Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Virol ; 91(10)2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28250131

RESUMO

Measles virus (MeV) is a member of the family Paramixoviridae that causes a highly contagious respiratory disease but has emerged as a promising oncolytic platform. Previous studies of MeV entry focused on the identification of cellular receptors. However, the endocytic and trafficking pathways utilized during MeV entry remain poorly described. The contribution of each endocytic pathway has been examined in cells that express the MeV receptors SLAM (signaling lymphocyte-activating molecule) and PVRL4 (poliovirus receptor-like 4) (nectin-4). Recombinant MeVs expressing either firefly luciferase or green fluorescent protein together with a variety of inhibitors were used. The results showed that MeV uptake was dynamin independent in the Vero.hPVRL4, Vero.hSLAM, and PVRL4-positive MCF7 breast cancer cell lines. However, MeV infection was blocked by 5-(N-ethyl-N-propyl)amiloride (EIPA), the hallmark inhibitor of macropinocytosis, as well as inhibitors of actin polymerization. By using phalloidin staining, MeV entry was shown to induce actin rearrangements and the formation of membrane ruffles accompanied by transient elevated fluid uptake. Small interfering RNA (siRNA) knockdown of p21-activated kinase 1 (PAK1) demonstrated that MeV enters both Vero.hPVRL4 and Vero.hSLAM cells in a PAK1-independent manner using a macropinocytosis-like pathway. In contrast, MeV entry into MCF7 human breast cancer cells relied upon Rac1 and its effector PAK1 through a PVRL4-mediated macropinocytosis pathway. MeV entry into DLD-1 colon and HTB-20 breast cancer cells also appeared to use the same pathway. Overall, these findings provide new insight into the life cycle of MeV, which could lead to therapies that block virus entry or methods that improve the uptake of MeV by cancer cells during oncolytic therapy.IMPORTANCE In the past decades, measles virus (MeV) has emerged as a promising oncolytic platform. Previous studies concerning MeV entry focused mainly on the identification of putative receptors for MeV. Nectin-4 (PVRL4) was recently identified as the epithelial cell receptor for MeV. However, the specific endocytic and trafficking pathways utilized during MeV infections are poorly documented. In this study, we demonstrated that MeV enters host cells via a dynamin-independent and actin-dependent endocytic pathway. Moreover, we show that MeV gains entry into MCF7, DLD-1, and HTB-20 cancer cells through a PVRL4-mediated macropinocytosis pathway and identified the typical cellular GTPase and kinase involved. Our findings provide new insight into the life cycle of MeV, which may lead to the development of therapies that block the entry of the virus into the host cell or alternatively promote the uptake of oncolytic MeV into cancer cells.


Assuntos
Moléculas de Adesão Celular/metabolismo , Vírus do Sarampo/fisiologia , Pinocitose , Internalização do Vírus , Actinas/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Neoplasias da Mama , Linhagem Celular , Chlorocebus aethiops , Neoplasias do Colo , Células Epiteliais/virologia , Feminino , Humanos , Células MCF-7 , Vírus do Sarampo/efeitos dos fármacos , Vírus do Sarampo/genética , Vírus Oncolíticos/fisiologia , Pinocitose/efeitos dos fármacos , RNA Interferente Pequeno/genética , Células Vero , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
2.
J Virol ; 91(11)2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28331086

RESUMO

In addition to humans, only certain nonhuman primates are naturally susceptible to measles virus (MeV) infection. Disease severity is species dependent, ranging from mild to moderate for macaques to severe and even lethal for certain New World monkey species. To investigate if squirrel monkeys (Saimiri sciureus), which are reported to develop a course of disease similar to humans, may be better suited than macaques for the identification of virulence determinants or the evaluation of therapeutics, we infected them with a green fluorescent protein-expressing MeV. Compared to cynomolgus macaques (Macaca fascicularis) infected with the same virus, the squirrel monkeys developed more-severe immunosuppression, higher viral load, and a broader range of clinical signs typical for measles. In contrast, infection with an MeV unable to interact with the epithelial receptor nectin-4, while causing immunosuppression, resulted in only a mild and transient rash and a short-lived elevation of the body temperature. Similar titers of the wild-type and nectin-4-blind MeV were detected in peripheral blood mononuclear cells and lymph node homogenates, but only the wild-type virus was found in tracheal lavage fluids and urine. Thus, our study demonstrates the importance of MeV interactions with nectin-4 for clinical disease in the new and better-performing S. sciureus model of measles pathogenesis.IMPORTANCE The characterization of mechanisms underlying measles virus clinical disease has been hampered by the lack of an animal model that reproduces the course of disease seen in human patients. Here, we report that infection of squirrel monkeys (Saimiri sciureus) fulfills these requirements. Comparative infection with wild-type and epithelial cell receptor-blind viruses demonstrated the importance of epithelial cell infection for clinical disease, highlighting the spread to epithelia as an attractive target for therapeutic strategies.


Assuntos
Moléculas de Adesão Celular/metabolismo , Vírus do Sarampo/patogenicidade , Sarampo/virologia , Modelos Animais , Saimiri , Animais , Células Epiteliais/virologia , Proteínas de Fluorescência Verde , Humanos , Leucócitos Mononucleares/química , Leucócitos Mononucleares/virologia , Macaca fascicularis , Vírus do Sarampo/fisiologia , Nectinas , Carga Viral , Virulência
3.
J Virol ; 91(23)2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28904193

RESUMO

The inhibitors carbobenzoxy (Z)-d-Phe-l-Phe-Gly (fusion inhibitor peptide [FIP]) and 4-nitro-2-phenylacetyl amino-benzamide (AS-48) have similar efficacies in blocking membrane fusion and syncytium formation mediated by measles virus (MeV). Other homologues, such as Z-d-Phe, are less effective but may act through the same mechanism. In an attempt to map the site of action of these inhibitors, we generated mutant viruses that were resistant to the inhibitory effects of Z-d-Phe-l-Phe-Gly. These 10 mutations were localized to the heptad repeat B (HRB) region of the fusion protein, and no changes were observed in the viral hemagglutinin, which is the receptor attachment protein. Mutations were validated in a luciferase-based membrane fusion assay, using transfected fusion and hemagglutinin expression plasmids or with syncytium-based assays in Vero, Vero-SLAM, and Vero-Nectin 4 cell lines. The changes I452T, D458N, D458G/V459A, N462K, N462H, G464E, and I483R conferred resistance to both FIP and AS-48 without compromising membrane fusion. The inhibitors did not block hemagglutinin protein-mediated binding to the target cell. Edmonston vaccine/laboratory and IC323 wild-type strains were equally affected by the inhibitors. Escape mutations were mapped upon a three-dimensional (3D) structure modeled from the published crystal structure of parainfluenzavirus 5 fusion protein. The most effective mutations were situated in a region located near the base of the globular head and its junction with the alpha-helical stalk of the prefusion protein. We hypothesize that the fusion inhibitors could interfere with the structural changes that occur between the prefusion and postfusion conformations of the fusion protein.IMPORTANCE Due to lapses in vaccination worldwide that have caused localized outbreaks, measles virus (MeV) has regained importance as a pathogen. Antiviral agents against measles virus are not commercially available but could be useful in conjunction with MeV eradication vaccine programs and as a safeguard in oncolytic viral therapy. Three decades ago, the small hydrophobic peptide Z-d-Phe-l-Phe-Gly (FIP) was shown to block MeV infections and syncytium formation in monkey kidney cell lines. The exact mechanism of its action has yet to be determined, but it does appear to have properties similar to those of another chemical inhibitor, AS-48, which appears to interfere with the conformational change in the viral F protein that is required to elicit membrane fusion. Escape mutations were used to map the site of action for FIP. Knowledge gained from these studies could help in the design of new inhibitors against morbilliviruses and provide additional knowledge concerning the mechanism of virus-mediated membrane fusion.


Assuntos
Vírus do Sarampo/efeitos dos fármacos , Vírus do Sarampo/genética , Mutação , Oligopeptídeos/farmacologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo , Animais , Antivirais/farmacologia , Benzamidas/farmacologia , Chlorocebus aethiops , Hemaglutininas Virais/genética , Hemaglutininas Virais/metabolismo , Fusão de Membrana/efeitos dos fármacos , Modelos Moleculares , Ligação Proteica , Células Vero , Proteínas Virais de Fusão/química , Internalização do Vírus/efeitos dos fármacos
4.
J Virol ; 87(5): 2526-34, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23255790

RESUMO

After the contagion measles virus (MV) crosses the respiratory epithelium within myeloid cells that express the primary receptor signaling lymphocytic activation molecule (SLAM), it replicates briskly in SLAM-expressing cells in lymphatic organs. Later, the infection spreads to epithelia expressing nectin-4, an adherens junction protein expressed preferentially in the trachea, but how it gets there is not understood. To characterize the mechanisms of spread, we infected groups of 5 or 6 cynomolgus monkeys (Macaca fascicularis) with either a wild-type MV or its "N4-blind" derivative, which is unable to enter nectin-4-expressing cells because of the targeted mutation of two hemagglutinin residues. As expected, both viruses caused similar levels of immunosuppression, as monitored by reductions in white blood cell counts and lymphocyte proliferation activity. However, monkeys infected with the N4-blind MV cleared infection more rapidly. Wild-type virus-infected monkeys secreted virus, while marginal virus titers were detected in tracheal lavage fluid cells of N4-blind MV-infected hosts. Analyses of tracheal rings obtained at necropsy (day 12) documented widespread infection of individual cells or small cell clusters in the subepithelial lamina propria of monkeys infected with either virus. However, only wild-type MV spread to the epithelium, forming numerous infectious centers comprised of many contiguous columnar cells. Infected CD11c(+) myeloid (macrophage or dendritic) cells were frequently observed in the lamina propria below epithelial infectious centers. Thus, MV may use myeloid cells as vehicles not only immediately after contagion but also to infect epithelia of tissues expressing nectin-4, including the trachea.


Assuntos
Moléculas de Adesão Celular/metabolismo , Macaca fascicularis/virologia , Vírus do Sarampo/fisiologia , Mucosa/imunologia , Mucosa Respiratória/virologia , Traqueia/imunologia , Traqueia/virologia , Animais , Antígenos CD/biossíntese , Moléculas de Adesão Celular/biossíntese , Chlorocebus aethiops/imunologia , Chlorocebus aethiops/virologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Células Epiteliais/virologia , Feminino , Proteínas de Fluorescência Verde/metabolismo , Terapia de Imunossupressão , Macrófagos/imunologia , Macrófagos/virologia , Sarampo/metabolismo , Sarampo/virologia , Vírus do Sarampo/genética , Vírus do Sarampo/metabolismo , Mucosa/virologia , Receptores de Superfície Celular/biossíntese , Receptores Virais/metabolismo , Mucosa Respiratória/imunologia , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Células Vero
5.
J Virol ; 86(16): 8527-35, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22647692

RESUMO

In the context of viral infections, autophagy induction can be beneficial or inhibitory. Within the Paramyxoviridae family, only morbilliviruses have been investigated and are reported to induce autophagy. Here we show that morbilliviruses rapidly induce autophagy and require this induction for efficient cell-to-cell spread. Coexpression of both glycoproteins in cells expressing one of the cellular receptors was required for autophagy induction, and LC3 punctum formation, indicative of autophagy, was mainly observed in syncytia. A similar correlation between syncytium formation and autophagy induction was also observed for other paramyxovirus glycoproteins, suggesting that membrane fusion-mediated autophagy may be common among paramyxoviruses and possibly other enveloped viruses.


Assuntos
Autofagia , Células Gigantes/virologia , Fusão de Membrana , Morbillivirus/patogenicidade , Animais , Chlorocebus aethiops , Células Vero
6.
Virologie (Montrouge) ; 16(3): 158-167, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-33065872

RESUMO

Morbilliviruses cause a severe and sometimes lethal disease in their respective hosts, which is characterized by a generalized immunosuppression, respiratory and gastro-intestinal clinical signs, and occasional neurological complications. This similarity in the biology of different members of the morbillivirus genus constitutes the basis for the study of canine distemper virus in its natural hosts as a model for the characterization of morbillivirus pathogenesis. In combination with the reverse genetics technology, which allows the production of recombinant viruses carrying specific genetic modifications, this model has made important contributions to our understanding of the mechanisms underlying morbillivirus immunosuppression, dissemination, and neuroinvasion.

7.
J Virol ; 84(6): 3004-15, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20071589

RESUMO

Ebola virus (EBOV) protein VP35 is a double-stranded RNA (dsRNA) binding inhibitor of host interferon (IFN)-alpha/beta responses that also functions as a viral polymerase cofactor. Recent structural studies identified key features, including a central basic patch, required for VP35 dsRNA binding activity. To address the functional significance of these VP35 structural features for EBOV replication and pathogenesis, two point mutations, K319A/R322A, that abrogate VP35 dsRNA binding activity and severely impair its suppression of IFN-alpha/beta production were identified. Solution nuclear magnetic resonance (NMR) spectroscopy and X-ray crystallography reveal minimal structural perturbations in the K319A/R322A VP35 double mutant and suggest that loss of basic charge leads to altered function. Recombinant EBOVs encoding the mutant VP35 exhibit, relative to wild-type VP35 viruses, minimal growth attenuation in IFN-defective Vero cells but severe impairment in IFN-competent cells. In guinea pigs, the VP35 mutant virus revealed a complete loss of virulence. Strikingly, the VP35 mutant virus effectively immunized animals against subsequent wild-type EBOV challenge. These in vivo studies, using recombinant EBOV viruses, combined with the accompanying biochemical and structural analyses directly correlate VP35 dsRNA binding and IFN inhibition functions with viral pathogenesis. Moreover, these studies provide a framework for the development of antivirals targeting this critical EBOV virulence factor.


Assuntos
Ebolavirus/genética , Ebolavirus/patogenicidade , Cobaias/virologia , Mutação Puntual , RNA de Cadeia Dupla/metabolismo , Proteínas Virais Reguladoras e Acessórias , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Ebolavirus/imunologia , Feminino , Humanos , Fatores Imunológicos/genética , Fatores Imunológicos/imunologia , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/genética , Interferon beta/imunologia , Interferons/antagonistas & inibidores , Interferons/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , RNA de Cadeia Dupla/genética , Alinhamento de Sequência , Células Vero , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/metabolismo
8.
J Virol ; 83(18): 9596-601, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19587051

RESUMO

Ebolavirus (EBOV) is the etiological agent of a severe hemorrhagic fever with a high mortality rate. The spike glycoprotein (GP) is believed to be one of the major determinants of virus pathogenicity. In this study, we demonstrated the molecular mechanism responsible for the downregulation of surface markers caused by EBOV GP expression. We showed that expression of mature GP on the plasma membrane results in the masking of cellular surface proteins, including major histocompatibility complex class I. Overexpression of GP also results in the masking of certain antigenic epitopes on GP itself, causing an illusory effect of disappearance from the plasma membrane.


Assuntos
Ebolavirus/patogenicidade , Epitopos , Proteínas de Membrana , Proteínas do Envelope Viral/fisiologia , Linhagem Celular , Membrana Celular/virologia , Regulação para Baixo , Ebolavirus/química , Antígenos de Histocompatibilidade Classe I , Humanos , Transfecção , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
9.
J Clin Virol ; 31(2): 140-7, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15364271

RESUMO

BACKGROUND: High risk human papillomavirus (HR-HPV) load determined by quantitative methods has already been considered as highly predictive of future development of high grade cervical lesions. Some studies also demonstrated that Hybrid Capture II (HCII) results can be considered as a reflection of HPV DNA load, while others did not. HCI assay, well suited for routine HR-HPV screening, is not especially dedicated for quantitative use. However, we have recently shown that women with high viral loads assessed by HCII were at increased risk of cervical precancer. OBJECTIVES: The aim of the study was to determine if the values given by the HCII assay can be considered as quantitative. STUDY DESIGN: We used a real-time PCR allowing precise quantification of both HPV16 genome and albumin gene to normalize the measuring HPV16 load in cervical cells and to compare the data with those obtained by HCIIin a series of 40 HR-HPV positive samples. RESULTS: Reproducibility of the HPV16 real-time PCR, assessed from nine independent experiments of serial dilutions of SiHa cell DNA, was reflected in coefficients of variation for standard curves of crossing point (Cp) values below 5%. The HPV16 loads with a broad individual variability were significantly related to the cumulative load estimated by HCII and did not depend on the cellularity of samples. CONCLUSIONS: We assume that the HCII values can be used as a quantitative measure of HR-HPV DNA, so long as cervical specimens are collected using standardized protocols.


Assuntos
Hibridização de Ácido Nucleico/métodos , Papillomaviridae/genética , Papillomaviridae/isolamento & purificação , Reação em Cadeia da Polimerase/métodos , Sequência de Bases , Sondas de DNA de HPV/genética , DNA Viral/análise , DNA Viral/genética , Feminino , Humanos , Papillomaviridae/classificação , Papillomaviridae/patogenicidade , Infecções por Papillomavirus/diagnóstico , Infecções por Papillomavirus/virologia , Esfregaço Vaginal , Virologia/métodos
10.
Virology ; 454-455: 109-17, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24725937

RESUMO

The entry of canine distemper virus (CDV) is a multistep process that involves the attachment of CDV hemagglutinin (H) to its cellular receptor, followed by fusion between virus and cell membranes. Our laboratory recently identified PVRL4 (nectin-4) to be the epithelial receptor for measles and canine distemper viruses. In this study, we demonstrate that the V domain of PVRL4 is critical for CDV entry and virus cell-to-cell spread. Furthermore, four key amino acid residues within the V domain of dog PVRL4 and two within the CDV hemagglutinin were shown to be essential for receptor-mediated virus entry.


Assuntos
Moléculas de Adesão Celular/metabolismo , Vírus da Cinomose Canina/fisiologia , Interações Hospedeiro-Patógeno , Receptores Virais/metabolismo , Internalização do Vírus , Animais , Cães , Hemaglutininas Virais/metabolismo
11.
Viruses ; 6(6): 2268-86, 2014 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-24892636

RESUMO

PVRL4 (nectin-4) was recently identified as the epithelial receptor for members of the Morbillivirus genus, including measles virus, canine distemper virus and peste des petits ruminants virus. Here, we describe the role of PVRL4 in morbillivirus pathogenesis and its promising use in cancer therapies. This discovery establishes a new paradigm for the spread of virus from lymphocytes to airway epithelial cells and its subsequent release into the environment. Measles virus vaccine strains have emerged as a promising oncolytic platform for cancer therapy in the last ten years. Given that PVRL4 is a well-known tumor-associated marker for several adenocarcinoma (lung, breast and ovary), the measles virus could potentially be used to specifically target, infect and destroy cancers expressing PVRL4.


Assuntos
Biomarcadores Tumorais , Moléculas de Adesão Celular/metabolismo , Células Epiteliais/metabolismo , Morbillivirus/fisiologia , Receptores Virais/metabolismo , Animais , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/genética , Humanos , Vírus do Sarampo/fisiologia , Infecções por Morbillivirus/imunologia , Infecções por Morbillivirus/metabolismo , Infecções por Morbillivirus/virologia , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Receptores Virais/química , Receptores Virais/genética , Internalização do Vírus
12.
Virology ; 436(1): 210-20, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23260107

RESUMO

Canine distemper virus (CDV) was shown to use dog nectin-4 as a receptor to gain entry into epithelial cells. RNA from dog placenta or MDCK kidney cells was isolated and cDNAs were prepared. Two splice variants of dog nectin-4 were identified. A deletion of 25 amino acids was found in the cytoplasmic domain of dog nectin-4 from MDCK cells, corresponding to a splice variant that is also seen in murine nectin-4, and did not affect its role as a receptor. Both dog nectin-4 and human nectin-4 could function as an entry factor for CDV containing an EGFP reporter gene. Inhibition of dog nectin-4 expression by RNAi or nectin-4 antibodies decreased CDV titers and EGFP fluorescence. Finally, dog nectin-4 also promotes syncytia formation, which could be inhibited by siRNA treatment. These data confirm that dog nectin-4 can be used by CDV to gain entry into epithelial cells, and facilitate virus spread.


Assuntos
Moléculas de Adesão Celular/metabolismo , Vírus da Cinomose Canina/fisiologia , Células Epiteliais/virologia , Células Gigantes/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Animais , Anticorpos/imunologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Chlorocebus aethiops , Vírus da Cinomose Canina/genética , Vírus da Cinomose Canina/imunologia , Cães , Feminino , Células Gigantes/virologia , Proteínas de Fluorescência Verde/genética , Humanos , Células MCF-7 , Células Madin Darby de Rim Canino , Dados de Sequência Molecular , Nectinas , Placenta , Gravidez , Isoformas de Proteínas/genética , Interferência de RNA , RNA Interferente Pequeno , Receptores Virais , Deleção de Sequência , Células Vero
13.
Curr Opin Virol ; 2(6): 773-83, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23146309

RESUMO

This review takes a general approach to describing host cell factors that facilitate measles virus (MeV) infection and replication. It relates our current understanding of MeV entry receptors, with emphasis on how these host cell surface proteins contribute to pathogenesis within its host. The roles of SLAM/CD150 lymphocyte receptor and the newly discovered epithelial receptor PVRL4/nectin-4 are highlighted. Host cell factors such as HSP72, Prdx1, tubulin, casein kinase, and actin, which are known to impact viral RNA synthesis and virion assembly, are also discussed. Finally the review describes strategies used by measles virus to circumvent innate immunity and confound the effects of interferon within the host cell. Proteomic studies and genome wide RNAi screens will undoubtedly advance our knowledge in the future.


Assuntos
Interações Hospedeiro-Patógeno , Vírus do Sarampo/fisiologia , Internalização do Vírus , Replicação Viral , Humanos , Evasão da Resposta Imune , Vírus do Sarampo/imunologia , Vírus do Sarampo/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA