteins to be modeled were chosen to test a range of sequence identities, sequence lengths, and protein motifs and all are of scientific importance. These include HIV-1 protease, kinases, dihydrofolate reductase, a viral capsid protein, and factor Xa among others. For the most part, the programs produce results that are similar. For example, all programs are able to produce reasonable models when sequence identities are >30% and all programs have difficulties producing complete models when sequence identities are lower. However, certain programs fare slightly better than others in certain situations and we attempt to provide insight on this topic.


Assuntos
Algoritmos , Proteínas/química , Homologia Estrutural de Proteína , Sítios de Ligação , Simulação por Computador , Bases de Dados de Proteínas , Modelos Moleculares , Alinhamento de Sequência/métodos , Software
18.
Life Sci ; 90(11-12): 440-5, 2012 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-22269828

RESUMO

AIM: To compare the efficacy of ribavirin and oseltamivir in reducing mortality, lung injury and cytokine response profile in pandemic influenza H1N1 (2009) infection. MAIN METHODS: We assessed survival, weight loss, lung viral load (by RT-PCR), lung injury (by protein content in bronchoalveolar lavage), and inflammation (cell counts, differentials and cytokines in the bronchoalveolar lavage) in BALB/c mice after infection with mouse-adapted pandemic influenza strain A/California/04/2009. KEY FINDINGS: Our results indicate that ribavirin (80 mg kg(-1)) and oseltamivir (50 mg kg(-1)) are equally effective in improving survival (100% vs. 0% in water treated controls), while ribavirin proved to be more effective in significantly preventing weight loss. Both drugs diminished the injury of the alveolar-capillary barrier by decreasing the protein detected in the BAL to baseline levels, and they were also equally effective in reduction lung viral loads by 100-fold. Administration of either drug did not decrease the amount of inflammatory infiltrate in the lung, but ribavirin significantly reduced the percentage comprised of lymphocytes. This study shows that these antivirals differentially regulate inflammatory cytokines and chemokines with ribavirin significantly reducing most of the cytokines/chemokines measured. Ribavirin treatment leads to a Th1 cytokine response while oseltamivir leads to a Th2 cytokine response with significant increase in the levels of the anti-inflammatory cytokine IL-10. SIGNIFICANCE: This study reveals new mechanistic insights in the way that ribavirin and oseltamivir exert their antiviral activity and supports the theory that ribavirin could potentially serve as an efficacious therapeutic alternative for oseltamivir resistant pandemic H1N1 strains.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Pulmão/virologia , Infecções por Orthomyxoviridae/tratamento farmacológico , Oseltamivir/farmacologia , Ribavirina/farmacologia , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Quimiocinas/análise , Citocinas/análise , Primers do DNA/genética , Feminino , Estimativa de Kaplan-Meier , Pulmão/efeitos dos fármacos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/patologia , Oseltamivir/uso terapêutico , Reação em Cadeia da Polimerase em Tempo Real , Ribavirina/uso terapêutico
19.
J Biomol Screen ; 17(2): 194-203, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21948801

RESUMO

The authors conducted a high-throughput screening campaign for inhibitors of SV40 large T antigen ATPase activity to identify candidate antivirals that target the replication of polyomaviruses. The primary assay was adapted to 1536-well microplates and used to screen the National Institutes of Health Molecular Libraries Probe Centers Network library of 306 015 compounds. The primary screen had an Z value of ~0.68, signal/background = 3, and a high (5%) DMSO tolerance. Two counterscreens and two secondary assays were used to prioritize hits by EC(50), cytotoxicity, target specificity, and off-target effects. Hits that inhibited ATPase activity by >44% in the primary screen were tested in dose-response efficacy and eukaryotic cytotoxicity assays. After evaluation of hit cytotoxicity, drug likeness, promiscuity, and target specificity, three compounds were chosen for chemical optimization. Chemical optimization identified a class of bisphenols as the most effective biochemical inhibitors. Bisphenol A inhibited SV40 large T antigen ATPase activity with an IC(50) of 41 µM in the primary assay and 6.2 µM in a cytoprotection assay. This compound class is suitable as probes for biochemical investigation of large T antigen ATPase activity, but because of their cytotoxicity, further optimization is necessary for their use in studying polyomavirus replication in vivo.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Antígenos Transformantes de Poliomavirus/metabolismo , Ensaios de Triagem em Larga Escala/métodos , Fenóis/farmacologia , Animais , Antivirais/farmacologia , Compostos Benzidrílicos , Linhagem Celular , Chlorocebus aethiops , Cães , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Polyomavirus/enzimologia , Bibliotecas de Moléculas Pequenas/análise
20.
Antimicrob Agents Chemother ; 55(11): 5054-62, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21844323

RESUMO

Poxvirus uracil DNA glycosylase D4 in association with A20 and the catalytic subunit of DNA polymerase forms the processive polymerase complex. The binding of D4 and A20 is essential for processive polymerase activity. Using an AlphaScreen assay, we identified compounds that inhibit protein-protein interactions between D4 and A20. Effective interaction inhibitors exhibited both antiviral activity and binding to D4. These results suggest that novel antiviral agents that target the protein-protein interactions between D4 and A20 can be developed for the treatment of infections with poxviruses, including smallpox.


Assuntos
Antivirais/farmacologia , Vaccinia virus/efeitos dos fármacos , Proteínas Virais/metabolismo , Linhagem Celular , DNA Glicosilases/metabolismo , Humanos , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...