Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 817
Filtrar
1.
Anal Bioanal Chem ; 413(29): 7157-7178, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34490501

RESUMEN

The objective of this critical review is to provide an overview of how emerging bioanalytical techniques are expanding our understanding of the complex physicochemical nature of virus interactions with host cell surfaces. Herein, selected model viruses representing both non-enveloped (simian virus 40 and human norovirus) and enveloped (influenza A virus, human herpes simplex virus, and human immunodeficiency virus type 1) viruses are highlighted. The technologies covered utilize a wide range of cell membrane mimics, from supported lipid bilayers (SLBs) containing a single purified host membrane component to SLBs derived from the plasma membrane of a target cell, which can be compared with live-cell experiments to better understand the role of individual interaction pairs in virus attachment and entry. These platforms are used to quantify binding strengths, residence times, diffusion characteristics, and binding kinetics down to the single virus particle and single receptor, and even to provide assessments of multivalent interactions. The technologies covered herein are surface plasmon resonance (SPR), quartz crystal microbalance with dissipation (QCM-D), dynamic force spectroscopy (DFS), total internal reflection fluorescence (TIRF) microscopy combined with equilibrium fluctuation analysis (EFA) and single particle tracking (SPT), and finally confocal microscopy using multi-labeling techniques to visualize entry of individual virus particles in live cells. Considering the growing scientific and societal needs for untangling, and interfering with, the complex mechanisms of virus binding and entry, we hope that this review will stimulate the community to implement these emerging tools and strategies in conjunction with more traditional methods. The gained knowledge will not only contribute to a better understanding of the virus biology, but may also facilitate the design of effective inhibitors to block virus entry.


Asunto(s)
Membrana Celular/virología , Interacciones Huésped-Patógeno/fisiología , Biología Molecular/métodos , Membrana Celular/química , Membrana Celular/metabolismo , Glicosaminoglicanos/metabolismo , VIH-1/patogenicidad , VIH-1/fisiología , Herpesvirus Humano 1/patogenicidad , Herpesvirus Humano 1/fisiología , Humanos , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/fisiología , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Norovirus/patogenicidad , Norovirus/fisiología , Polisacáridos/metabolismo , Virus 40 de los Simios/patogenicidad , Virus 40 de los Simios/fisiología , Internalización del Virus
2.
Antiviral Res ; 179: 104819, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32389733

RESUMEN

During virus entry, members of the Polyomaviridae transit the endolysosomal network en route to the endoplasmic reticulum (ER), from which degraded capsids escape into the cytoplasm and enter the nucleus. Emerging evidence suggests that viruses require both endosomal acidification and the correct ionic balance of K+ and Ca2+ ions in endosomes for correct virus trafficking and genome release. Here, using two polyomaviruses with different capsid architectures, namely Simian virus 40 (SV40) and Merkel cell polyomavirus (MCPyV), we describe methods to rapidly quantify virus infection using IncuCyte ZOOM imaging analysis, and use this system to investigate the role of both K+ and Ca2+ channels during the early stages of virus entry. Using broad spectrum blockers of both K+ and Ca2+ channels to specifically target host cell ion channel functionality, we show that MCPyV, but not SV40 can be inhibited by K+ channel modulators, whilst both viruses are restricted by the broad spectrum Ca2+ channel inhibitor verapamil. Using a panel of more specific Ca2+ blockers, we show that both MCPyV and SV40 are dependent on the activity of two-pore Ca2+ channels (TPCs), as the TPC-specific blocker tetrandrine prevented capsid disassembly and nuclear transport required for virus entry. We therefore reveal a novel target to restrict the entry of polyomaviruses, which given the known role of TPCs during endolysosomal-ER fusion, is likely to be applicable to other viruses that transit this pathway.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Endosomas/fisiología , Poliomavirus/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Internalización del Virus/efectos de los fármacos , Animales , Bencilisoquinolinas/farmacología , Línea Celular , Movimiento Celular , Chlorocebus aethiops , Descubrimiento de Drogas , Endosomas/virología , Células HEK293 , Humanos , Poliomavirus de Células de Merkel/efectos de los fármacos , Poliomavirus de Células de Merkel/fisiología , Poliomavirus/fisiología , Virus 40 de los Simios/efectos de los fármacos , Virus 40 de los Simios/fisiología , Verapamilo/farmacología , Células Vero
3.
Nat Commun ; 11(1): 1127, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32111841

RESUMEN

Although viruses must navigate the complex host endomembrane system to infect cells, the strategies used to achieve this is unclear. During entry, polyomavirus SV40 is sorted from the late endosome (LE) to the endoplasmic reticulum (ER) to cause infection, yet how this is accomplished remains enigmatic. Here we find that EMC4 and EMC7, two ER membrane protein complex (EMC) subunits, support SV40 infection by promoting LE-to-ER targeting of the virus. They do this by engaging LE-associated Rab7, presumably to stabilize contact between the LE and ER. These EMC subunits also bind to the ER-resident fusion machinery component syntaxin18, which is required for SV40-arrival to the ER. Our data suggest that EMC4 and EMC7 act as molecular tethers, inter-connecting two intracellular compartments to enable efficient transport of a virus between these compartments. As LE-to-ER transport of cellular cargos is unclear, our results have broad implications for illuminating inter-organelle cargo transport.


Asunto(s)
Retículo Endoplásmico/metabolismo , Membranas Intracelulares/metabolismo , Proteínas de la Membrana/metabolismo , Internalización del Virus , Animales , Sitios de Unión , Células COS , Línea Celular , Chlorocebus aethiops , Retículo Endoplásmico/virología , Endosomas/metabolismo , Endosomas/virología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Membranas Intracelulares/virología , Proteínas de la Membrana/genética , Unión Proteica , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo , Virus 40 de los Simios/fisiología , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
4.
Viruses ; 11(10)2019 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-31590347

RESUMEN

During entry, a virus must be transported through the endomembrane system of the host cell, penetrate a cellular membrane, and undergo capsid disassembly, to reach the cytosol and often the nucleus in order to cause infection. To do so requires the virus to coordinately exploit the action of cellular membrane transport, penetration, and disassembly machineries. How this is accomplished remains enigmatic for many viruses, especially for viruses belonging to the nonenveloped virus family. In this review, we present the current model describing infectious entry of the nonenveloped polyomavirus (PyV) SV40. Insights from SV40 entry are likely to provide strategies to combat PyV-induced diseases, and to illuminate cellular trafficking, membrane transport, and disassembly mechanisms.


Asunto(s)
Transporte Biológico/fisiología , Membranas/metabolismo , Infecciones por Polyomavirus/metabolismo , Virus 40 de los Simios/fisiología , Núcleo Celular/virología , Citosol/metabolismo , Citosol/virología , Endocitosis , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Endosomas/virología , Aparato de Golgi , Humanos , Membranas/virología , Virus 40 de los Simios/patogenicidad , Infecciones Tumorales por Virus/metabolismo , Ensamble de Virus/fisiología , Internalización del Virus , Replicación Viral
5.
J Virol ; 93(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30700597

RESUMEN

JC polyomavirus (JCPyV) establishes a persistent, lifelong, asymptomatic infection within the kidney of the majority of the human population. Under conditions of severe immunosuppression or immune modulation, JCPyV can reactivate in the central nervous system (CNS) and cause progressive multifocal leukoencephalopathy (PML), a fatal demyelinating disease. Initiation of infection is mediated through viral attachment to α2,6-sialic acid-containing lactoseries tetrasaccharide c (LSTc) on the surface of host cells. JCPyV internalization is dependent on serotonin 5-hydroxytryptamine subfamily 2 receptors (5-HT2Rs), and entry is thought to occur by clathrin-mediated endocytosis (CME). However, the JCPyV entry process and the cellular factors involved in viral internalization remain poorly understood. Treatment of cells with small-molecule chemical inhibitors and RNA interference of 5-HT2R endocytic machinery, including ß-arrestin, clathrin, AP2, and dynamin, significantly reduced JCPyV infection. However, infectivity of the polyomavirus simian virus 40 (SV40) was not affected by CME-specific treatments. Inhibition of clathrin or ß-arrestin specifically reduced JCPyV internalization but did not affect viral attachment. Furthermore, mutagenesis of a ß-arrestin binding domain (Ala-Ser-Lys) within the intracellular C terminus of 5-HT2AR severely diminished internalization and infection, suggesting that ß-arrestin interactions with 5-HT2AR are critical for JCPyV infection and entry. These conclusions illuminate key host factors that regulate clathrin-mediated endocytosis of JCPyV, which is necessary for viral internalization and productive infection.IMPORTANCE Viruses usurp cellular factors to invade host cells. Activation and utilization of these proteins upon initiation of viral infection are therefore required for productive infection and resultant viral disease. The majority of healthy individuals are asymptomatically infected by JC polyomavirus (JCPyV), but if the host immune system is compromised, JCPyV can cause progressive multifocal leukoencephalopathy (PML), a rare, fatal, demyelinating disease. Individuals infected with HIV or taking prolonged immunomodulatory therapies have a heightened risk for developing PML. The cellular proteins and pathways utilized by JCPyV to mediate viral entry are poorly understood. Our findings further characterize how JCPyV utilizes the clathrin-mediated endocytosis pathway to invade host cells. We have identified specific components of this pathway that are necessary for the viral entry process and infection. Collectively, the conclusions increase our understanding of JCPyV infection and pathogenesis and may contribute to the future development of novel therapeutic strategies for PML.


Asunto(s)
Clatrina/metabolismo , Endocitosis , Virus JC/fisiología , Internalización del Virus , beta-Arrestinas/metabolismo , Células HEK293 , Humanos , Receptores de Serotonina/metabolismo , Virus 40 de los Simios/fisiología
6.
J Virol ; 93(1)2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30333173

RESUMEN

Host range (HR) mutants of simian virus 40 (SV40) containing mutations in the C terminus of large T antigen fail to replicate efficiently or form plaques in restrictive cell types. HR mutant viruses exhibit impairments at several stages of the viral life cycle, including early and late gene and protein expression, DNA replication, and virion assembly, although the underlying mechanism for these defects is unknown. Host protein FAM111A, whose depletion rescues early and late gene expression and plaque formation for SV40 HR viruses, has been shown to play a role in cellular DNA replication. SV40 viral DNA replication occurs in the nucleus of infected cells in viral replication centers where viral proteins and cellular replication factors localize. Here, we examined the role of viral replication center formation and DNA replication in the FAM111A-mediated HR phenotype. We found that SV40 HR virus rarely formed viral replication centers in restrictive cells, a phenotype that could be rescued by FAM111A depletion. Furthermore, while FAM111A localized to nucleoli in uninfected cells in a cell cycle-dependent manner, FAM111A relocalized to viral replication centers after infection with SV40 wild-type or HR viruses. We also found that inhibition of viral DNA replication through aphidicolin treatment or through the use of replication-defective SV40 mutants diminished the effects of FAM111A depletion on viral gene expression. These results indicate that FAM111A restricts SV40 HR viral replication center formation and that viral DNA replication contributes to the FAM111A-mediated effect on early gene expression.IMPORTANCE SV40 has served as a powerful tool for understanding fundamental viral and cellular processes; however, despite extensive study, the SV40 HR mutant phenotype remains poorly understood. Mutations in the C terminus of large T antigen that disrupt binding to the host protein FAM111A render SV40 HR viruses unable to replicate in restrictive cell types. Our work reveals a defect of HR mutant viruses in the formation of viral replication centers that can be rescued by depletion of FAM111A. Furthermore, inhibition of viral DNA replication reduces the effects of FAM111A restriction on viral gene expression. Additionally, FAM111A is a poorly characterized cellular protein whose mutation leads to two severe human syndromes, Kenny-Caffey syndrome and osteocraniostenosis. Our findings regarding the role of FAM111A in restricting viral replication and its localization to nucleoli and viral replication centers provide further insight into FAM111A function that could help reveal the underlying disease-associated mechanisms.


Asunto(s)
Antígenos Virales de Tumores/genética , Proteínas de Ciclo Celular/metabolismo , ADN Viral/metabolismo , Virus 40 de los Simios/fisiología , Animales , Antígenos Virales de Tumores/química , Proteínas de Ciclo Celular/genética , Línea Celular , Núcleo Celular/virología , Chlorocebus aethiops , Regulación Viral de la Expresión Génica , Especificidad del Huésped , Humanos , Mutación , Fenotipo , Virus 40 de los Simios/genética , Virus 40 de los Simios/inmunología , Replicación Viral
7.
Angew Chem Int Ed Engl ; 57(43): 14032-14036, 2018 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-30063096

RESUMEN

We report a strategy to construct peptidyl virus-like particles (pVLPs) by mimicking the human immunodeficiency virus and simian virus 40. We designed two viral peptides with cell/nucleus-targeting capabilities that can co-assemble in their active conformations into well-defined nanoparticles. The self-assembled nanoparticles can encapsulate the DNA of clustered regularly interspaced short palindromic repeat associated proteins 9 (CRISPR/Cas9) to form biodegradable pVLPs with excellent cell-targeting ability and biocompatibility. The pVLPs can penetrate the cellular membrane and deliver genetic cargos into the nucleus through the viral entry route. The results provide a promising pathway for engineering artificial viruses with desired functions.


Asunto(s)
Técnicas de Transferencia de Gen , Péptidos/química , Virión/química , Sistemas CRISPR-Cas , Línea Celular , Ensayo de Cambio de Movilidad Electroforética , VIH/química , VIH/fisiología , Humanos , Fusión de Membrana , Microscopía de Fuerza Atómica , Microscopía Electrónica de Transmisión , Nanopartículas/química , Virus 40 de los Simios/química , Virus 40 de los Simios/fisiología
8.
J Virol ; 92(12)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29593037

RESUMEN

During entry, polyomavirus (PyV) is endocytosed and sorts to the endoplasmic reticulum (ER), where it penetrates the ER membrane to reach the cytosol. From the cytosol, the virus moves to the nucleus to cause infection. How PyV is transported from the cytosol into the nucleus, a crucial infection step, is unclear. We found that upon reaching the cytosol, the archetypal PyV simian virus 40 (SV40) recruits the cytoplasmic dynein motor, which disassembles the viral particle. This reaction enables the resulting disassembled virus to enter the nucleus to promote infection. Our findings reveal how a cytosolic motor can be hijacked to impart conformational changes to a viral particle, a process essential for successful infection.IMPORTANCE How a nonenveloped virus successfully traffics from the cell surface to the nucleus to cause infection remains enigmatic in many instances. In the case of the nonenveloped PyV, the viral particle is sorted from the plasma membrane to the ER and then the cytosol, from which it enters the nucleus to promote infection. The molecular mechanism by which PyV reaches the nucleus from the cytosol is not entirely clear. Here we demonstrate that the prototype PyV SV40 recruits dynein upon reaching the cytosol. Importantly, this cellular motor disassembles the viral particle during cytosol-to-nucleus transport to cause infection.


Asunto(s)
Citosol/virología , Dineínas/metabolismo , Mapeo de Interacción de Proteínas/métodos , Virus 40 de los Simios/patogenicidad , Animales , Células COS , Línea Celular , Núcleo Celular/virología , Chlorocebus aethiops , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/virología , Virus 40 de los Simios/química , Virus 40 de los Simios/fisiología , Internalización del Virus
9.
Antiviral Res ; 152: 68-75, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29458134

RESUMEN

The ability of antiviral polyamides (AVP) to act upon polyomaviruses (PyV) was evaluated. Initial studies found that a single treatment of AVP protected SV40-infected BSC-1 cells from cytopathic effect (CPE) for as long as 11 days p.i.. AVP substantially suppressed SV40 genome copy numbers over the duration of the experiment. Immunofluorescence analysis of ataxia-telangiectasia mutated (ATM) activation and large T antigen (LTag) expression clearly demonstrated that AVP treatment at day 1 p.i. delayed the onset of productive SV40 replication by approximately 3 days, and substantially limited the infection relative to vehicle-treated controls. AVP dose-response experiments recorded IC50s in the low nM range that were similar to IC50s previously reported for HPV16. The ability of AVPs to act on BKPyV was next examined. Again, IC50s in the low nM range were obtained with the exception of an AVP (PA1) that gave an IC50 of 437 nM against the BKPyV Dunlop strain. The Mre11 inhibitor Mirin substantially reduced the AVP IC50 against SV40 demonstrating that Mre11 protects PyV genomes from AVP action as previously shown for HPV. Together these experiments show that AVPs are potent antiviral agents for PyV that act via a mechanism with similarities to that found for HPV. The results demonstrate that AVPs are useful tools for controlling and studying PyV biology. The potential use of these agents against BKPyV and other PyV pathogens also has clinical implications.


Asunto(s)
Antivirales/farmacología , Virus BK/efectos de los fármacos , Imidazoles/farmacología , Nylons/farmacología , Infecciones por Polyomavirus/virología , Pirroles/farmacología , Virus 40 de los Simios/efectos de los fármacos , Infecciones Tumorales por Virus/virología , Antivirales/química , Virus BK/genética , Virus BK/fisiología , Replicación del ADN/efectos de los fármacos , Humanos , Imidazoles/química , Nylons/química , Pirroles/química , Virus 40 de los Simios/genética , Virus 40 de los Simios/fisiología
10.
J Med Primatol ; 47(1): 81-84, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28671309

RESUMEN

Recrudescence of latent and dormant viruses may lead to overwhelming viremia in immunosuppressed hosts. In immunocompromised hosts, Simian virus 40 (SV40) reactivation is known to cause nephritis and demyelinating central nervous system disease. Here, we report SV40 viremia leading to fatal interstitial pneumonia in an immunosuppressed host following renal allotransplantation.


Asunto(s)
Huésped Inmunocomprometido , Enfermedades Renales/fisiopatología , Macaca mulatta , Enfermedades de los Monos/fisiopatología , Neumonía/fisiopatología , Infecciones por Polyomavirus/veterinaria , Virus 40 de los Simios/fisiología , Infecciones Tumorales por Virus/veterinaria , Animales , Enfermedades Renales/virología , Trasplante de Riñón/veterinaria , Enfermedades de los Monos/virología , Neumonía/virología , Infecciones por Polyomavirus/complicaciones , Infecciones Tumorales por Virus/complicaciones
11.
Trends Microbiol ; 26(6): 525-537, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29079499

RESUMEN

Disruption of host membranes by nonenveloped viruses, which allows the nucleocapsid or genome to enter the cytosol, is a mechanistically diverse process. Although the membrane-penetrating agents are usually small, hydrophobic or amphipathic peptides deployed from the capsid interior during entry, their manner of membrane interaction varies substantially. In this review, we discuss recent data about the molecular pathways for externalization of viral peptides amidst conformational alterations in the capsid, as well as mechanisms of membrane penetration, which is influenced by structural features of the peptides themselves as well as physicochemical properties of membranes, and other host factors. The membrane-penetrating components of nonenveloped viruses constitute an interesting class of cell-penetrating peptides, and may have potential therapeutic value for gene transfer.


Asunto(s)
Proteínas de la Cápside/fisiología , Membrana Celular/virología , Interacciones Microbiota-Huesped , Internalización del Virus , Cápside/fisiología , Péptidos de Penetración Celular/fisiología , Citosol/virología , Humanos , Poliomavirus/fisiología , Virus 40 de los Simios/fisiología , Virión/fisiología
12.
mBio ; 8(5)2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28974621

RESUMEN

Inflammatory responses, while essential for pathogen clearance, can also be deleterious to the host. Chemical inhibition of topoisomerase 1 (Top1) by low-dose camptothecin (CPT) can suppress transcriptional induction of antiviral and inflammatory genes and protect animals from excessive and damaging inflammatory responses. We describe the unexpected finding that minor DNA damage from topoisomerase 1 inhibition with low-dose CPT can trigger a strong antiviral immune response through cyclic GMP-AMP synthase (cGAS) detection of cytoplasmic DNA. This argues against CPT having only anti-inflammatory activity. Furthermore, expression of the simian virus 40 (SV40) large T antigen was paramount to the proinflammatory antiviral activity of CPT, as it potentiated cytoplasmic DNA leakage and subsequent cGAS recruitment in human and mouse cell lines. This work suggests that the capacity of Top1 inhibitors to blunt inflammatory responses can be counteracted by viral oncogenes and that this should be taken into account for their therapeutic development.IMPORTANCE Recent studies suggest that low-dose DNA-damaging compounds traditionally used in cancer therapy can have opposite effects on antiviral responses, either suppressing (with the example of CPT) or potentiating (with the example of doxorubicin) them. Our work demonstrates that the minor DNA damage promoted by low-dose CPT can also trigger strong antiviral responses, dependent on the presence of viral oncogenes. Taken together, these results call for caution in the therapeutic use of low-dose chemotherapy agents to modulate antiviral responses in humans.


Asunto(s)
ADN-Topoisomerasas de Tipo I/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Nucleótidos Cíclicos/metabolismo , Virus 40 de los Simios/efectos de los fármacos , Inhibidores de Topoisomerasa I/farmacología , Animales , Antígenos Virales de Tumores/genética , Antígenos Virales de Tumores/inmunología , Antivirales/farmacología , Camptotecina/farmacología , Línea Celular , Técnicas de Cocultivo , Daño del ADN , ADN-Topoisomerasas de Tipo I/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Humanos , Inflamación , Ratones , Virus 40 de los Simios/inmunología , Virus 40 de los Simios/fisiología , Virosis/tratamiento farmacológico , Virosis/inmunología , Virosis/virología
13.
Curr Protoc Microbiol ; 46: 14F.3.1-14F.3.26, 2017 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-28800155

RESUMEN

Simian virus 40 (SV40) is one of the best-characterized members of the polyomavirus family of small DNA tumor viruses. It has a small genome of 5243 bp and utilizes cellular proteins for its molecular biology, with the exception of the T-antigen protein, which is coded by the virus and is involved in regulating transcription and directing replication. Importantly, SV40 exists as chromatin in both the virus particle and intracellular minichromosomes. These facts, combined with high yields of virus and minichromosomes following infection and ease of manipulation, have made SV40 an extremely useful model to study all aspects of eukaryotic molecular biology. This unit describes procedures for working with SV40 and preparing SV40 chromatin from infected cells and virus particles, as well as procedures for using SV40 chromatin to study epigenetic regulation. © 2017 by John Wiley & Sons, Inc.


Asunto(s)
Epigenómica/métodos , Infecciones por Polyomavirus/virología , Virus 40 de los Simios/genética , Infecciones Tumorales por Virus/virología , Cultivo de Virus/métodos , Animales , Epigénesis Genética , Humanos , Virus 40 de los Simios/fisiología
14.
PLoS Pathog ; 13(6): e1006439, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28614383

RESUMEN

The molecular mechanism by which non-enveloped viruses penetrate biological membranes remains enigmatic. The non-enveloped polyomavirus SV40 penetrates the endoplasmic reticulum (ER) membrane to reach the cytosol and cause infection. We previously demonstrated that SV40 creates its own membrane penetration structure by mobilizing select transmembrane proteins to distinct puncta in the ER membrane called foci that likely function as the cytosol entry sites. How these ER membrane proteins reorganize into the foci is unknown. B12 is a transmembrane J-protein that mobilizes into the foci to promote cytosol entry of SV40. Here we identify two closely related ER membrane proteins Erlin1 and Erlin2 (Erlin1/2) as B12-interaction partners. Strikingly, SV40 recruits B12 to the foci by inducing release of this J-protein from Erlin1/2. Our data thus reveal how a non-enveloped virus promotes its own membrane translocation by triggering the release and recruitment of a critical transport factor to the membrane penetration site.


Asunto(s)
Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Virus 40 de los Simios/fisiología , Internalización del Virus , Línea Celular , Retículo Endoplásmico/virología , Técnicas de Silenciamiento del Gen , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/virología , Infecciones por Polyomavirus/metabolismo
15.
Nat Commun ; 8: 15496, 2017 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-28537258

RESUMEN

Viruses exploit cellular machineries to penetrate a host membrane and cause infection, a process that remains enigmatic for non-enveloped viruses. Here we probe how the non-enveloped polyomavirus SV40 penetrates the endoplasmic reticulum (ER) membrane to reach the cytosol, a crucial infection step. We find that the microtubule-based motor kinesin-1 is recruited to the ER membrane by binding to the transmembrane J-protein B14. Strikingly, this motor facilitates SV40 ER-to-cytosol transport by constructing a penetration site on the ER membrane called a 'focus'. Neither kinesin-2, kinesin-3 nor kinesin-5 promotes foci formation or infection. The specific use of kinesin-1 is due to its unique ability to select posttranslationally modified microtubules for cargo transport and thereby spatially restrict focus formation to the perinucleus. These findings support the idea of a 'tubulin code' for motor-dependent trafficking and establish a distinct kinesin-1 function in which a motor is exploited to create a viral membrane penetration site.


Asunto(s)
Retículo Endoplásmico/metabolismo , Membranas Intracelulares/metabolismo , Cinesinas/metabolismo , Virus 40 de los Simios/fisiología , Internalización del Virus , Animales , Células COS , Chlorocebus aethiops , Citosol/metabolismo , Citosol/virología , Retículo Endoplásmico/virología , Técnicas de Silenciamiento del Gen , Células HEK293 , Proteínas del Choque Térmico HSP47/genética , Proteínas del Choque Térmico HSP47/metabolismo , Humanos , Membranas Intracelulares/virología , Microscopía Intravital , Cinesinas/genética , Microtúbulos/metabolismo , Chaperonas Moleculares , ARN Interferente Pequeño/metabolismo , Virus 40 de los Simios/patogenicidad , Virión/metabolismo
16.
J Virol ; 91(12)2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28356524

RESUMEN

Membrane penetration by nonenveloped viruses remains enigmatic. In the case of the nonenveloped polyomavirus simian virus 40 (SV40), the virus penetrates the endoplasmic reticulum (ER) membrane to reach the cytosol and then traffics to the nucleus to cause infection. We previously demonstrated that the cytosolic Hsc70-SGTA-Hsp105 complex is tethered to the ER membrane, where Hsp105 and SGTA facilitate the extraction of SV40 from the ER and transport of the virus into the cytosol. We now find that Hsc70 also ejects SV40 from the ER into the cytosol in a step regulated by SGTA. Although SGTA's N-terminal domain, which mediates homodimerization and recruits cellular adaptors, is dispensable during ER-to-cytosol transport of SV40, this domain appears to exert an unexpected post-ER membrane translocation function during SV40 entry. Our study thus establishes a critical function of Hsc70 within the Hsc70-SGTA-Hsp105 complex in promoting SV40 ER-to-cytosol membrane penetration and unveils a role of SGTA in controlling this step.IMPORTANCE How a nonenveloped virus transports across a biological membrane to cause infection remains mysterious. One enigmatic step is whether host cytosolic components are co-opted to transport the viral particle into the cytosol. During ER-to-cytosol membrane transport of the nonenveloped polyomavirus SV40, a decisive infection step, a cytosolic complex composed of Hsc70-SGTA-Hsp105 was previously shown to associate with the ER membrane. SGTA and Hsp105 have been shown to extract SV40 from the ER and transport the virus into the cytosol. We demonstrate here a critical role of Hsc70 in SV40 ER-to-cytosol penetration and reveal how SGTA controls Hsc70 to impact this process.


Asunto(s)
Proteínas Portadoras/metabolismo , Citosol/virología , Retículo Endoplásmico/virología , Proteínas del Choque Térmico HSC70/metabolismo , Virus 40 de los Simios/fisiología , Internalización del Virus , Animales , Transporte Biológico/fisiología , Células COS , Proteínas Portadoras/genética , Línea Celular , Chlorocebus aethiops , Citosol/metabolismo , Retículo Endoplásmico/fisiología , Regulación de la Expresión Génica , Células HEK293 , Proteínas del Choque Térmico HSC70/genética , Interacciones Huésped-Patógeno/genética , Humanos , Membranas Intracelulares/virología , Chaperonas Moleculares/metabolismo , ARN Interferente Pequeño
17.
J Cell Physiol ; 232(11): 3060-3066, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27925194

RESUMEN

The Mesenchymal Stromal Cells from umbilical cord Wharton's jelly (WJSCs) are a source of cells with high potentiality for the treatment of human immunological disorders. Footprints of the oncogenic viruses Simian Virus 40 (SV40) and JC Virus (JCPyV) have been recently detected in human WJSCs specimens. The aim of this study is to evaluate if WJSCs can be efficiently infected by these Polyomaviruses and if they can potentially exert tumoral activity. Cell culture experiments indicated that WJSCs could sustain both SV40 and JCPyV infections. A transient and lytic replication was observed for JCPyV, while SV40 persistently infected WJSCs over a long period of time, releasing a viral progeny at low titer without evident cytopathic effect (CPE). Considering the association between SV40 and human tumors and the reported ability of the oncogenic viruses to drive the host innate immune response to cell transformation, the expression profile of a large panel of immune mediators was evaluated in supernatants by the Bioplex platform. RANTES, IL-3, MIG, and IL-12p40, involved in chronic inflammation, cells differentiation, and transformation, were constantly measured at high concentration comparing to control. These findings represent a new aspect of SV40 biological activity in the humans, highlighting its interaction with specific host cellular pathways. In view of these results, it seems to be increasingly urgent to consider Polyomaviruses in the management of WJSCs for their safely use as promising therapeutic source. J. Cell. Physiol. 232: 3060-3066, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Transformación Celular Viral , Mediadores de Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/virología , Virus 40 de los Simios/fisiología , Gelatina de Wharton/citología , Línea Celular Transformada , Separación Celular/métodos , Quimiocina CCL5/metabolismo , Quimiocina CXCL9/metabolismo , Efecto Citopatogénico Viral , ADN Viral/biosíntesis , ADN Viral/genética , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/inmunología , Subunidad p40 de la Interleucina-12/metabolismo , Interleucina-3/metabolismo , Virus JC/fisiología , Células Madre Mesenquimatosas/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Virus 40 de los Simios/genética , Virus 40 de los Simios/inmunología , Factores de Tiempo , Regulación hacia Arriba , Carga Viral , Replicación Viral
18.
Elife ; 52016 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-28012275

RESUMEN

Destabilization of a non-enveloped virus generates a membrane transport-competent viral particle. Here we probe polyomavirus SV40 endoplasmic reticulum (ER)-to-cytosol membrane transport, a decisive infection step where destabilization initiates this non-enveloped virus for membrane penetration. We find that a member of the ER membrane protein complex (EMC) called EMC1 promotes SV40 ER membrane transport and infection. Surprisingly, EMC1 does so by using its predicted transmembrane residue D961 to bind to and stabilize the membrane-embedded partially destabilized SV40, thereby preventing premature viral disassembly. EMC1-dependent stabilization enables SV40 to engage a cytosolic extraction complex that ejects the virus into the cytosol. Thus EMC1 acts as a molecular chaperone, bracing the destabilized SV40 in a transport-competent state. Our findings reveal the novel principle that coordinated destabilization-stabilization drives membrane transport of a non-enveloped virus.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas/metabolismo , Virus 40 de los Simios/fisiología , Internalización del Virus , Animales , Transporte Biológico , Células COS , Células HEK293 , Humanos , Proteínas de la Membrana
19.
Oncotarget ; 7(33): 52643-52660, 2016 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-27462916

RESUMEN

SV40 large T-antigen (T-ag) has been known for decades to inactivate the tumor suppressor p53 by sequestration and additional mechanisms. Our present study revealed that the struggle between p53 and T-ag begins very early in the infection cycle. We found that p53 is activated early after SV40 infection and defends the host against the infection. Using live cell imaging and single cell analyses we found that p53 dynamics are variable among individual cells, with only a subset of cells activating p53 immediately after SV40 infection. This cell-to-cell variabilty had clear consequences on the outcome of the infection. None of the cells with elevated p53 at the beginning of the infection proceeded to express T-ag, suggesting a p53-dependent decision between abortive and productive infection. In addition, we show that artificial elevation of p53 levels prior to the infection reduces infection efficiency, supporting a role for p53 in defending against SV40. We further found that the p53-mediated host defense mechanism against SV40 is not facilitated by apoptosis nor via interferon-stimulated genes. Instead p53 binds to the viral DNA at the T-ag promoter region, prevents its transcriptional activation by Sp1, and halts the progress of the infection. These findings shed new light on the long studied struggle between SV40 T-ag and p53, as developed during virus-host coevolution. Our studies indicate that the fate of SV40 infection is determined as soon as the viral DNA enters the nucleus, before the onset of viral gene expression.


Asunto(s)
Antígenos Transformadores de Poliomavirus/genética , Regulación Viral de la Expresión Génica , Virus 40 de los Simios/genética , Proteína p53 Supresora de Tumor/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/virología , Línea Celular , Regulación Neoplásica de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Células MCF-7 , Microscopía Confocal , Regiones Promotoras Genéticas/genética , Unión Proteica , Virus 40 de los Simios/fisiología , Factor de Transcripción Sp1/metabolismo , Imagen de Lapso de Tiempo/métodos , Proteína p53 Supresora de Tumor/metabolismo
20.
ILAR J ; 57(1): 44-54, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27034394

RESUMEN

The SV40 viral oncogene has been used since the 1970s as a reliable and reproducible method to generate transgenic mouse models. This seminal discovery has taught us an immense amount about how tumorigenesis occurs, and its success has led to the evolution of many mouse models of cancer. Despite the development of more modern and targeted approaches for developing genetically engineered mouse models of cancer, SV40-induced mouse models still remain frequently used today. This review discusses a number of cancer types in which SV40 mouse models of cancer have been developed and highlights their relevance and importance to preclinical research.


Asunto(s)
Ratones Transgénicos , Neoplasias/virología , Animales , Ratones , Virus 40 de los Simios/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...