Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 93(4)2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30541831

RESUMO

Respiratory syncytial virus (RSV) is a leading cause of hospitalization of infants and young children, causing considerable respiratory disease and repeat infections that may lead to chronic respiratory conditions such as asthma, wheezing, and bronchitis. RSV causes ∼34 million new episodes of lower respiratory tract illness (LRTI) in children younger than 5 years of age, with >3 million hospitalizations due to severe RSV-associated LRTI. The standard of care is limited to symptomatic relief as there are no approved vaccines and few effective antiviral drugs; thus, a safe and efficacious RSV therapeutic is needed. Therapeutic targeting of host proteins hijacked by RSV to facilitate replication is a promising antiviral strategy as targeting the host reduces the likelihood of developing drug resistance. The nuclear export of the RSV M protein, mediated by the nuclear export protein exportin 1 (XPO1), is crucial for RSV assembly and budding. Inhibition of RSV M protein export by leptomycin B correlated with reduced RSV replication in vitro In this study, we evaluated the anti-RSV efficacy of Verdinexor (KPT-335), a small molecule designed to reversibly inhibit XPO1-mediated nuclear export. KPT-335 inhibited XPO1-mediated transport and reduced RSV replication in vitro KPT-335 was effective against RSV A and B strains and reduced viral replication following prophylactic or therapeutic administration. Inhibition of RSV replication by KPT-335 was due to a combined effect of reduced XPO1 expression, disruption of the nuclear export of RSV M protein, and inactivation of the NF-κB signaling pathway.IMPORTANCE RSV is an important cause of LRTI in infants and young children for which there are no suitable antiviral drugs offered. We evaluated the efficacy of KPT-335 as an anti-RSV drug and show that KPT-335 inhibits XPO1-mediated nuclear export, leading to nuclear accumulation of RSV M protein and reduction in RSV levels. KPT-335 treatment also resulted in inhibition of proinflammatory pathways, which has important implications for its effectiveness in vivo.


Assuntos
Acrilamidas/farmacologia , Hidrazinas/farmacologia , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Células A549 , Acrilamidas/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Antivirais/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Glicoproteínas/imunologia , Humanos , Hidrazinas/metabolismo , Carioferinas/efeitos dos fármacos , Carioferinas/metabolismo , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/metabolismo , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Células Vero , Proteína Exportina 1
2.
Viruses ; 10(1)2018 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-29361733

RESUMO

Two primary causes of respiratory tract infections are respiratory syncytial virus (RSV) and influenza viruses, both of which remain major public health concerns. There are a limited number of antiviral drugs available for the treatment of RSV and influenza, each having limited effectiveness and each driving selective pressure for the emergence of drug-resistant viruses. Novel broad-spectrum antivirals are needed to circumvent problems with current disease intervention strategies, while improving the cytokine-induced immunopathology associated with RSV and influenza infections. In this review, we examine the use of Verdinexor (KPT-335, a novel orally bioavailable drug that functions as a selective inhibitor of nuclear export, SINE), as an antiviral with multifaceted therapeutic potential. KPT-335 works to (1) block CRM1 (i.e., Chromosome Region Maintenance 1; exportin 1 or XPO1) mediated export of viral proteins critical for RSV and influenza pathogenesis; and (2) repress nuclear factor κB (NF-κB) activation, thus reducing cytokine production and eliminating virus-associated immunopathology. The repurposing of SINE compounds as antivirals shows promise not only against RSV and influenza virus but also against other viruses that exploit the nucleus as part of their viral life cycle.


Assuntos
Acrilamidas/uso terapêutico , Antivirais/uso terapêutico , Hidrazinas/uso terapêutico , Carioferinas/antagonistas & inibidores , Orthomyxoviridae/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Acrilamidas/farmacologia , Animais , Antivirais/farmacologia , Apoptose , Linhagem Celular Tumoral , Ensaios Clínicos como Assunto , Descoberta de Drogas , Humanos , Hidrazinas/farmacologia , Fatores Imunológicos/farmacologia , Fatores Imunológicos/uso terapêutico , Influenza Humana/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Proteína Exportina 1
3.
PLoS One ; 11(5): e0155917, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27224013

RESUMO

Respiratory syncytial virus (RSV) and human metapneumovirus (HMPV) are paramyxoviruses that are responsible for substantial human health burden, particularly in children and the elderly. The fusion (F) glycoproteins are major targets of the neutralizing antibody response and studies have mapped dominant antigenic sites in F. Here we grafted a major neutralizing site of RSV F, recognized by the prophylactic monoclonal antibody palivizumab, onto HMPV F, generating a chimeric protein displaying epitopes of both viruses. We demonstrate that the resulting chimeric protein (RPM-1) is recognized by both anti-RSV and anti-HMPV F neutralizing antibodies indicating that it can be used to map the epitope specificity of antibodies raised against both viruses. Mice immunized with the RPM-1 chimeric antigen generate robust neutralizing antibody responses to MPV but weak or no cross-reactive recognition of RSV F, suggesting that grafting of the single palivizumab epitope stimulates a comparatively limited antibody response. The RPM-1 protein provides a new tool for characterizing the immune responses resulting from RSV and HMPV infections and provides insights into the requirements for developing a chimeric subunit vaccine that could induce robust and balanced immunity to both virus infections.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Epitopos/imunologia , Metapneumovirus/imunologia , Proteínas Recombinantes de Fusão/imunologia , Vírus Sinciciais Respiratórios/imunologia , Proteínas Virais de Fusão/imunologia , Animais , Especificidade de Anticorpos , Epitopos/genética , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/genética , Proteínas Virais de Fusão/genética
4.
Vaccine ; 34(7): 950-6, 2016 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-26772634

RESUMO

Human respiratory syncytial virus (hRSV) and human metapneumovirus (hMPV) are major causes of illness among children, the elderly, and the immunocompromised. No vaccine has been licensed for protection against either of these viruses. We tested the ability of two Venezuelan equine encephalitis virus-based viral replicon particle (VEE-VRP) vaccines that express the hRSV or hMPV fusion (F) protein to confer protection against hRSV or hMPV in African green monkeys. Animals immunized with VEE-VRP vaccines developed RSV or MPV F-specific antibodies and serum neutralizing activity. Compared to control animals, immunized animals were better able to control viral load in the respiratory mucosa following challenge and had lower levels of viral genome in nasopharyngeal and bronchoalveolar lavage fluids. The high level of immunogenicity and protective efficacy induced by these vaccine candidates in nonhuman primates suggest that they hold promise for further development.


Assuntos
Infecções por Paramyxoviridae/prevenção & controle , Replicon , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas Virais/imunologia , Alphavirus , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Líquido da Lavagem Broncoalveolar/virologia , Chlorocebus aethiops , Vírus da Encefalite Equina Venezuelana , Imunoglobulina G/sangue , Metapneumovirus , Testes de Neutralização , Nariz/virologia , Vírus Sincicial Respiratório Humano , Proteínas Virais de Fusão/imunologia
5.
J Clin Invest ; 125(6): 2523-31, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25985274

RESUMO

Several HIV envelope-targeting (Env-targeting) antibodies with broad and potent neutralizing activity have been identified and shown to have unusual features. Of these, the PG9 antibody has a long heavy chain complementarity determining region 3 (HCDR3) and possesses unique structural elements that interact with protein and glycan features of the HIV Env glycoprotein. Here, we used the Rosetta software suite to design variants of the PG9 antibody HCDR3 loop with the goal of identifying variants with increased potency and breadth of neutralization for diverse HIV strains. One variant, designated PG9_N100(F)Y, possessed increased potency and was able to neutralize a diverse set of PG9-resistant HIV strains, including those lacking the Env N160 glycan, which is critical for PG9 binding. An atomic resolution structure of the PG9_N100(F)Y fragment antigen binding (Fab) confirmed that the mutated residue retains the paratope surface when compared with WT PG9. Differential scanning calorimetry experiments revealed that the mutation caused a modest increase in thermodynamic stability of the Fab, a feature predicted by the computational model. Our findings suggest that thermodynamic stabilization of the long HCDR3 in its active conformation is responsible for the increased potency of PG9_N100(F)Y, and strategies aimed at stabilizing this region in other HIV antibodies could become an important approach to in silico optimization of antibodies.


Assuntos
Regiões Determinantes de Complementaridade/química , Anticorpos Anti-HIV/química , Proteína gp160 do Envelope de HIV/química , HIV-1 , Modelos Moleculares , Software , Regiões Determinantes de Complementaridade/genética , Regiões Determinantes de Complementaridade/imunologia , Anticorpos Anti-HIV/genética , Anticorpos Anti-HIV/imunologia , Proteína gp160 do Envelope de HIV/antagonistas & inibidores , Proteína gp160 do Envelope de HIV/genética , Proteína gp160 do Envelope de HIV/imunologia , Humanos , Estrutura Terciária de Proteína
6.
PLoS One ; 7(6): e38067, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22675507

RESUMO

Direct transmission of avian influenza viruses to mammals has become an increasingly investigated topic during the past decade; however, isolates that have been primarily investigated are typically ones originating from human or poultry outbreaks. Currently there is minimal comparative information on the behavior of the innumerable viruses that exist in the natural wild bird host. We have previously demonstrated the capacity of numerous North American avian influenza viruses isolated from wild birds to infect and induce lesions in the respiratory tract of mice. In this study, two isolates from shorebirds that were previously examined in mice (H1N9 and H6N1 subtypes) are further examined through experimental inoculations in the ferret with analysis of viral shedding, histopathology, and antigen localization via immunohistochemistry to elucidate pathogenicity and transmission of these viruses. Using sequence analysis and glycan binding analysis, we show that these avian viruses have the typical avian influenza binding pattern, with affinity for cell glycoproteins/glycolipids having terminal sialic acid (SA) residues with α 2,3 linkage [Neu5Ac(α2,3)Gal]. Despite the lack of α2,6 linked SA binding, these AIVs productively infected both the upper and lower respiratory tract of ferrets, resulting in nasal viral shedding and pulmonary lesions with minimal morbidity. Moreover, we show that one of the viruses is able to transmit to ferrets via direct contact, despite its binding affinity for α 2,3 linked SA residues. These results demonstrate that avian influenza viruses, which are endemic in aquatic birds, can potentially infect humans and other mammals without adaptation. Finally this work highlights the need for additional study of the wild bird subset of influenza viruses in regard to surveillance, transmission, and potential for reassortment, as they have zoonotic potential.


Assuntos
Adaptação Fisiológica/imunologia , Animais Selvagens/virologia , Furões/virologia , Vírus da Influenza A/isolamento & purificação , Vírus da Influenza A/fisiologia , Influenza Aviária/transmissão , Replicação Viral/fisiologia , Aminoácidos/metabolismo , Animais , Antígenos Virais/imunologia , Aves/virologia , Agregação Eritrocítica , Hemaglutinação , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Humanos , Vírus da Influenza A/patogenicidade , Influenza Aviária/imunologia , Influenza Aviária/patologia , Influenza Aviária/virologia , Influenza Humana/virologia , Camundongos , Dados de Sequência Molecular , Polissacarídeos/metabolismo , Receptores Virais/metabolismo , Sistema Respiratório/patologia , Sistema Respiratório/virologia , Especificidade da Espécie , Virulência , Eliminação de Partículas Virais
7.
PLoS One ; 6(6): e21183, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21731666

RESUMO

Avian influenza viruses (AIV) are an important emerging threat to public health. It is thought that sialic acid (sia) receptors are barriers in cross-species transmission where the binding preferences of AIV and human influenza viruses are sias α2,3 versus α2,6, respectively. In this study, we show that a normal fully differentiated, primary human bronchial epithelial cell model is readily infected by low pathogenic H5N1, H5N2 and H5N3 AIV, which primarily bind to sia α2,3 moieties, and replicate in these cells independent of specific sias on the cell surface. NHBE cells treated with neuraminidase prior to infection are infected by AIV despite removal of sia α2,3 moieties. Following AIV infection, higher levels of IP-10 and RANTES are secreted compared to human influenza virus infection, indicating differential chemokine expression patterns, a feature that may contribute to differences in disease pathogenesis between avian and human influenza virus infections in humans.


Assuntos
Aves/virologia , Brônquios/patologia , Células Epiteliais/virologia , Vírus da Influenza A/fisiologia , Influenza Aviária/virologia , Influenza Humana/virologia , Ácido N-Acetilneuramínico/metabolismo , Adolescente , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Quimiocinas/metabolismo , Cílios/efeitos dos fármacos , Cílios/metabolismo , Cílios/patologia , Cães , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Células Caliciformes/efeitos dos fármacos , Células Caliciformes/metabolismo , Células Caliciformes/patologia , Humanos , Vírus da Influenza A/efeitos dos fármacos , Influenza Aviária/patologia , Influenza Humana/patologia , Masculino , Neuraminidase/farmacologia , Receptores de Superfície Celular/metabolismo , Replicação Viral/efeitos dos fármacos , Replicação Viral/fisiologia , Eliminação de Partículas Virais/efeitos dos fármacos , Eliminação de Partículas Virais/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa