Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Virol J ; 3: 21, 2006 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-16579862

RESUMO

BACKGROUND: We have recently shown that amphotropic murine leukemia virus (A-MLV) can enter the mouse fibroblast cell line NIH3T3 via caveola-dependent endocytosis. But due to the size and omega-like shape of caveolae it is possible that A-MLV initially binds cells outside of caveolae. Rafts have been suggested to be pre-caveolae and we here investigate whether A-MLV initially binds to its receptor Pit2, a sodium-dependent phosphate transporter, in rafts or caveolae or outside these cholesterol-rich microdomains. RESULTS: Here, we show that a high amount of cell-bound A-MLV was attached to large rafts of NIH3T3 at the time of investigation. These large rafts were not enriched in caveolin-1, a major structural component of caveolae. In addition, they are rather of natural occurrence in NIH3T3 cells than a result of patching of smaller rafts by A-MLV. Thus cells incubated in parallel with vesicular stomatitis virus glycoprotein (VSV-G) pseudotyped MLV particles showed the same pattern of large rafts as cells incubated with A-MLV, but VSV-G pseudotyped MLV particles did not show any preference to attach to these large microdomains. CONCLUSION: The high concentration of A-MLV particles bound to large rafts of NIH3T3 cells suggests a role of these microdomains in early A-MLV binding events.


Assuntos
Colesterol/metabolismo , Fibroblastos/virologia , Vírus da Leucemia Murina/metabolismo , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/virologia , Animais , Cavéolas/metabolismo , Cavéolas/virologia , Membrana Celular/química , Membrana Celular/virologia , Fibroblastos/metabolismo , Técnica Indireta de Fluorescência para Anticorpo/métodos , Microdomínios da Membrana/química , Camundongos , Células NIH 3T3 , Proteínas Ativadoras de Esfingolipídeos/metabolismo
11.
Uirusu ; 55(1): 19-26, 2005 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-16308526

RESUMO

The endocytic function of caveolae has been controversial for a long time. However, a real-time-imaging analysis of Simian virus 40 (SV40) 's entry in cells has indicated the existence of caveolar endocytosis during virus entry. The caveolae engulfed SV40 virions begin budding from plasma membrane depending on dynamin. SV40 enclosed in caveolae vesicles move to the caveosome, then to the endoplasmic reticulum. In addition, it was demonstrated that human coronavirus-229E enters the cell through caveolae. This review examines the involvement of caveolae in endocytosis used by the viral entry system.


Assuntos
Cavéolas/fisiologia , Cavéolas/virologia , Coronavirus Humano 229E/patogenicidade , Endocitose/fisiologia , Vírus 40 dos Símios/patogenicidade , Caveolina 1/fisiologia , Membrana Celular/virologia , Coronavirus Humano 229E/ultraestrutura , Dinaminas/fisiologia , Retículo Endoplasmático/virologia , Endossomos/fisiologia , Endossomos/virologia , Interpretação de Imagem Assistida por Computador/métodos , Microscopia Eletrônica/métodos , Vírus 40 dos Símios/ultraestrutura , Vírion/crescimento & desenvolvimento
12.
Cell Mol Biol Lett ; 9(1): 47-60, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15048150

RESUMO

Clathrin-mediated endocytosis has been described as the primary internalization pathway for many viruses, including the influenza virus. However, caveolae, an alternative clathrin-independent endocytotic pathway, has also been described as mediating the entry of some molecules, including viruses. To address the question of pathway selection by the influenza virus, we have investigated whether the virus is internalized via clathrin-coated pits and/or caveolae in Madin Darby canine kidney (MDCK) cells. By applying pharmacological manipulations to selectively disrupt the cell internalization pathways, we found that, in MDCK cells, the influenza virus may be internalized via caveolae in addition to entry by clathrin-mediated endocytosis. However, a small contribution by another mode of entry, as recently proposed, cannot be excluded.


Assuntos
Cavéolas/metabolismo , Membrana Celular/metabolismo , Invaginações Revestidas da Membrana Celular/metabolismo , Células Epiteliais/metabolismo , Orthomyxoviridae/metabolismo , Animais , Transporte Biológico/fisiologia , Cavéolas/virologia , Membrana Celular/virologia , Células Cultivadas , Invaginações Revestidas da Membrana Celular/virologia , Vesículas Revestidas/metabolismo , Vesículas Revestidas/virologia , Cães , Endocitose/fisiologia , Células Epiteliais/virologia , Filipina/química
13.
J Virol ; 77(4): 2615-22, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12552000

RESUMO

The murine polyomavirus (Py) enters mouse fibroblasts and kidney epithelial cells via an endocytic pathway that is caveola-independent (as well as clathrin-independent). In contrast, uptake of simian virus 40 into the same cells is dependent on caveola. Following the initial uptake of Py, both microtubules and microfilaments play roles in trafficking of the virus to the nucleus. Colcemid, which disrupts microtubules, inhibits the ability of Py to reach the nucleus and replicate. Paclitaxel, which stabilizes microtubules and prevents microtubule turnover, has no effect, indicating that intact but not dynamic microtubules are required for Py infectivity. Compounds that disrupt actin filaments enhance Py uptake while stabilization of actin filaments impedes Py infection. Virus particles are seen in association with actin in cells treated with microfilament-disrupting or filament-stabilizing agents at levels comparable to those in untreated cells, suggesting that a dynamic state of the microfilament system is important for Py infectivity.


Assuntos
Polyomavirus/fisiologia , Polyomavirus/patogenicidade , Vírus 40 dos Símios/fisiologia , Vírus 40 dos Símios/patogenicidade , Células 3T3/virologia , Actinas/metabolismo , Animais , Cavéolas/efeitos dos fármacos , Cavéolas/virologia , Células Cultivadas , Citoesqueleto/metabolismo , Demecolcina/farmacologia , Células Epiteliais/virologia , Rim/citologia , Rim/virologia , Camundongos , Microtúbulos , Nistatina/farmacologia , Paclitaxel/farmacologia
14.
Science ; 296(5567): 535-9, 2002 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-11964480

RESUMO

Simian virus 40 (SV40) utilizes endocytosis through caveolae for infectious entry into host cells. We found that after binding to caveolae, virus particles induced transient breakdown of actin stress fibers. Actin was then recruited to virus-loaded caveolae as actin patches that served as sites for actin "tail" formation. Dynamin II was also transiently recruited. These events depended on the presence of cholesterol and on the activation of tyrosine kinases that phosphorylated proteins in caveolae. They were necessary for formation of caveolae-derived endocytic vesicles and for infection of the cell. Thus, caveolar endocytosis is ligand-triggered and involves extensive rearrangement of the actin cytoskeleton.


Assuntos
Citoesqueleto de Actina/fisiologia , Actinas/metabolismo , Cavéolas/metabolismo , Depsipeptídeos , Endocitose , GTP Fosfo-Hidrolases/metabolismo , Vírus 40 dos Símios/fisiologia , Citoesqueleto de Actina/ultraestrutura , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Cavéolas/ultraestrutura , Cavéolas/virologia , Caveolina 1 , Caveolinas/metabolismo , Linhagem Celular , Colesterol/fisiologia , Dinaminas , GTP Fosfo-Hidrolases/genética , Haplorrinos , Peptídeos Cíclicos/farmacologia , Fosforilação , Fosfotirosina/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Fibras de Estresse/metabolismo , Tiazóis/farmacologia , Tiazolidinas , Vesículas Transportadoras/metabolismo
15.
J Virol ; 76(10): 5156-66, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11967331

RESUMO

Simian virus 40 (SV40) enters cells by atypical endocytosis mediated by caveolae that transports the virus to the endoplasmic reticulum (ER) instead of to the endosomal-lysosomal compartment, which is the usual destination for viruses and other cargo that enter by endocytosis. We show here that SV4O is transported to the ER via an intermediate compartment that contains beta-COP, which is best known as a component of the COPI coatamer complexes that are required for the retrograde retrieval pathway from the Golgi to the ER. Additionally, transport of SV40 to the ER, as well as infection, is sensitive to brefeldin A. This drug acts by specifically inhibiting the ARF1 GTPase, which is known to regulate assembly of COPI coat complexes on Golgi cisternae. Moreover, some beta-COP colocalizes with intracellular caveolin-1, which was previously shown to be present on a new organelle (termed the caveosome) that is an intermediate in the transport of SV40 to the ER (L. Pelkmans, J. Kartenbeck, and A. Helenius, Nat. Cell Biol. 3:473-483, 2001). We also show that the internal SV40 capsid proteins VP2 and VP3 become accessible to immunostaining starting at about 5 h. Most of that immunostaining overlays the ER, with some appearing outside of the ER. In contrast, immunostaining with anti-SV40 antisera remains confined to the ER.


Assuntos
Antivirais/farmacologia , Brefeldina A/farmacologia , Cavéolas/virologia , Retículo Endoplasmático/virologia , Vírus 40 dos Símios/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Capsídeo/análise , Capsídeo/metabolismo , Cavéolas/metabolismo , Proteína Coatomer/análise , Proteína Coatomer/metabolismo , Endocitose , Retículo Endoplasmático/metabolismo , Replicação Viral
16.
J Virol ; 76(4): 1856-65, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11799180

RESUMO

Echovirus 1 (EV1) is a human pathogen which belongs to the Picornaviridae family of RNA viruses. We have analyzed the early events of infection after EV1 binding to its receptor alpha 2 beta 1 integrin and elucidated the route by which EV1 gains access to the host cell. EV1 binding onto the cell surface and subsequent entry resulted in conformational changes of the viral capsid as demonstrated by sucrose gradient sedimentation analysis. After 15 min to 2 h postinfection (p.i.) EV1 capsid proteins were seen in vesicular structures that were negative for markers of the clathrin-dependent endocytic pathway. In contrast, immunofluorescence confocal microscopy showed that EV1, alpha 2 beta 1 integrin, and caveolin-1 were internalized together in vesicular structures to the perinuclear area. Electron microscopy showed the presence of EV1 particles inside caveolae. Furthermore, infective EV1 could be isolated with anti-caveolin-1 beads 15 min p.i., confirming a close association with caveolin-1. Finally, the expression of dominant negative caveolin in cells markedly inhibited EV1 infection, indicating the importance of caveolae for the viral replication cycle of EV1.


Assuntos
Cavéolas/virologia , Enterovirus Humano B/patogenicidade , Infecções por Enterovirus/virologia , Animais , Capsídeo/metabolismo , Cavéolas/ultraestrutura , Caveolina 1 , Caveolinas/metabolismo , Clatrina/metabolismo , Enterovirus Humano B/ultraestrutura , Humanos , Integrinas/metabolismo , Microscopia Confocal , Microscopia Eletrônica , Coelhos , Receptores de Colágeno , Células Tumorais Cultivadas/ultraestrutura , Células Tumorais Cultivadas/virologia , Microglobulina beta-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA