Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Med Virol ; 96(4): e29620, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38647027

RESUMO

Vertical transmission has been described following monkeypox virus (MPXV) infection in pregnant women. The presence of MPXV has been reported in the placenta from infected women, but whether pathogens colonize placenta remains unexplored. We identify trophoblasts as a target cell for MPXV replication. In a pan-microscopy approach, we decipher the specific infectious cycle of MPXV and inner cellular structures in trophoblasts. We identified the formation of a specialized region for viral morphogenesis and replication in placental cells. We also reported infection-induced cellular remodeling. We found that MPXV stimulates cytoskeleton reorganization with intercellular extensions for MPXV cell spreading specifically to trophoblastic cells. Altogether, the specific infectious cycle of MPXV in trophoblast cells and these protrusions that were structurally and morphologically similar to filopodia reveal new insights into the infection of MPXV.


Assuntos
Monkeypox virus , Pseudópodes , Trofoblastos , Trofoblastos/virologia , Humanos , Pseudópodes/virologia , Feminino , Gravidez , Monkeypox virus/fisiologia , Liberação de Vírus , Replicação Viral , Citoesqueleto/virologia , Placenta/virologia , Placenta/citologia , Vírion/ultraestrutura , Microscopia/métodos , Linhagem Celular
2.
Acta Virol ; 64(2): 154-166, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32551784

RESUMO

The most frequent way of virus dissemination is through the canonical receptor-mediated pathway. However, when unfavorable conditions, such as presence of antibodies appear, the viruses use more peculiar routes of transmission to protect themselves. Here we describe most of the routes, from syncytia formation, tunneling nanotubes and filopodia, through immunological and virological synapses to actin comets formation. We describe the cell-to-cell transport in different viral families to show that this way of virus distribution is present in almost all the mammalian virus families and is not as uncommon as it was thought. The knowledge of the ways of viral transport might lead us to exploit more successful therapeutic approaches and fight the most threatening diseases. Keywords: cell-to-cell transmission; tunneling nanotubes; viral synapses; filopodia; actin comets.


Assuntos
Actinas , Células Gigantes/virologia , Sinapses Imunológicas/virologia , Pseudópodes/virologia , Fenômenos Fisiológicos Virais , Vírus/patogenicidade , Animais
3.
Virol J ; 15(1): 182, 2018 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-30477508

RESUMO

Cellular protein eukaryotic translation elongation factor 1A (eEF1A) is an actin binding protein that plays a role in the formation of filamentous actin (F-actin) bundles. F-Actin regulates multiple stages of respiratory syncytial virus (RSV) replication including assembly and budding. Our previous study demonstrated that eEF1A knock-down significantly reduced RSV replication. Here we investigated if the eEF1A function in actin bundle formation was important for RSV replication and release. To investigate this, eEF1A function was impaired in HEp-2 cells by either knock-down of eEF1A with siRNA, or treatment with an eEF1A inhibitor, didemnin B (Did B). Cell staining and confocal microscopy analysis showed that both eEF1A knock-down and treatment with Did B resulted in disruption of cellular stress fiber formation and elevated accumulation of F-actin near the plasma membrane. When treated cells were then infected with RSV, there was also reduced formation of virus-induced cellular filopodia. Did B treatment, similarly to eEF1A knock-down, reduced the release of infectious RSV, but unlike eEF1A knock-down, did not significantly affect RSV genome replication. The lower infectious virus production in Did B treated cells also reduced RSV-induced cell death. In conclusion, the cellular factor eEF1A plays an important role in the regulation of F-actin stress fiber formation required for RSV assembly and release.


Assuntos
Actinas/metabolismo , Fator 1 de Elongação de Peptídeos/genética , Vírus Sincicial Respiratório Humano/fisiologia , Fibras de Estresse/fisiologia , Replicação Viral , Actinas/genética , Linhagem Celular Tumoral , Depsipeptídeos/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Técnicas de Silenciamento de Genes , Humanos , Pseudópodes/fisiologia , Pseudópodes/virologia , Vírus Sincicial Respiratório Humano/genética
4.
Front Immunol ; 9: 260, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29515578

RESUMO

While HIV-1 infection of target cells with cell-free viral particles has been largely documented, intercellular transmission through direct cell-to-cell contact may be a predominant mode of propagation in host. To spread, HIV-1 infects cells of the immune system and takes advantage of their specific particularities and functions. Subversion of intercellular communication allows to improve HIV-1 replication through a multiplicity of intercellular structures and membrane protrusions, like tunneling nanotubes, filopodia, or lamellipodia-like structures involved in the formation of the virological synapse. Other features of immune cells, like the immunological synapse or the phagocytosis of infected cells are hijacked by HIV-1 and used as gateways to infect target cells. Finally, HIV-1 reuses its fusogenic capacity to provoke fusion between infected donor cells and target cells, and to form infected syncytia with high capacity of viral production and improved capacities of motility or survival. All these modes of cell-to-cell transfer are now considered as viral mechanisms to escape immune system and antiretroviral therapies, and could be involved in the establishment of persistent virus reservoirs in different host tissues.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Sinapses Imunológicas/virologia , Junções Intercelulares/virologia , Animais , Linfócitos T CD4-Positivos/virologia , Reservatórios de Doenças , Infecções por HIV/transmissão , Humanos , Evasão da Resposta Imune , Fusão de Membrana , Nanotubos/virologia , Pseudópodes/virologia
5.
Viruses ; 10(2)2018 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-29473915

RESUMO

Herpes simplex virus type 1 (HSV-1) is a neuroinvasive human pathogen that has the ability to infect and replicate within epithelial cells and neurons and establish a life-long latent infection in sensory neurons. HSV-1 depends on the host cellular cytoskeleton for entry, replication, and exit. Therefore, HSV-1 has adapted mechanisms to promote its survival by exploiting the microtubule and actin cytoskeletons to direct its active transport, infection, and spread between neurons and epithelial cells during primary and recurrent infections. This review will focus on the currently known mechanisms utilized by HSV-1 to harness the neuronal cytoskeleton, molecular motors, and the secretory and exocytic pathways for efficient virus entry, axonal transport, replication, assembly, and exit from the distinct functional compartments (cell body and axon) of the highly polarized sensory neurons.


Assuntos
Citoesqueleto/metabolismo , Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Interações Hospedeiro-Patógeno , Neurônios/virologia , Actinas/metabolismo , Animais , Transporte Axonal , Axônios/metabolismo , Axônios/virologia , Gânglios Espinais/metabolismo , Gânglios Espinais/virologia , Humanos , Microtúbulos/metabolismo , Proteínas Motores Moleculares/metabolismo , Pseudópodes/metabolismo , Pseudópodes/virologia , Montagem de Vírus , Internalização do Vírus , Replicação Viral
6.
J Virol ; 92(2)2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29093086

RESUMO

Cell motility and migration is a complex, multistep, and multicomponent process intrinsic to progression and metastasis. Motility is dependent on the activities of integrin receptors and Rho family GTPases, resulting in the remodeling of the actin cytoskeleton and formation of various motile actin-based protrusions. Merkel cell carcinoma (MCC) is an aggressive skin cancer with a high likelihood of recurrence and metastasis. Merkel cell polyomavirus (MCPyV) is associated with the majority of MCC cases, and MCPyV-induced tumorigenesis largely depends on the expression of the small tumor antigen (ST). Since the discovery of MCPyV, a number of mechanisms have been suggested to account for replication and tumorigenesis, but to date, little is known about potential links between MCPyV T antigen expression and the metastatic nature of MCC. Previously, we described the action of MCPyV ST on the microtubule network and how it impacts cell motility and migration. Here, we demonstrate that MCPyV ST affects the actin cytoskeleton to promote the formation of filopodia through a mechanism involving the catalytic subunit of protein phosphatase 4 (PP4C). We also show that MCPyV ST-induced cell motility is dependent upon the activities of the Rho family GTPases Cdc42 and RhoA. In addition, our results indicate that the MCPyV ST-PP4C interaction results in the dephosphorylation of ß1 integrin, likely driving the cell motility pathway. These findings describe a novel mechanism by which a tumor virus induces cell motility, which may ultimately lead to cancer metastasis, and provides opportunities and strategies for targeted interventions for disseminated MCC.IMPORTANCE Merkel cell polyomavirus (MCPyV) is the most recently discovered human tumor virus. It causes the majority of cases of Merkel cell carcinoma (MCC), an aggressive skin cancer. However, the molecular mechanisms implicating MCPyV-encoded proteins in cancer development are yet to be fully elucidated. This study builds upon our previous observations, which demonstrated that the MCPyV ST antigen enhances cell motility, providing a potential link between MCPyV protein expression and the highly metastatic nature of MCC. Here, we show that MCPyV ST remodels the actin cytoskeleton, promoting the formation of filopodia, which is essential for MCPyV ST-induced cell motility, and we also implicate the activity of specific Rho family GTPases, Cdc42 and RhoA, in these processes. Moreover, we describe a novel mechanism for the activation of Rho-GTPases and the cell motility pathway due to the interaction between MCPyV ST and the cellular phosphatase catalytic subunit PP4C, which leads to the specific dephosphorylation of ß1 integrin. These findings may therefore provide novel strategies for therapeutic intervention for disseminated MCC.


Assuntos
Antígenos Virais de Tumores/imunologia , Movimento Celular , Poliomavírus das Células de Merkel/fisiologia , Pseudópodes/metabolismo , Pseudópodes/virologia , Proteínas rho de Ligação ao GTP/metabolismo , Actinas/metabolismo , Antígenos Virais de Tumores/genética , Carcinoma de Célula de Merkel/virologia , Expressão Gênica , Humanos , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Infecções por Polyomavirus/virologia , Ligação Proteica , Infecções Tumorais por Vírus/virologia
7.
PLoS Pathog ; 12(12): e1006062, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27926942

RESUMO

Human respiratory syncytial virus (RSV) is an enveloped RNA virus that is the most important viral cause of acute pediatric lower respiratory tract illness worldwide, and lacks a vaccine or effective antiviral drug. The involvement of host factors in the RSV replicative cycle remains poorly characterized. A genome-wide siRNA screen in human lung epithelial A549 cells identified actin-related protein 2 (ARP2) as a host factor involved in RSV infection. ARP2 knockdown did not reduce RSV entry, and did not markedly reduce gene expression during the first 24 hr of infection, but decreased viral gene expression thereafter, an effect that appeared to be due to inhibition of viral spread to neighboring cells. Consistent with reduced spread, there was a 10-fold reduction in the release of infectious progeny virions in ARP2-depleted cells at 72 hr post-infection. In addition, we found that RSV infection induced filopodia formation and increased cell motility in A549 cells and that this phenotype was ARP2 dependent. Filopodia appeared to shuttle RSV to nearby uninfected cells, facilitating virus spread. Expression of the RSV F protein alone from a plasmid or heterologous viral vector in A549 cells induced filopodia, indicating a new role for the RSV F protein, driving filopodia induction and virus spread. Thus, this study identified roles for ARP2 and filopodia in RSV-induced cell motility, RSV production, and RSV cell-to-cell spread.


Assuntos
Proteína 2 Relacionada a Actina/metabolismo , Pseudópodes/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/patogenicidade , Células A549 , Western Blotting , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Humanos , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Pseudópodes/ultraestrutura , Reação em Cadeia da Polimerase em Tempo Real , Internalização do Vírus
8.
Mol Cell Biol ; 33(15): 3036-49, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23732912

RESUMO

The mechanisms underlying the cellular entry of the HIV-1 Tat protein transduction domain (TatP) and the molecular information necessary to improve the transduction efficiency of TatP remain unclear due to the technical limitations for direct visualization of TatP's behavior in cells. Using confocal microscopy, total internal reflection fluorescence microscopy, and four-dimensional microscopy, we developed a single-molecule tracking assay for TatP labeled with quantum dots (QDs) to examine the kinetics of TatP initially and immediately before, at the beginning of, and immediately after entry into living cells. We report that even when the number of multivalent TatP (mTatP)-QDs bound to a cell was low, each single mTatP-QD first locally induced the cell's lateral transport machinery to move the mTatP-QD toward the center of the cell body upon cross-linking of heparan sulfate proteoglycans. The centripetal and lateral movements were linked to the integrity and flow of actomyosin and microtubules. Individual mTatP underwent lipid raft-mediated temporal confinement, followed by complete immobilization, which ultimately led to endocytotic internalization. However, bivalent TatP did not sufficiently promote either cell surface movement or internalization. Together, these findings provide clues regarding the mechanisms of TatP cell entry and indicate that increasing the valence of TatP on nanoparticles allows them to behave as cargo delivery nanomachines.


Assuntos
Endocitose , Infecções por HIV/virologia , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Internalização do Vírus , Produtos do Gene tat do Vírus da Imunodeficiência Humana/análise , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Actinas/metabolismo , Infecções por HIV/metabolismo , Células HeLa , Humanos , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/virologia , Microscopia Confocal/métodos , Microtúbulos/metabolismo , Microtúbulos/virologia , Imagem Molecular/métodos , Pseudópodes/metabolismo , Pseudópodes/virologia , Pontos Quânticos
9.
PLoS Pathog ; 8(6): e1002762, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22685410

RESUMO

Paramount to the success of persistent viral infection is the ability of viruses to navigate hostile environments en route to future targets. In response to such obstacles, many viruses have developed the ability of establishing actin rich-membrane bridges to aid in future infections. Herein through dynamic imaging of HIV infected dendritic cells, we have observed how viral high-jacking of the actin/membrane network facilitates one of the most efficient forms of HIV spread. Within infected DC, viral egress is coupled to viral filopodia formation, with more than 90% of filopodia bearing immature HIV on their tips at extensions of 10 to 20 µm. Live imaging showed HIV filopodia routinely pivoting at their base, and projecting HIV virions at µm.sec⁻¹ along repetitive arc trajectories. HIV filopodial dynamics lead to up to 800 DC to CD4 T cell contacts per hour, with selection of T cells culminating in multiple filopodia tethering and converging to envelope the CD4 T-cell membrane with budding HIV particles. Long viral filopodial formation was dependent on the formin diaphanous 2 (Diaph2), and not a dominant Arp2/3 filopodial pathway often associated with pathogenic actin polymerization. Manipulation of HIV Nef reduced HIV transfer 25-fold by reducing viral filopodia frequency, supporting the potency of DC HIV transfer was dependent on viral filopodia abundance. Thus our observations show HIV corrupts DC to CD4 T cell interactions by physically embedding at the leading edge contacts of long DC filopodial networks.


Assuntos
Proteínas de Transporte/metabolismo , Células Dendríticas/virologia , HIV , Interações Hospedeiro-Parasita/fisiologia , Pseudópodes/virologia , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Forminas , Humanos , Microscopia Eletrônica de Transmissão , Pseudópodes/ultraestrutura , Vírion/metabolismo , Vírion/ultraestrutura
10.
Proc Natl Acad Sci U S A ; 107(20): 9346-51, 2010 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-20439710

RESUMO

To enter host cells, vaccinia virus, a prototype poxvirus, can induce transient macropinocytosis followed by endocytic internalization and penetration through the limiting membrane of pinosomes by membrane fusion. Although mature virions (MVs) of the Western reserve (WR) strain do this in HeLa cells by activating transient plasma membrane blebbing, MVs from the International Health Department-J strain were found to induce rapid formation (and lengthening) of filopodia. When the signaling pathways underlying these responses were compared, differences were observed at the level of Rho GTPases. Key to the filopodial formation was the virus-induced activation of Cdc42, and for the blebbing response the activation of Rac1. In addition, unlike WR, International Health Department-J MVs did not rely on genistein-sensitive tyrosine kinase and PI(3)K activities. Only WR MVs had membrane fusion activity at low pH. Inhibitor profiling showed that MVs from both strains entered cells by macropinocytosis and that this was induced by virion-exposed phosphatidylserine. Both MVs relied on the activation of epidermal growth factor receptor, on serine/threonine kinases, protein kinase C, and p21-activated kinase 1. The results showed that different strains of the same virus can elicit dramatically different responses in host cells during entry, and that different macropinocytic mechanisms are possible in the same cell line through subtle differences in the activating ligand.


Assuntos
Pinocitose/fisiologia , Transdução de Sinais/fisiologia , Vaccinia virus/fisiologia , Internalização do Vírus , Proteínas rho de Ligação ao GTP/metabolismo , Receptores ErbB/metabolismo , Citometria de Fluxo , Proteínas de Fluorescência Verde/metabolismo , Concentração de Íons de Hidrogênio , Pseudópodes/virologia , RNA Interferente Pequeno/genética , Especificidade da Espécie , Transfecção
11.
Biochem Biophys Res Commun ; 391(1): 176-81, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19909728

RESUMO

Heparan sulfate (HS) moieties on cell surfaces are known to provide attachment sites for many viruses including herpes simplex virus type-1 (HSV-1). Here, we demonstrate that cells respond to HSV-1 infection by enhancing filopodia formation. Filopodia express HS and are subsequently utilized for the transport of HSV-1 virions to cell bodies in a surfing-like phenomenon, which is facilitated by the underlying actin cytoskeleton and is regulated by transient activation of a small Rho GTPase, Cdc42. We also demonstrate that interaction between a highly conserved herpesvirus envelope glycoprotein B (gB) and HS is required for surfing. A HSV-1 mutant that lacks gB fails to surf and quantum dots conjugated with gB demonstrate surfing-like movements. Our data demonstrates a novel use of a common receptor, HS, which could also be exploited by multiple viruses and quite possibly, many additional ligands for transport along the plasma membrane.


Assuntos
Heparitina Sulfato/fisiologia , Herpesvirus Humano 1/fisiologia , Pseudópodes/virologia , Proteínas do Envelope Viral/fisiologia , Ligação Viral , Internalização do Vírus , Membrana Celular/virologia , Herpesvirus Humano 1/genética , Humanos , Mutação , Pontos Quânticos , Proteínas do Envelope Viral/genética
12.
J Virol ; 83(20): 10504-14, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19656887

RESUMO

Canine parvovirus (CPV) and feline panleukopenia virus (FPV) are closely related parvoviruses that differ in their host ranges for cats and dogs. Both viruses bind their host transferrin receptor (TfR), enter cells by clathrin-mediated endocytosis, and traffic with that receptor through endosomal pathways. Infection by these viruses appears to be inefficient and slow, with low numbers of virions infecting the cell after a number of hours. Species-specific binding to TfR controls viral host range, and in this study FPV and strains of CPV differed in the levels of cell attachment, uptake, and infection in canine and feline cells. During infection, CPV particles initially bound and trafficked passively on the filopodia of canine cells while they bound to the cell body of feline cells. That binding was associated with the TfR as it was disrupted by anti-TfR antibodies. Capsids were taken up from the cell surface with different kinetics in canine and feline cells but, unlike transferrin, most did not recycle. Capsids labeled with fluorescent markers were seen in Rab5-, Rab7-, or Rab11-positive endosomal compartments within minutes of uptake, but reached the nucleus. Constitutively active or dominant negative Rab mutants changed the intracellular distribution of capsids and affected the infectivity of virus in cells.


Assuntos
Células/virologia , Vírus da Panleucopenia Felina/patogenicidade , Interações Hospedeiro-Patógeno , Parvovirus Canino/patogenicidade , Pseudópodes/virologia , Receptores da Transferrina/metabolismo , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Gatos , Linhagem Celular , Cães , Endossomos/fisiologia , Vírus da Panleucopenia Felina/metabolismo , Parvovirus Canino/metabolismo , Receptores Virais/metabolismo , Especificidade da Espécie , Proteínas rab de Ligação ao GTP/metabolismo
13.
J Virol ; 83(12): 6234-46, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19369333

RESUMO

Human immunodeficiency virus type 1 (HIV-1) efficiently propagates through cell-to-cell contacts, which include virological synapses (VS), filopodia, and nanotubes. Here, we quantified and characterized further these diverse modes of contact in lymphocytes. We report that viral transmission mainly occurs across VS and through "polysynapses," a rosette-like structure formed between one infected cell and multiple adjacent recipients. Polysynapses are characterized by simultaneous HIV clustering and transfer at multiple membrane regions. HIV Gag proteins often adopt a ring-like supramolecular organization at sites of intercellular contacts and colocalize with CD63 tetraspanin and raft components GM1, Thy-1, and CD59. In donor cells engaged in polysynapses, there is no preferential accumulation of Gag proteins at contact sites facing the microtubule organizing center. The LFA-1 adhesion molecule, known to facilitate viral replication, enhances formation of polysynapses. Altogether, our results reveal an underestimated mode of viral transfer through polysynapses. In HIV-infected individuals, these structures, by promoting concomitant infection of multiple targets in the vicinity of infected cells, may facilitate exponential viral growth and escape from immune responses.


Assuntos
Linfócitos T CD4-Positivos/virologia , Infecções por HIV/virologia , HIV-1/fisiologia , Antígeno-1 Associado à Função Linfocitária/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Animais , Linfócitos T CD4-Positivos/ultraestrutura , Feminino , Humanos , Células Jurkat , Macaca , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Pseudópodes/virologia , Replicação Viral , Produtos do Gene env do Vírus da Imunodeficiência Humana/metabolismo
14.
FEBS J ; 275(21): 5272-85, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18803666

RESUMO

Herpes simplex virus 1 (HSV-1) demonstrates a unique ability to infect a variety of host cell types. Retinal pigment epithelial (RPE) cells form the outermost layer of the retina and provide a potential target for viral invasion and permanent vision impairment. Here we examine the initial cellular and molecular mechanisms that facilitate HSV-1 invasion of human RPE cells. High-resolution confocal microscopy demonstrated initial interaction of green fluorescent protein (GFP)-tagged virions with filopodia-like structures present on cell surfaces. Unidirectional movement of the virions on filopodia to the cell body was detected by live cell imaging of RPE cells, which demonstrated susceptibility to pH-dependent HSV-1 entry and replication. Use of RT-PCR indicated expression of nectin-1, herpes virus entry mediator (HVEM) and 3-O-sulfotransferase-3 (as a surrogate marker for 3-O-sulfated heparan sulfate). HVEM and nectin-1 expression was subsequently verified by flow cytometry. Nectin-1 expression in murine retinal tissue was also demonstrated by immunohistochemistry. Antibodies against nectin-1, but not HVEM, were able to block HSV-1 infection. Similar blocking effects were seen with a small interfering RNA construct specifically directed against nectin-1, which also blocked RPE cell fusion with HSV-1 glycoprotein-expressing Chinese hamster ovary (CHO-K1) cells. Anti-nectin-1 antibodies and F-actin depolymerizers were also successful in blocking the cytoskeletal changes that occur upon HSV-1 entry into cells. Our findings shed new light on the cellular and molecular mechanisms that help the virus to enter the cells of the inner eye.


Assuntos
Moléculas de Adesão Celular/fisiologia , Herpesvirus Humano 1/patogenicidade , Epitélio Pigmentado da Retina/virologia , Internalização do Vírus , Replicação Viral , Animais , Células CHO , Fusão Celular , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Camundongos , Nectinas , Pseudópodes/virologia , Membro 14 de Receptores do Fator de Necrose Tumoral/fisiologia , Epitélio Pigmentado da Retina/química , Epitélio Pigmentado da Retina/citologia , Vírion/metabolismo
15.
Invest Ophthalmol Vis Sci ; 49(9): 4026-35, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18502984

RESUMO

PURPOSE: The human conjunctiva is a natural target for herpes simplex virus (HSV)-1 infection. The goals of this study were to investigate the cellular and molecular mechanisms of HSV-1 entry into human conjunctival epithelial (HCjE) cells. Specific features of entry studied included the method of initial viral binding to cells, pH dependency, and expression and usage of specific HSV-1 entry receptors. METHODS: To observe HSV-1 initial binding, live cell imaging was performed on HSV-1-infected HCjE cells. Reporter HSV-1 virions expressing beta-galactosidase were used to determine entry of wild-type HSV-1(KOS) and a mutant, HSV-1(KOS)Rid1, into HCjE cells. HSV-1 replication in HCjE cells was determined by plaque assays. Lysosomotropic agents were used to determine whether viral entry was pH dependent. Reverse transcription (RT)-PCR, flow cytometry, and immunohistochemistry were used to determine the expression of receptors. Receptor-specific siRNAs were used to define the role of individual entry receptors. RESULTS: HSV-1 virions attach to filopodia present on HCjE cells and use them to reach the cell body for entry. Cultured HCjE cells demonstrate susceptibility to HSV-1 entry and form plaques confirming viral replication. Blocking vesicular acidification significantly reduces entry, implicating a pH-dependent mode of entry. Multiple assays confirm the expression of entry receptors nectin-1, HVEM, and 3-O-sulfated heparan sulfate (3-OS HS) on the HCjE cell membrane. Knocking down of gD receptors by siRNAs interference implicates nectin-1 and HVEM as the major mediators of entry. CONCLUSIONS: HSV-1 entry into HCjE cells is a pH-dependent process that is aided by targeted virus travel on filopodia. HCjE cells express all three major entry receptors, with nectin-1 and HVEM playing the predominant role in mediating entry.


Assuntos
Túnica Conjuntiva/virologia , Células Epiteliais/virologia , Herpes Simples/prevenção & controle , Herpesvirus Humano 1/patogenicidade , Proteínas de Filamentos Intermediários/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Membro 14 de Receptores do Fator de Necrose Tumoral/fisiologia , Internalização do Vírus , Animais , Células CHO , Túnica Conjuntiva/fisiologia , Cricetinae , Cricetulus , Células Epiteliais/fisiologia , Herpesvirus Humano 1/fisiologia , Humanos , Nestina , Pseudópodes/virologia , Ensaio de Placa Viral
16.
J Virol ; 82(6): 2821-35, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18184710

RESUMO

Varicella-zoster virus (VZV) is renowned for its very low titer when grown in cultured cells. There remains no single explanation for the low infectivity. In this study, viral particles on the surfaces of infected cells were examined by several imaging technologies. Few surface particles were detected at 48 h postinfection (hpi), but numerous particles were observed at 72 and 96 hpi. At 72 hpi, 75% of the particles resembled light (L) particles, i.e., envelopes without capsids. By 96 hpi, 85% of all particles resembled L particles. Subsequently, the envelopes of complete virions and L particles were investigated to determine their glycoprotein constituents. Glycoproteins gE, gI, and gB were detected in the envelopes of both types of particles in similar numbers; i.e., there appeared to be no difference in the glycoprotein content of the L particles. The viral particles emerged onto the cell surface amid actin-based filopodia, which were present in abundance within viral highways. Viral particles were easily detected at the base of and along the exterior surfaces of the filopodia. VZV particles were not detected within filopodia. In short, these results demonstrate that VZV infection of cultured cells produces a larger proportion of aberrant coreless particles than has been seen with any other previously examined alphaherpesvirus. Further, these results suggested a major disassociation between capsid formation and envelopment as an explanation for the invariably low VZV titer in cultured cells.


Assuntos
Herpesvirus Humano 3/fisiologia , Luz , Pseudópodes/virologia , Linhagem Celular , Glicoproteínas/fisiologia , Herpesvirus Humano 3/crescimento & desenvolvimento , Humanos , Microscopia Eletrônica , Proteínas do Envelope Viral/fisiologia
18.
Nat Cell Biol ; 9(3): 310-5, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17293854

RESUMO

The spread of retroviruses between cells is estimated to be 2-3 orders of magnitude more efficient when cells can physically interact with each other. The underlying mechanism is largely unknown, but transfer is believed to occur through large-surface interfaces, called virological or infectious synapses. Here, we report the direct visualization of cell-to-cell transmission of retroviruses in living cells. Our results reveal a mechanism of virus transport from infected to non-infected cells, involving thin filopodial bridges. These filopodia originate from non-infected cells and interact, through their tips, with infected cells. A strong association of the viral envelope glycoprotein (Env) in an infected cell with the receptor molecules in a target cell generates a stable bridge. Viruses then move along the outer surface of the filopodial bridge toward the target cell. Our data suggest that retroviruses spread by exploiting an inherent ability of filopodia to transport ligands from cell to cell.


Assuntos
Comunicação Celular/fisiologia , Células Eucarióticas/virologia , Pseudópodes/virologia , Retroviridae/fisiologia , Animais , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Antígenos CD4/genética , Antígenos CD4/metabolismo , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Endocitose/fisiologia , Células Eucarióticas/metabolismo , HIV-1/fisiologia , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Mutação , Pseudópodes/ultraestrutura , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
19.
Cell Microbiol ; 9(4): 939-51, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17140405

RESUMO

Viruses exploit the cytoskeleton of host cells to transport their components and spread to neighbouring cells. Here we show that the actin cytoskeleton is involved in the release of Marburgvirus (MARV) particles. We found that peripherally located nucleocapsids and envelope precursors of MARV are located either at the tip or at the side of filopodial actin bundles. Importantly, viral budding was almost exclusively detected at filopodia. Inhibiting actin polymerization in MARV-infected cells significantly diminished the amount of viral particles released into the medium. This suggested that dynamic polymerization of actin in filopodia is essential for efficient release of MARV. The viral matrix protein VP40 plays a key role in the release of MARV particles and we found that the intracellular localization of recombinant VP40 and its release in form of virus-like particles were strongly influenced by overexpression or inhibition of myosin 10 and Cdc42, proteins important in filopodia formation and function. We suggest that VP40, which is capable of interacting with viral nucleocapsids, provides an interface of MARV subviral particles and filopodia. As filopodia are in close contact with neighbouring cells, usurpation of these structures may facilitate spread of MARV to adjacent cells.


Assuntos
Marburgvirus/metabolismo , Pseudópodes/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Chlorocebus aethiops , Eletroforese em Gel de Poliacrilamida , Humanos , Imunoprecipitação , Marburgvirus/crescimento & desenvolvimento , Marburgvirus/ultraestrutura , Microscopia Eletrônica de Varredura , Microscopia Imunoeletrônica , Nucleocapsídeo/metabolismo , Nucleocapsídeo/ultraestrutura , Pseudópodes/ultraestrutura , Pseudópodes/virologia , Células Vero , Proteínas da Matriz Viral/metabolismo , Vírion/metabolismo , Vírion/ultraestrutura
20.
J Cell Biol ; 170(2): 317-25, 2005 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-16027225

RESUMO

Viruses have often been observed in association with the dense microvilli of polarized epithelia as well as the filopodia of nonpolarized cells, yet whether interactions with these structures contribute to infection has remained unknown. Here we show that virus binding to filopodia induces a rapid and highly ordered lateral movement, "surfing" toward the cell body before cell entry. Virus cell surfing along filopodia is mediated by the underlying actin cytoskeleton and depends on functional myosin II. Any disruption of virus cell surfing significantly reduces viral infection. Our results reveal another example of viruses hijacking host machineries for efficient infection by using the inherent ability of filopodia to transport ligands to the cell body.


Assuntos
Actinas/fisiologia , Vírus da Leucose Aviária/fisiologia , Vírus da Leucemia Murina/fisiologia , Miosinas/fisiologia , Pseudópodes/fisiologia , Animais , Vírus da Leucose Aviária/efeitos dos fármacos , Vírus da Leucose Aviária/ultraestrutura , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Membrana Celular/virologia , Citocalasina D/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Vírus da Leucemia Murina/efeitos dos fármacos , Vírus da Leucemia Murina/ultraestrutura , Camundongos , Microscopia Eletrônica , Pseudópodes/ultraestrutura , Pseudópodes/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...