Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 12(5)2020 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-32357558

RESUMO

Virus infection has drawn extensive attention since it causes serious or even deadly diseases, consequently inducing a series of social and public health problems. Caveolin-1 is the most important structural protein of caveolae, a membrane invagination widely known for its role in endocytosis and subsequent cytoplasmic transportation. Caveolae/caveolin-1 is tightly associated with a wide range of biological processes, including cholesterol homeostasis, cell mechano-sensing, tumorigenesis, and signal transduction. Intriguingly, the versatile roles of caveolae/caveolin-1 in virus infections have increasingly been appreciated. Over the past few decades, more and more viruses have been identified to invade host cells via caveolae-mediated endocytosis, although other known pathways have been explored. The subsequent post-entry events, including trafficking, replication, assembly, and egress of a large number of viruses, are caveolae/caveolin-1-dependent. Deprivation of caveolae/caveolin-1 by drug application or gene editing leads to abnormalities in viral uptake, viral protein expression, or virion release, whereas the underlying mechanisms remain elusive and must be explored holistically to provide potential novel antiviral targets and strategies. This review recapitulates our current knowledge on how caveolae/caveolin-1 functions in every step of the viral infection cycle and various relevant signaling pathways, hoping to provide a new perspective for future viral cell biology research.


Assuntos
Cavéolas/virologia , Caveolina 1/metabolismo , Viroses/metabolismo , Fenômenos Fisiológicos Virais , Animais , Cavéolas/metabolismo , Caveolina 1/genética , Endocitose , Humanos , Viroses/genética , Viroses/fisiopatologia , Viroses/virologia , Vírus/genética
2.
FEBS J ; 287(17): 3664-3671, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32428379

RESUMO

The quest for the effective treatment against coronavirus disease 2019 pneumonia caused by the severe acute respiratory syndrome (SARS)-coronavirus 2(CoV-2) coronavirus is hampered by the lack of knowledge concerning the basic cell biology of the infection. Given that most viruses use endocytosis to enter the host cell, mechanistic investigation of SARS-CoV-2 infection needs to consider the diversity of endocytic pathways available for SARS-CoV-2 entry in the human lung epithelium. Taking advantage of the well-established methodology of membrane trafficking studies, this research direction allows for the rapid characterisation of the key cell biological mechanism(s) responsible for SARS-CoV-2 infection. Furthermore, 11 clinically approved generic drugs are identified as potential candidates for repurposing as blockers of several potential routes for SARS-CoV-2 endocytosis. More broadly, the paradigm of targeting a fundamental aspect of human cell biology to protect against infection may be advantageous in the context of future pandemic outbreaks.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , Reposicionamento de Medicamentos , Endocitose/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/virologia , Amilorida/farmacologia , COVID-19/metabolismo , COVID-19/patologia , COVID-19/virologia , Cavéolas/efeitos dos fármacos , Cavéolas/virologia , Clorpromazina/farmacologia , Vesículas Revestidas por Clatrina/efeitos dos fármacos , Vesículas Revestidas por Clatrina/virologia , Endossomos/efeitos dos fármacos , Endossomos/virologia , Humanos , Itraconazol/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Lisossomos/efeitos dos fármacos , Lisossomos/virologia , Nistatina/farmacologia , Pinocitose/efeitos dos fármacos , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Vimblastina/farmacologia
3.
mBio ; 11(1)2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-32047126

RESUMO

Venezuelan and western equine encephalitis viruses (VEEV and WEEV, respectively) invade the central nervous system (CNS) early during infection, via neuronal and hematogenous routes. While viral replication mediates host shutoff, including expression of type I interferons (IFN), few studies have addressed how alphaviruses gain access to the CNS during established infection or the mechanisms of viral crossing at the blood-brain barrier (BBB). Here, we show that hematogenous dissemination of VEEV and WEEV into the CNS occurs via caveolin-1 (Cav-1)-mediated transcytosis (Cav-MT) across an intact BBB, which is impeded by IFN and inhibitors of RhoA GTPase. Use of reporter and nonreplicative strains also demonstrates that IFN signaling mediates viral restriction within cells comprising the neurovascular unit (NVU), differentially rendering brain endothelial cells, pericytes, and astrocytes permissive to viral replication. Transmission and immunoelectron microscopy revealed early events in virus internalization and Cav-1 association within brain endothelial cells. Cav-1-deficient mice exhibit diminished CNS VEEV and WEEV titers during early infection, whereas viral burdens in peripheral tissues remained unchanged. Our findings show that alphaviruses exploit Cav-MT to enter the CNS and that IFN differentially restricts this process at the BBB.IMPORTANCE VEEV, WEEV, and eastern equine encephalitis virus (EEEV) are emerging infectious diseases in the Americas, and they have caused several major outbreaks in the human and horse population during the past few decades. Shortly after infection, these viruses can infect the CNS, resulting in severe long-term neurological deficits or death. Neuroinvasion has been associated with virus entry into the CNS directly from the bloodstream; however, the underlying molecular mechanisms have remained largely unknown. Here, we demonstrate that following peripheral infection alphavirus augments vesicular formation/trafficking at the BBB and utilizes Cav-MT to cross an intact BBB, a process regulated by activators of Rho GTPases within brain endothelium. In vivo examination of early viral entry in Cav-1-deficient mice revealed significantly lower viral burdens in the brain than in similarly infected wild-type animals. These studies identify a potentially targetable pathway to limit neuroinvasion by alphaviruses.


Assuntos
Barreira Hematoencefálica/virologia , Cavéolas/virologia , Vírus da Encefalite Equina Venezuelana/fisiologia , Vírus da Encefalite Equina do Oeste/fisiologia , Transcitose , Internalização do Vírus , Animais , Caveolina 1/genética , Linhagem Celular , Sistema Nervoso Central/virologia , Células Endoteliais/virologia , Masculino , Camundongos Endogâmicos C57BL , Replicação Viral
4.
Virol J ; 16(1): 37, 2019 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-30909932

RESUMO

BACKGROUND: Porcine sapelovirus (PSV), a species of the genus Sapelovirus within the family Picornaviridae, are a significant cause of enteritis, pneumonia, polioencephalomyelitis and reproductive disorders in pigs. However, the life cycle of PSV on the molecular level is largely unknown. METHODS: Here, we used chemical inhibitors, RNA interference, and overexpression of dominant negative (DN) mutant plasmids to verify the roles of distinct endocytic pathways involved in PSV entry into porcine small intestinal epithelial cell line (IPEC-J2). RESULTS: Our experiments indicated that PSV infection was inhibited when cells were pre-treated with NH4Cl or chloroquine. Inhibitors nystatin, methyl-ß-cyclodextrin, dynasore and wortmannin dramatically reduced PSV entry efficiency, whereas the inhibitors chlorpromazine and EIPA had no effect. Furthermore, overexpression caveolin DN mutant and siRNA against caveolin also decreased virus titers and VP1 protein synthesis, whereas overexpression EPS15 DN mutant and siRNA against EPS15 did not reduce virus infection. CONCLUSIONS: Our findings suggest that PSV entry into IPEC-J2 cells depends on caveolae/lipid raft mediated-endocytosis, that is pH-dependent and requires dynamin and PI3K but is independent of clathrin and macropinocytosis.


Assuntos
Cavéolas/virologia , Endocitose , Células Epiteliais/virologia , Picornaviridae/fisiologia , Internalização do Vírus/efeitos dos fármacos , Cloreto de Amônio/farmacologia , Animais , Linhagem Celular , Cloroquina/farmacologia , Clatrina/metabolismo , Dinaminas/metabolismo , Hidrazonas/farmacologia , Nistatina/farmacologia , Picornaviridae/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno , Suínos
5.
J Virol ; 92(15)2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29769350

RESUMO

The members of Flaviviridae utilize several endocytic pathways to enter a variety of host cells. Our previous work showed that classical swine fever virus (CSFV) enters porcine kidney (PK-15) cells through a clathrin-dependent pathway that requires Rab5 and Rab7. The entry mechanism for CSFV into other cell lines remains unclear, for instance, porcine alveolar macrophages (3D4/21 cells). More importantly, the trafficking of CSFV within endosomes controlled by Rab GTPases is unknown in 3D4/21 cells. In this study, entry and postinternalization of CSFV were analyzed using chemical inhibitors, RNA interference, and dominant-negative (DN) mutants. Our data demonstrated that CSFV entry into 3D4/21 cells depends on caveolae, dynamin, and cholesterol but not clathrin or macropinocytosis. The effects of DN mutants and knockdown of four Rab proteins that regulate endosomal trafficking were examined on CSFV infection, respectively. The results showed that Rab5, Rab7, and Rab11, but not Rab9, regulate CSFV endocytosis. Confocal microscopy showed that virus particles colocalize with Rab5, Rab7, or Rab11 within 30 min after virus entry and further with lysosomes, suggesting that after internalization CSFV moves to early, late, and recycling endosomes and then into lysosomes before the release of the viral genome. Our findings provide insights into the life cycle of pestiviruses in macrophages.IMPORTANCE Classical swine fever, is caused by classical swine fever virus (CSFV). The disease is notifiable to World Organisation for Animal Health (OIE) in most countries and causes significant financial losses to the pig industry globally. Understanding the processes of CSFV endocytosis and postinternalization will advance our knowledge of the disease and provide potential novel drug targets against CSFV. With this objective, we used systematic approaches to dissect these processes in CSFV-infected 3D4/21 cells. The data presented here demonstrate for the first time to our knowledge that CSFV is able to enter cells via caveola-mediated endocytosis that requires Rab5, Rab7 and Rab11, in addition to the previously described classical clathrin-dependent pathway that requires Rab5 and Rab7. The characterization of CSFV entry will further promote our current understanding of Pestivirus cellular entry pathways and provide novel targets for antiviral drug development.


Assuntos
Cavéolas/metabolismo , Vírus da Febre Suína Clássica/metabolismo , Endocitose , Macrófagos Alveolares/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Animais , Cavéolas/virologia , Vírus da Febre Suína Clássica/genética , Macrófagos Alveolares/virologia , Suínos , Proteínas rab de Ligação ao GTP/genética , Proteínas rab5 de Ligação ao GTP/genética , proteínas de unión al GTP Rab7
6.
Vet Res ; 49(1): 16, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29439726

RESUMO

Cell-penetrating peptide (CPP) is a promising cargo for delivering bioactive molecules. In this study, the N terminus of VP1 from chicken anemia virus, designated as CVP1, was found to carry enriched arginine residues with α-helix. By confocal imaging, flow cytometry and MTT assay, we identified CVP1 as a novel, safe and efficient CPP. CVP1-FITC peptide could entry different types of cells tested with dose dependence, but without cytotoxic effects. Compared with TAT-FITC peptide, the CVP1-FITC peptide showed much higher cell-penetrating activity. Moreover, CVP1 could successfully deliver ß-glycosidase, poly (I:C) and plasmid into HCT116 cells. Inhibitors and temperature sensitivity analysis further indicated that the cell-penetrating activity of CVP1 was based on ATP-dependent and caveolae-mediated endocytosis. All these data demonstrate that CVP1 has efficient cell-penetrating activity and great potential for developing a novel delivery vector.


Assuntos
Cavéolas/fisiologia , Peptídeos Penetradores de Células/administração & dosagem , Vírus da Anemia da Galinha/fisiologia , Animais , Cavéolas/virologia , Linhagem Celular , Galinhas , Cães , Sistemas de Liberação de Medicamentos/veterinária , Endocitose/fisiologia , Células HCT116 , Células HEK293 , Humanos , Células Madin Darby de Rim Canino
7.
Vet Res ; 45: 17, 2014 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-24517254

RESUMO

Monocytes infected with feline infectious peritonitis virus, a coronavirus, express viral proteins in their plasma membranes. Upon binding of antibodies, these proteins are quickly internalised through a new clathrin- and caveolae-independent internalisation pathway. By doing so, the infected monocytes can escape antibody-dependent cell lysis. In the present study, we investigated which kinases and cytoskeletal proteins are of importance during internalisation and subsequent intracellular transport. The experiments showed that myosin light chain kinase (MLCK) and myosin 1 are crucial for the initiation of the internalisation. With co-localisation stainings, it was found that MLCK and myosin 1 co-localise with antigens even before internalisation started. Myosin 6 co-localised with the internalising complexes during passage through the cortical actin, were it might play a role in moving or disintegrating actin filaments, to overcome the actin barrier. One minute after internalisation started, vesicles had passed the cortical actin, co-localised with microtubules and association with myosin 6 was lost. The vesicles were further transported over the microtubules and accumulated at the microtubule organising centre after 10 to 30 min. Intracellular trafficking over microtubules was mediated by MLCK, myosin 1 and a small actin tail. Since inhibiting MLCK with ML-7 was so efficient in blocking the internalisation pathway, this target can be used for the development of a new treatment for FIPV.


Assuntos
Actinas/metabolismo , Coronavirus Felino/fisiologia , Peritonite Infecciosa Felina/metabolismo , Microtúbulos/metabolismo , Miosinas/metabolismo , Internalização do Vírus , Actinas/genética , Animais , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Gatos , Cavéolas/fisiologia , Cavéolas/virologia , Clatrina/fisiologia , Peritonite Infecciosa Felina/virologia , Regulação da Expressão Gênica , Microtúbulos/genética , Monócitos/virologia , Miosinas/genética
8.
PLoS Pathog ; 6(9): e1001121, 2010 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-20886108

RESUMO

Ebolavirus (EBOV) is an enveloped, single-stranded, negative-sense RNA virus that causes severe hemorrhagic fever with mortality rates of up to 90% in humans and nonhuman primates. Previous studies suggest roles for clathrin- or caveolae-mediated endocytosis in EBOV entry; however, ebolavirus virions are long, filamentous particles that are larger than the plasma membrane invaginations that characterize clathrin- or caveolae-mediated endocytosis. The mechanism of EBOV entry remains, therefore, poorly understood. To better understand Ebolavirus entry, we carried out internalization studies with fluorescently labeled, biologically contained Ebolavirus and Ebolavirus-like particles (Ebola VLPs), both of which resemble authentic Ebolavirus in their morphology. We examined the mechanism of Ebolavirus internalization by real-time analysis of these fluorescently labeled Ebolavirus particles and found that their internalization was independent of clathrin- or caveolae-mediated endocytosis, but that they co-localized with sorting nexin (SNX) 5, a marker of macropinocytosis-specific endosomes (macropinosomes). Moreover, the internalization of Ebolavirus virions accelerated the uptake of a macropinocytosis-specific cargo, was associated with plasma membrane ruffling, and was dependent on cellular GTPases and kinases involved in macropinocytosis. A pseudotyped vesicular stomatitis virus possessing the Ebolavirus glycoprotein (GP) also co-localized with SNX5 and its internalization and infectivity were affected by macropinocytosis inhibitors. Taken together, our data suggest that Ebolavirus is internalized into cells by stimulating macropinocytosis in a GP-dependent manner. These findings provide new insights into the lifecycle of Ebolavirus and may aid in the development of therapeutics for Ebolavirus infection.


Assuntos
Ebolavirus/fisiologia , Doença pelo Vírus Ebola/virologia , Pinocitose/fisiologia , Nexinas de Classificação/metabolismo , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Western Blotting , Cavéolas/metabolismo , Cavéolas/virologia , Células Cultivadas , Chlorocebus aethiops , Clatrina/metabolismo , Endocitose/fisiologia , Doença pelo Vírus Ebola/metabolismo , Humanos , Microscopia de Fluorescência , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Nexinas de Classificação/genética , Células Vero , Vesiculovirus , Proteínas do Envelope Viral/genética , Vírion/genética , Replicação Viral , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
9.
PLoS Pathog ; 6(9): e1001110, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20862315

RESUMO

Zaire ebolavirus (ZEBOV), a highly pathogenic zoonotic virus, poses serious public health, ecological and potential bioterrorism threats. Currently no specific therapy or vaccine is available. Virus entry is an attractive target for therapeutic intervention. However, current knowledge of the ZEBOV entry mechanism is limited. While it is known that ZEBOV enters cells through endocytosis, which of the cellular endocytic mechanisms used remains unclear. Previous studies have produced differing outcomes, indicating potential involvement of multiple routes but many of these studies were performed using noninfectious surrogate systems such as pseudotyped retroviral particles, which may not accurately recapitulate the entry characteristics of the morphologically distinct wild type virus. Here we used replication-competent infectious ZEBOV as well as morphologically similar virus-like particles in specific infection and entry assays to demonstrate that in HEK293T and Vero cells internalization of ZEBOV is independent of clathrin, caveolae, and dynamin. Instead the uptake mechanism has features of macropinocytosis. The binding of virus to cells appears to directly stimulate fluid phase uptake as well as localized actin polymerization. Inhibition of key regulators of macropinocytosis including Pak1 and CtBP/BARS as well as treatment with the drug EIPA, which affects macropinosome formation, resulted in significant reduction in ZEBOV entry and infection. It is also shown that following internalization, the virus enters the endolysosomal pathway and is trafficked through early and late endosomes, but the exact site of membrane fusion and nucleocapsid penetration in the cytoplasm remains unclear. This study identifies the route for ZEBOV entry and identifies the key cellular factors required for the uptake of this filamentous virus. The findings greatly expand our understanding of the ZEBOV entry mechanism that can be applied to development of new therapeutics as well as provide potential insight into the trafficking and entry mechanism of other filoviruses.


Assuntos
Ebolavirus/fisiologia , Endossomos/metabolismo , Endossomos/virologia , Doença pelo Vírus Ebola/prevenção & controle , Pinocitose/fisiologia , Internalização do Vírus/efeitos dos fármacos , Oxirredutases do Álcool/genética , Oxirredutases do Álcool/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Western Blotting , Cavéolas/metabolismo , Cavéolas/virologia , Células Cultivadas , Chlorocebus aethiops , Clatrina/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endocitose/fisiologia , Doença pelo Vírus Ebola/metabolismo , Doença pelo Vírus Ebola/virologia , Humanos , Rim/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Fusão de Membrana , Microdomínios da Membrana/fisiologia , Fosforilação , Pinocitose/efeitos dos fármacos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células Vero , Replicação Viral/efeitos dos fármacos , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
10.
Virology ; 384(2): 389-99, 2009 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-19157478

RESUMO

This review summarizes the field's major findings related to the characterization of polyomavirus structures and to the characterization of virus receptors and mechanisms of host cell invasion. The four members of the family that have received the most attention in this regard are the mouse polyomavirus (mPyV), the monkey polyomavirus SV40, and the two human polyomaviruses, JCV and BKV. The structures of both the mPyV and SV40 alone and in complex with receptor fragments have been solved to high resolution. The majority of polyomaviruses recognize terminal sialic acid in either an alpha2,3 linkage or an alpha2,6 linkage to the underlying galactose. Studies on virus structure, receptor utilization and mechanisms of entry have led to new insights into how these viruses interact in an active way with cells to ensure the nuclear delivery and expression of their genomes. Critical work on virus entry has led to the discovery of a pH neutral endocytic compartment that accepts cargo from caveolae and to novel roles for endoplasmic reticulum (ER) associated factors in virus uncoating and penetration of ER membranes. This review will summarize the major findings and compare and contrast the mechanisms used by these viruses to infect cells.


Assuntos
Infecções por Polyomavirus/virologia , Polyomavirus/fisiologia , Receptores Virais/metabolismo , Cavéolas/virologia , Retículo Endoplasmático/virologia , Interações Hospedeiro-Patógeno , Polyomavirus/genética , Polyomavirus/metabolismo , Ácidos Siálicos/metabolismo
11.
J Gen Virol ; 89(Pt 11): 2731-2740, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18931069

RESUMO

Infection with feline infectious peritonitis virus (FIPV), a feline coronavirus, frequently leads to death in spite of a strong humoral immune response. In previous work, we reported that infected monocytes, the in vivo target cells of FIPV, express viral proteins in their plasma membranes. These proteins are quickly internalized upon binding of antibodies. As the cell surface is cleared from viral proteins, internalization might offer protection against antibody-dependent cell lysis. Here, the internalization and subsequent trafficking of the antigen-antibody complexes were characterized using biochemical, cell biological and genetic approaches. Internalization occurred through a clathrin- and caveolae-independent pathway that did not require dynamin, rafts, actin or rho-GTPases. These findings indicate that the viral antigen-antibody complexes were not internalized through any of the previously described pathways. Further characterization showed that this internalization process was independent from phosphatases and tyrosine kinases but did depend on serine/threonine kinases. After internalization, the viral antigen-antibody complexes passed through the early endosomes, where they resided only briefly, and accumulated in the late endosomes. Between 30 and 60 min after antibody addition, the complexes left the late endosomes but were not degraded in the lysosomes. This study reveals what is probably a new internalization pathway into primary monocytes, confirming once more the complexity of endocytic processes.


Assuntos
Cavéolas/fisiologia , Clatrina/fisiologia , Coronavirus Felino/fisiologia , Peritonite Infecciosa Felina/fisiopatologia , Proteínas Virais/biossíntese , Internalização do Vírus , Animais , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Gatos , Cavéolas/virologia , Coronavirus Felino/imunologia , Endossomos/imunologia , Endossomos/virologia , Peritonite Infecciosa Felina/imunologia , Genes Reporter , Modelos Biológicos , Monócitos/virologia , Plasmídeos
12.
PLoS One ; 3(10): e3313, 2008 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-18836553

RESUMO

BACKGROUND: Infectious entry of human papillomaviruses into their host cells is an important step in the viral life cycle. For cell binding these viruses use proteoglycans as initial attachment sites. Subsequent transfer to a secondary receptor molecule seems to be involved in virus uptake. Depending on the papillomavirus subtype, it has been reported that entry occurs by clathrin- or caveolin-mediated mechanisms. Regarding human papillomavirus type 16 (HPV16), the primary etiologic agent for development of cervical cancer, clathrin-mediated endocytosis was described as infectious entry pathway. METHODOLOGY/PRINCIPAL FINDINGS: Using immunofluorescence and infection studies we show in contrast to published data that infectious entry of HPV16 occurs in a clathrin- and caveolin-independent manner. Inhibition of clathrin- and caveolin/raft-dependent endocytic pathways by dominant-negative mutants and siRNA-mediated knockdown, as well as inhibition of dynamin function, did not impair infection. Rather, we provide evidence for involvement of tetraspanin-enriched microdomains (TEMs) in HPV16 endocytosis. Following cell attachment, HPV16 particles colocalized with the tetraspanins CD63 and CD151 on the cell surface. Notably, tetraspanin-specific antibodies and siRNA inhibited HPV16 cell entry and infection, confirming the importance of TEMs for infectious endocytosis of HPV16. CONCLUSIONS/SIGNIFICANCE: Tetraspanins fulfill various roles in the life cycle of a number of important viral pathogens, including human immunodeficiency virus (HIV) and hepatitis C virus (HCV). However, their involvement in endocytosis of viral particles has not been proven. Our data indicate TEMs as a novel clathrin- and caveolin-independent invasion route for viral pathogens and especially HPV16.


Assuntos
Papillomavirus Humano 16/metabolismo , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Internalização do Vírus , Antígenos CD/metabolismo , Antígenos CD/ultraestrutura , Cavéolas/metabolismo , Cavéolas/virologia , Linhagem Celular , Clatrina/genética , Clatrina/metabolismo , Endocitose , Feminino , Células HeLa , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/patogenicidade , Papillomavirus Humano 16/ultraestrutura , Humanos , Rim/citologia , Microdomínios da Membrana/ultraestrutura , Proteínas de Membrana/ultraestrutura , Glicoproteínas da Membrana de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/ultraestrutura , Tetraspanina 24 , Tetraspanina 30 , Vírion/genética , Vírion/metabolismo , Vírion/ultraestrutura
13.
J Gen Virol ; 89(Pt 9): 2147-2156, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18753224

RESUMO

Feline infectious peritonitis virus (FIPV), a coronavirus that causes a lethal chronic disease in cats, enters feline monocytes via endocytosis. In this study, the pathway of internalization is characterized by evaluating the effect of chemical inhibitors and/or expression of dominant-negative (DN) proteins on the percentage of internalized virions per cell and infection. Further, co-localization studies were performed to determine the involvement of certain cellular internalization proteins. FIPV is not internalized through a clathrin-mediated pathway, as chlorpromazine, amantadine and DN eps15 did not influence virus uptake and FIPV did not co-localize with clathrin. The caveolae-mediated pathway could be excluded based on the inability of genistein and DN caveolin-1 to inhibit virus uptake and lack of co-localization between FIPV and caveolin-1. Dynamin inhibitory peptide and DN dynamin effectively inhibited virus internalization. The inhibitor strongly reduced uptake to 20.3+/-1.1% of uptake in untreated cells. In the presence of DN dynamin, uptake was 58.7+/-3.9% relative to uptake in untransduced cells. Internalization of FIPV was slightly reduced to 85.0+/-1.4 and 87.4+/-6.1% of internalization in control cells by the sterol-binding drugs nystatin and methyl-beta-cyclodextrin, respectively. Rho GTPases were inhibited by Clostridium difficile toxin B, but no effect was observed. These results were confirmed with infection studies showing that infection was not influenced by chlorpromazine, amantadine and genistein, but was significantly reduced by dynamin inhibition and nystatin. In conclusion, these results indicate that FIPV enters monocytes through a clathrin- and caveolae-independent pathway that strongly depends on dynamin and is slightly sensitive to cholesterol depletion.


Assuntos
Clatrina/fisiologia , Coronavirus Felino/fisiologia , Coronavirus Felino/patogenicidade , Dinaminas/fisiologia , Monócitos/fisiologia , Monócitos/virologia , Amantadina/farmacologia , Animais , Gatos , Cavéolas/efeitos dos fármacos , Cavéolas/virologia , Caveolina 1/antagonistas & inibidores , Caveolina 1/genética , Caveolina 1/fisiologia , Clorpromazina/farmacologia , Coronavirus Felino/efeitos dos fármacos , Dinaminas/antagonistas & inibidores , Dinaminas/genética , Endocitose/efeitos dos fármacos , Genisteína/farmacologia , Técnicas In Vitro , Monócitos/efeitos dos fármacos , Nistatina/farmacologia , Internalização do Vírus/efeitos dos fármacos , beta-Ciclodextrinas/farmacologia , Proteínas rho de Ligação ao GTP/fisiologia
14.
J Virol ; 82(18): 9075-85, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18614639

RESUMO

Foot-and-mouth disease virus (FMDV) utilizes different cell surface macromolecules to facilitate infection of cultured cells. Virus, which is virulent for susceptible animals, infects cells via four members of the alpha(V) subclass of cellular integrins. In contrast, tissue culture adaptation of some FMDV serotypes results in the loss of viral virulence in the animal, accompanied by the loss of virus' ability to use integrins as receptors. These avirulent viral variants acquire positively charged amino acids on surface-exposed structural proteins, resulting in the utilization of cell surface heparan sulfate (HS) molecules as receptors. We have recently shown that FMDV serotypes utilizing integrin receptors enter cells via a clathrin-mediated mechanism into early endosomes. Acidification within the endosome results in a breakdown of the viral capsid, releasing the RNA, which enters the cytoplasm by a still undefined mechanism. Since there is evidence that HS internalizes bound ligands via a caveola-mediated mechanism, it was of interest to analyze the entry of FMDV by cell-surface HS. Using a genetically engineered variant of type O(1)Campos (O(1)C3056R) which can utilize both integrins and HS as receptors and a second variant (O(1)C3056R-KGE) which can utilize only HS as a receptor, we followed viral entry using confocal microscopy. After virus bound to cells at 4 degrees C, followed by a temperature shift to 37 degrees C, type O(1)C3056R-KGE colocalized with caveolin-1, while O(1)C3056R colocalized with both clathrin and caveolin-1. Compounds which either disrupt or inhibit the formation of lipid rafts inhibited the replication of O(1)C3056R-KGE. Furthermore, a caveolin-1 knockdown by RNA interference also considerably reduced the efficiency of O(1)C3056R-KGE infection. These results indicate that HS-binding FMDV enters the cells via the caveola-mediated endocytosis pathway and that caveolae can associate and traffic with endosomes. In addition, these results further suggest that the route of FMDV entry into cells is a function solely of the viral receptor.


Assuntos
Cavéolas/virologia , Endocitose/fisiologia , Células Epiteliais/virologia , Vírus da Febre Aftosa/patogenicidade , Heparitina Sulfato/metabolismo , Receptores Virais/metabolismo , Animais , Células COS/virologia , Cavéolas/fisiologia , Linhagem Celular , Chlorocebus aethiops , Vírus da Febre Aftosa/metabolismo , Humanos , Glândulas Mamárias Humanas/citologia
15.
Trends Microbiol ; 16(2): 44-7, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18191571

RESUMO

To initiate infection, poliovirus must release its RNA genome into the cytoplasm of a target cell, a process called 'uncoating'. How this occurs has remained uncertain, despite studies over several decades. Two new studies re-address the question of poliovirus entry. The results suggest that poliovirus enters different cells by different mechanisms, and point to a role for virus-induced intracellular signals in the process.


Assuntos
Corantes Fluorescentes/metabolismo , Poliovirus/fisiologia , Internalização do Vírus , Cavéolas/virologia , Vesículas Revestidas por Clatrina/virologia , Citoplasma/virologia , Endocitose , Células Endoteliais/virologia , Células HeLa , Humanos , Proteínas de Membrana/metabolismo , Microscopia de Fluorescência , Poliovirus/ultraestrutura , RNA Viral/metabolismo , Receptores Virais/metabolismo , Coloração e Rotulagem , Vírion/metabolismo
16.
Cell Res ; 18(2): 290-301, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18227861

RESUMO

While severe acute respiratory syndrome coronavirus (SARS-CoV) was initially thought to enter cells through direct fusion with the plasma membrane, more recent evidence suggests that virus entry may also involve endocytosis. We have found that SARS-CoV enters cells via pH- and receptor-dependent endocytosis. Treatment of cells with either SARS-CoV spike protein or spike-bearing pseudoviruses resulted in the translocation of angiotensin-converting enzyme 2 (ACE2), the functional receptor of SARS-CoV, from the cell surface to endosomes. In addition, the spike-bearing pseudoviruses and early endosome antigen 1 were found to colocalize in endosomes. Further analyses using specific endocytic pathway inhibitors and dominant-negative Eps15 as well as caveolin-1 colocalization study suggested that virus entry was mediated by a clathrin- and caveolae-independent mechanism. Moreover, cholesterol- and sphingolipid-rich lipid raft microdomains in the plasma membrane, which have been shown to act as platforms for many physiological signaling pathways, were shown to be involved in virus entry. Endocytic entry of SARS-CoV may expand the cellular range of SARS-CoV infection, and our findings here contribute to the understanding of SARS-CoV pathogenesis, providing new information for anti-viral drug research.


Assuntos
Endocitose , Endossomos/metabolismo , Glicoproteínas de Membrana/metabolismo , Peptidil Dipeptidase A/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Proteínas Adaptadoras de Transdução de Sinal , Enzima de Conversão de Angiotensina 2 , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Cavéolas/metabolismo , Cavéolas/virologia , Caveolina 1/genética , Caveolina 1/metabolismo , Linhagem Celular , Clatrina/genética , Clatrina/metabolismo , Endocitose/genética , Endossomos/virologia , Humanos , Concentração de Íons de Hidrogênio , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Glicoproteínas de Membrana/genética , Peptidil Dipeptidase A/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transporte Proteico , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , Síndrome Respiratória Aguda Grave/tratamento farmacológico , Síndrome Respiratória Aguda Grave/genética , Síndrome Respiratória Aguda Grave/metabolismo , Glicoproteína da Espícula de Coronavírus , Proteínas do Envelope Viral/genética
17.
Virology ; 369(1): 1-11, 2007 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-17698159

RESUMO

Arenaviruses are important causes of viral hemorrhagic fevers in humans. Arenavirus infection of cells occurs via a pH-dependent endocytic route, but detailed studies of entry pathways have not been done. We investigated the role of cell membrane cholesterol, caveolae, and clathrin coated pits in infection by Lassa virus (LASV), which utilizes alpha-dystroglycan (alpha-DG) as a receptor, and Pichindé virus (PICV), which does not. Depletion of cellular cholesterol by treatment with methyl betacyclodextrin (MbetaCD) or nystatin/progesterone inhibited PICV replication and transfer of packaged marker gene by LASV or PICV pseudotyped retroviral particles. In cells lacking caveolae due to silencing of the caveolin-1 gene, no inhibition of PICV infection or LASV pseudotype transduction was observed. However, PICV infection and LASV and PICV pseudotype transduction was inhibited when an Eps15 dominant negative mutant was used to inhibit clathrin-mediated endocytosis. Altogether, the results indicate that diverse arenaviruses have a common requirement for cell membrane cholesterol and clathrin mediated endocytosis in establishing infection.


Assuntos
Vesículas Revestidas por Clatrina/virologia , Endocitose/fisiologia , Vírus Lassa/fisiologia , Lipídeos de Membrana/fisiologia , Vírus Pichinde/fisiologia , Internalização do Vírus , Animais , Cavéolas/virologia , Caveolina 1/antagonistas & inibidores , Linhagem Celular , Chlorocebus aethiops , Colesterol/biossíntese , Inativação Gênica , Humanos , Camundongos , Transdução Genética , Células Vero , Replicação Viral/fisiologia
18.
Virology ; 369(1): 105-18, 2007 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-17714753

RESUMO

We have characterized the entry leading to productive infection of a type C FMDV in two cell lines widely used for virus growth, BHK-21 and IBRS-2. Inhibition of clathrin-mediated endocytosis by sucrose treatment decreased both cell entry and virus multiplication. Evidence of a direct requirement of clathrin for productive viral entry was obtained using BHK21-tTA/anti-CHC cells, which showed a significant reduction of viral entry and infection when the synthesis and functionality of clathrin heavy chain was inhibited (Tet- cells). This was also observed for vesicular stomatitis virus (VSV) productive entry. The effect of NH(4)Cl and concanamycin A on FMDV entry and infection was consistent with the requirement of acidic compartments for decapsidation and virus replication. As expected from its higher stability at acidic pH, this requirement was higher for VSV. Since BHK-21 and IBRS-2 cells expressed caveolin-1, we explored the effect on productive virus entry of drugs that interfere with caveolae-mediated endocytosis. Treatment with nystatin did not reduce entry and infection of FMDV or VSV, while cholesterol depletion with MbetaCD significantly inhibited both steps of the FMDV cycle, indicating that plasma membrane cholesterol is required for virus productive entry.


Assuntos
Vesículas Revestidas por Clatrina/virologia , Clatrina/fisiologia , Endocitose , Vírus da Febre Aftosa/fisiologia , Lipídeos de Membrana/fisiologia , Internalização do Vírus , Cloreto de Amônio/farmacologia , Animais , Antivirais/farmacologia , Cavéolas/virologia , Linhagem Celular , Clatrina/antagonistas & inibidores , Cricetinae , Vírus da Febre Aftosa/crescimento & desenvolvimento , Macrolídeos/farmacologia , Nistatina/farmacologia , Suínos , Vesiculovirus/crescimento & desenvolvimento , Vesiculovirus/fisiologia , Internalização do Vírus/efeitos dos fármacos
19.
J Virol ; 81(16): 8552-62, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17553887

RESUMO

In recent years, BK virus (BKV) nephritis after renal transplantation has become a severe problem. The exact mechanisms of BKV cell entry and subsequent intracellular trafficking remain unknown. Since human renal proximal tubular epithelial cells (HRPTEC) represent a main natural target of BKV nephritis, analysis of BKV infection of HRPTEC is necessary to obtain additional insights into BKV biology and to develop novel strategies for the treatment of BKV nephritis. We coincubated HRPTEC with BKV and the cholesterol-depleting agents methyl beta cyclodextrin (MBCD) and nystatin (Nys), drugs inhibiting caveolar endocytosis. The percentage of infected cells (detected by immunofluorescence) and the cellular levels of BKV large T antigen expression (detected by Western blot analysis) were significantly decreased in both MBCD- and Nys-treated HPRTEC compared to the level in HRPTEC incubated with BKV alone. HRPTEC infection by BKV was also tested after small interfering RNA (siRNA)-dependent depletion of either the caveolar structural protein caveolin-1 (Cav-1) or clathrin, the major structural protein of clathrin-coated pits. BKV infection was inhibited in HRPTEC transfected with Cav-1 siRNA but not in HRPTEC transfected with clathrin siRNA. The colocalization of labeled BKV particles with either Cav-1 or clathrin was investigated by using fluorescent microscopy and image cross-correlation spectroscopy. The rate of colocalization of BKV with Cav-1 peaked at 4 h after incubation. Colocalization with clathrin was insignificant at all time points. These results suggest that BKV entered into HRPTEC via caveolae, not clathrin-coated pits, and that BKV is maximally associated with caveolae at 4 h after infection, prior to relocation to a different intracellular compartment.


Assuntos
Vírus BK/fisiologia , Cavéolas/virologia , Endocitose , Túbulos Renais Proximais/virologia , Nefrite/virologia , Infecções por Polyomavirus/virologia , Internalização do Vírus , Cavéolas/química , Caveolina 1/análise , Caveolina 1/antagonistas & inibidores , Caveolina 1/metabolismo , Células Cultivadas , Clatrina/análise , Clatrina/antagonistas & inibidores , Clatrina/metabolismo , Endocitose/efeitos dos fármacos , Células Epiteliais/ultraestrutura , Células Epiteliais/virologia , Humanos , Túbulos Renais Proximais/fisiopatologia , Nistatina/farmacologia , Infecções por Polyomavirus/fisiopatologia , RNA Interferente Pequeno/farmacologia , beta-Ciclodextrinas/farmacologia
20.
J Virol ; 81(11): 5472-83, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17376898

RESUMO

Rotavirus NSP4, initially characterized as an endoplasmic reticulum intracellular receptor, is a multifunctional viral enterotoxin that induces diarrhea in murine pups. There have been recent reports of the secretion of a cleaved NSP4 fragment (residues 112 to 175) and of the association of NSP4 with LC3-positive autophagosomes, raft membranes, and microtubules. To determine if NSP4 traffics to a specific subset of rafts at the plasma membrane, we isolated caveolae from plasma membrane-enriched material that yielded caveola membranes free of endoplasmic reticulum and nonraft plasma membrane markers. Analyses of the newly isolated caveolae from rotavirus-infected MDCK cells revealed full-length, high-mannose glycosylated NSP4. The lack of Golgi network-specific processing of the caveolar NSP4 glycans supports studies showing that NSP4 bypasses the Golgi apparatus. Confocal imaging showed the colocalization of NSP4 with caveolin-1 early and late in infection, elucidating the temporal and spatial NSP4-caveolin-1 association during infection. These data were extended with fluorescent resonance energy transfer analyses that confirmed the NSP4 and caveolin-1 interaction in that the specific fluorescently tagged antibodies were within 10 nm of each other during infection. Cells transfected with NSP4 showed patterns of staining and colocalization with caveolin-1 similar to those of infected cells. This study presents an endoplasmic reticulum contaminant-free caveola isolation protocol; describes the presence of full-length, endoglycosidase H-sensitive NSP4 in plasma membrane caveolae; provides confirmation of the NSP4-caveolin interaction in the presence and absence of other viral proteins; and provides a final plasma membrane destination for Golgi network-bypassing NSP4 transport.


Assuntos
Cavéolas/metabolismo , Glicoproteínas/metabolismo , Microdomínios da Membrana/metabolismo , Toxinas Biológicas/metabolismo , Proteínas não Estruturais Virais/metabolismo , Animais , Cavéolas/ultraestrutura , Cavéolas/virologia , Linhagem Celular , Cães , Glicoproteínas/isolamento & purificação , Glicosilação , Células HT29 , Humanos , Microdomínios da Membrana/virologia , Octoxinol , Rotavirus/metabolismo , Toxinas Biológicas/isolamento & purificação , Proteínas não Estruturais Virais/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...