Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
2.
J Med Virol ; 96(4): e29620, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38647027

RESUMEN

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


Asunto(s)
Monkeypox virus , Seudópodos , Trofoblastos , Trofoblastos/virología , Humanos , Seudópodos/virología , Femenino , Embarazo , Monkeypox virus/fisiología , Liberación del Virus , Replicación Viral , Citoesqueleto/virología , Placenta/virología , Placenta/citología , Virión/ultraestructura , Microscopía/métodos , Línea Celular
3.
J Virol ; 96(12): e0220521, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35638850

RESUMEN

The pathogenesis of white spot syndrome virus (WSSV) is largely unclear. In this study, we found that actin nucleation and clathrin-mediated endocytosis were recruited for internalization of WSSV into crayfish hematopoietic tissue (Hpt) cells. This internalization was followed by intracellular transport of the invading virions via endocytic vesicles and endosomes. After envelope fusion within endosomes, the penetrated nucleocapsids were transported along microtubules toward the periphery of the nuclear pores. Furthermore, the nuclear transporter CqImportin α1/ß1, via binding of ARM repeat domain within CqImportin α1 to the nuclear localization sequences (NLSs) of viral cargoes and binding of CqImportin ß1 to the nucleoporins CqNup35/62 with the action of CqRan for docking to nuclear pores, was hijacked for both targeting of the incoming nucleocapsids toward the nuclear pores and import of the expressed viral structural proteins containing NLS into the cell nucleus. Intriguingly, dysfunction of CqImportin α1/ß1 resulted in significant accumulation of incoming nucleocapsids on the periphery of the Hpt cell nucleus, leading to substantially decreased introduction of the viral genome into the nucleus and remarkably reduced nuclear import of expressed viral structural proteins with NLS; both of these effects were accompanied by significantly inhibited viral propagation. Accordingly, the survival rate of crayfish post-WSSV challenge was significantly increased after dysfunction of CqImportin α1/ß1, also showing significantly reduced viral propagation, and was induced either by gene silencing or by pharmacological blockade via dietary administration of ivermectin per os. Collectively, our findings improve our understanding of WSSV pathogenesis and support future antiviral designing against WSSV. IMPORTANCE As one of the largest animal DNA viruses, white spot syndrome virus (WSSV) has been causing severe economical loss in aquaculture due to the limited knowledge on WSSV pathogenesis for an antiviral strategy. We demonstrate that the actin cytoskeleton, endocytic vesicles, endosomes, and microtubules are hijacked for WSSV invasion; importantly, the nuclear transporter CqImportin α1/ß1 together with CqRan were recruited, via binding of CqImportin ß1 to the nucleoporins CqNup35/62, for both the nuclear pore targeting of the incoming nucleocapsids and the nuclear import of expressed viral structural proteins containing the nuclear localization sequences (NLSs). This is the first report that NLSs from both viral structure proteins and host factor are elaborately recruited together to facilitate WSSV infection. Our findings provide a novel explanation for WSSV pathogenesis involving systemic hijacking of host factors, which can be used for antiviral targeting against WSSV disease, such as the blockade of CqImportin α1/ß1 with ivermectin.


Asunto(s)
Transporte Activo de Núcleo Celular , Citoesqueleto , Proteínas Estructurales Virales , Virus del Síndrome de la Mancha Blanca 1 , Animales , Antivirales , Astacoidea/virología , Citoesqueleto/virología , Ivermectina , Microtúbulos , Proteínas de Complejo Poro Nuclear , Replicación Viral , Virus del Síndrome de la Mancha Blanca 1/patogenicidad
4.
Retrovirology ; 18(1): 19, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34229718

RESUMEN

Microtubules (MTs) form a filamentous array that provide both structural support and a coordinated system for the movement and organization of macromolecular cargos within the cell. As such, they play a critical role in regulating a wide range of cellular processes, from cell shape and motility to cell polarization and division. The array is radial with filament minus-ends anchored at perinuclear MT-organizing centers and filament plus-ends continuously growing and shrinking to explore and adapt to the intracellular environment. In response to environmental cues, a small subset of these highly dynamic MTs can become stabilized, acquire post-translational modifications and act as specialized tracks for cargo trafficking. MT dynamics and stability are regulated by a subset of highly specialized MT plus-end tracking proteins, known as +TIPs. Central to this is the end-binding (EB) family of proteins which specifically recognize and track growing MT plus-ends to both regulate MT polymerization directly and to mediate the accumulation of a diverse array of other +TIPs at MT ends. Moreover, interaction of EB1 and +TIPs with actin-MT cross-linking factors coordinate changes in actin and MT dynamics at the cell periphery, as well as during the transition of cargos from one network to the other. The inherent structural polarity of MTs is sensed by specialized motor proteins. In general, dynein directs trafficking of cargos towards the minus-end while most kinesins direct movement toward the plus-end. As a pathogenic cargo, HIV-1 uses the actin cytoskeleton for short-range transport most frequently at the cell periphery during entry before transiting to MTs for long-range transport to reach the nucleus. While the fundamental importance of MT networks to HIV-1 replication has long been known, recent work has begun to reveal the underlying mechanistic details by which HIV-1 engages MTs after entry into the cell. This includes mimicry of EB1 by capsid (CA) and adaptor-mediated engagement of dynein and kinesin motors to elegantly coordinate early steps in infection that include MT stabilization, uncoating (conical CA disassembly) and virus transport toward the nucleus. This review discusses recent advances in our understanding of how MT regulators and their associated motors are exploited by incoming HIV-1 capsid during early stages of infection.


Asunto(s)
Cápside/metabolismo , Citoesqueleto/virología , VIH-1/metabolismo , Interacciones Huésped-Patógeno , Microtúbulos/virología , Transporte Biológico , Proteínas de la Cápside/metabolismo , Citoesqueleto/metabolismo , Humanos
5.
Plant Cell Rep ; 40(7): 1247-1267, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34028582

RESUMEN

KEY MESSAGE: PSV infection changed the abundance of host plant's transcripts and proteins associated with various cellular compartments, including ribosomes, chloroplasts, mitochondria, the nucleus and cytosol, affecting photosynthesis, translation, transcription, and splicing. Virus infection is a process resulting in numerous molecular, cellular, and physiological changes, a wide range of which can be analyzed due to development of many high-throughput techniques. Plant RNA viruses are known to replicate in the cytoplasm; however, the roles of chloroplasts and other cellular structures in the viral replication cycle and in plant antiviral defense have been recently emphasized. Therefore, the aim of this study was to analyze the small RNAs, transcripts, proteins, and phosphoproteins affected during peanut stunt virus strain P (PSV-P)-Nicotiana benthamiana interactions with or without satellite RNA (satRNA) in the context of their cellular localization or functional connections with particular cellular compartments to elucidate the compartments most affected during pathogenesis at the early stages of infection. Moreover, the processes associated with particular cell compartments were determined. The 'omic' results were subjected to comparative data analyses. Transcriptomic and small RNA (sRNA)-seq data were obtained to provide new insights into PSV-P-satRNA-plant interactions, whereas previously obtained proteomic and phosphoproteomic data were used to broaden the analysis to terms associated with cellular compartments affected by virus infection. Based on the collected results, infection with PSV-P contributed to changes in the abundance of transcripts and proteins associated with various cellular compartments, including ribosomes, chloroplasts, mitochondria, the nucleus and the cytosol, and the most affected processes were photosynthesis, translation, transcription, and mRNA splicing. Furthermore, sRNA-seq and phosphoproteomic analyses indicated that kinase regulation resulted in decreases in phosphorylation levels. The kinases were associated with the membrane, cytoplasm, and nucleus components.


Asunto(s)
Cucumovirus/patogenicidad , Nicotiana/citología , Nicotiana/virología , Biología de Sistemas/métodos , Núcleo Celular/genética , Núcleo Celular/virología , Cloroplastos/genética , Cloroplastos/virología , Citoesqueleto/genética , Citoesqueleto/virología , Citosol/virología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Interacciones Huésped-Patógeno/fisiología , MicroARNs , Nitrógeno/metabolismo , Fosfoproteínas/metabolismo , Células Vegetales/virología , Enfermedades de las Plantas/virología , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Mapas de Interacción de Proteínas/genética , Satélite de ARN , Nicotiana/genética
6.
Fungal Genet Biol ; 140: 103385, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32305452

RESUMEN

The rice blast fungus Magnaporthe oryzae differentiates a specialized infection structure called an appressorium, which is used to break into plant cells by directed application of enormous turgor force. Appressorium-mediated plant infection requires timely assembly of a higher-order septin ring structure at the base of the appressorium, which is needed to spatially orchestrate appressorium repolarization. Here we use quantitative 4D widefield fluorescence imaging to gain new insight into the spatiotemporal dynamics of septin ring formation, and septin-mediated actin re-organization, during appressorium morphogenesis by M. oryzae. We anticipate that the new knowledge will provide a quantitative framework for dissecting the molecular mechanisms of higher-order septin ring assembly in this devastating plant pathogenic fungus.


Asunto(s)
Ascomicetos/patogenicidad , Oryza/genética , Enfermedades de las Plantas/genética , Septinas/ultraestructura , Citoesqueleto/genética , Citoesqueleto/virología , Proteínas Fúngicas/genética , Morfogénesis/genética , Oryza/crecimiento & desarrollo , Oryza/virología , Enfermedades de las Plantas/virología , Septinas/química , Septinas/genética
7.
J Neurovirol ; 25(3): 301-312, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30850975

RESUMEN

Human immunodeficiency virus-1 (HIV) infection of the central nervous system damages synapses and promotes axonal injury, ultimately resulting in HIV-associated neurocognitive disorders (HAND). The mechanisms through which HIV causes damage to neurons are still under investigation. The cytoskeleton and associated proteins are fundamental for axonal and dendritic integrity. In this article, we review evidence that HIV proteins, such as the envelope protein gp120 and transactivator of transcription (Tat), impair the structure and function of the neuronal cytoskeleton. Investigation into the effects of viral proteins on the neuronal cytoskeleton may provide a better understanding of HIV neurotoxicity and suggest new avenues for additional therapies.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Complejo SIDA Demencia/patología , Citoesqueleto/virología , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Neuronas/virología , Citoesqueleto/metabolismo , Citoesqueleto/patología , Humanos , Neuronas/metabolismo , Neuronas/patología
8.
J Virol ; 91(7)2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28100610

RESUMEN

The signaling lymphocyte activation molecule F1 (SLAMF1) is both a microbial sensor and entry receptor for measles virus (MeV). Herein, we describe a new role for SLAMF1 to mediate MeV endocytosis that is in contrast with the alternative, and generally accepted, model that MeV genome enters cells only after fusion at the cell surface. We demonstrated that MeV engagement of SLAMF1 induces dramatic but transient morphological changes, most prominently in the formation of membrane blebs, which were shown to colocalize with incoming viral particles, and rearrangement of the actin cytoskeleton in infected cells. MeV infection was dependent on these dynamic cytoskeletal changes as well as fluid uptake through a macropinocytosis-like pathway as chemical inhibition of these processes inhibited entry. Moreover, we identified a role for the RhoA-ROCK-myosin II signaling axis in this MeV internalization process, highlighting a novel role for this recently characterized pathway in virus entry. Our study shows that MeV can hijack a microbial sensor normally involved in bacterial phagocytosis to drive endocytosis using a complex pathway that shares features with canonical viral macropinocytosis, phagocytosis, and mechanotransduction. This uptake pathway is specific to SLAMF1-positive cells and occurs within 60 min of viral attachment. Measles virus remains a significant cause of mortality in human populations, and this research sheds new light on the very first steps of infection of this important pathogen.IMPORTANCE Measles is a significant disease in humans and is estimated to have killed over 200 million people since records began. According to current World Health Organization statistics, it still kills over 100,000 people a year, mostly children in the developing world. The causative agent, measles virus, is a small enveloped RNA virus that infects a broad range of cells during infection. In particular, immune cells are infected via interactions between glycoproteins found on the surface of the virus and SLAMF1, the immune cell receptor. In this study, we have investigated the steps governing entry of measles virus into SLAMF1-positive cells and identified endocytic uptake of viral particles. This research will impact our understanding of morbillivirus-related immunosuppression as well as the application of measles virus as an oncolytic therapeutic.


Asunto(s)
Endocitosis , Virus del Sarampión/fisiología , Sarampión/virología , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/fisiología , Células A549 , Caveolinas/metabolismo , Clatrina/metabolismo , Citoesqueleto/ultraestructura , Citoesqueleto/virología , Dinamina II , Dinaminas/metabolismo , Células HEK293 , Humanos , Microdominios de Membrana/virología , Transducción de Señal , Virión/fisiología , Acoplamiento Viral , Internalización del Virus
9.
Retrovirology ; 12: 78, 2015 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-26376973

RESUMEN

BACKGROUND: HIV-1 replication results in mitochondrial damage that is enhanced during antiretroviral therapy (ART). The onset of HIV-1 replication is regulated by viral protein Tat, a 101-residue protein codified by two exons that elongates viral transcripts. Although the first exon of Tat (aa 1-72) forms itself an active protein, the presence of the second exon (aa 73-101) results in a more competent transcriptional protein with additional functions. RESULTS: Mitochondrial overall functions were analyzed in Jurkat cells stably expressing full-length Tat (Tat101) or one-exon Tat (Tat72). Representative results were confirmed in PBLs transiently expressing Tat101 and in HIV-infected Jurkat cells. The intracellular expression of Tat101 induced the deregulation of metabolism and cytoskeletal proteins which remodeled the function and distribution of mitochondria. Tat101 reduced the transcription of the mtDNA, resulting in low ATP production. The total amount of mitochondria increased likely to counteract their functional impairment. These effects were enhanced when Tat second exon was expressed. CONCLUSIONS: Intracellular Tat altered mtDNA transcription, mitochondrial content and distribution in CD4+ T cells. The importance of Tat second exon in non-transcriptional functions was confirmed. Tat101 may be responsible for mitochondrial dysfunctions found in HIV-1 infected patients.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , ADN Mitocondrial/genética , VIH-1/fisiología , Mitocondrias/ultraestructura , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/ultraestructura , Citoesqueleto/patología , Citoesqueleto/virología , ADN Mitocondrial/metabolismo , Exones , Glucólisis , Humanos , Células Jurkat , Leucocitos Mononucleares , Mitocondrias/genética
10.
Plant J ; 75(2): 290-308, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23379770

RESUMEN

Viruses use and subvert host cell mechanisms to support their replication and spread between cells, tissues and organisms. Microtubules and associated motor proteins play important roles in these processes in animal systems, and may also play a role in plants. Although transport processes in plants are mostly actin based, studies, in particular with Tobacco mosaic virus (TMV) and its movement protein (MP), indicate direct or indirect roles of microtubules in the cell-to-cell spread of infection. Detailed observations suggest that microtubules participate in the cortical anchorage of viral replication complexes, in guiding their trafficking along the endoplasmic reticulum (ER)/actin network, and also in developing the complexes into virus factories. Microtubules also play a role in the plant-to-plant transmission of Cauliflower mosaic virus (CaMV) by assisting in the development of specific virus-induced inclusions that facilitate viral uptake by aphids. The involvement of microtubules in the formation of virus factories and of other virus-induced inclusions suggests the existence of aggresomal pathways by which plant cells recruit membranes and proteins into localized macromolecular assemblies. Although studies related to the involvement of microtubules in the interaction of viruses with plants focus on specific virus models, a number of observations with other virus species suggest that microtubules may have a widespread role in viral pathogenesis.


Asunto(s)
Microtúbulos/virología , Virus de Plantas/fisiología , Replicación Viral , Animales , Caulimovirus/fisiología , Citoesqueleto/virología , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Insectos/virología , Enfermedades de las Plantas/virología , Proteínas de Movimiento Viral en Plantas/metabolismo , Virus de Plantas/patogenicidad , Virus del Mosaico del Tabaco/patogenicidad , Virus del Mosaico del Tabaco/fisiología
11.
J Immunol ; 188(3): 1222-33, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22227566

RESUMEN

Recruitment of virus-infected T lymphocytes into the CNS is an essential step in the development of virus-associated neuroinflammatory diseases, notably myelopathy induced by retrovirus human T leukemia virus-1 (HTLV-1). We have recently shown the key role of collapsin response mediator protein 2 (CRMP2), a phosphoprotein involved in cytoskeleton rearrangement, in the control of human lymphocyte migration and in brain targeting in animal models of virus-induced neuroinflammation. Using lymphocytes cloned from infected patients and chronically infected T cells, we found that HTLV-1 affects CRMP2 activity, resulting in an increased migratory potential. Elevated CRMP2 expression accompanies a higher phosphorylation level of CRMP2 and its more pronounced adhesion to tubulin and actin. CRMP2 forms, a full length and a shorter, cleaved one, are also affected. Tax transfection and extinction strategies show the involvement of this viral protein in enhanced full-length and active CRMP2, resulting in prominent migratory rate. A role for other viral proteins in CRMP2 phosphorylation is suspected. Full-length CRMP2 confers a migratory advantage possibly by preempting the negative effect of short CRMP2 we observe on T lymphocyte migration. In addition, HTLV-1-induced migration seems, in part, supported by the ability of infected cell to increase the proteosomal degradation of short CRMP2. Finally, gene expression in CD69(+) cells selected from patients suggests that HTLV-1 has the capacity to influence the CRMP2/PI3K/Akt axis thus to positively control cytoskeleton organization and lymphocyte migration. Our data provide an additional clue to understanding the infiltration of HTLV-1-infected lymphocytes into various tissues and suggest that the regulation of CRMP2 activity by virus infection is a novel aspect of neuroinflammation.


Asunto(s)
Movimiento Celular , Virus Linfotrópico T Tipo 1 Humano/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Linfocitos T/virología , Antígenos CD , Antígenos de Diferenciación de Linfocitos T , Citoesqueleto/virología , Humanos , Inflamación/virología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Lectinas Tipo C , Proteínas del Tejido Nervioso/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Linfocitos T/fisiología , Proteínas Virales
12.
J Immunol ; 189(2): 689-700, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22689882

RESUMEN

EWI motif-containing protein 2 (EWI-2) is a member of the Ig superfamily that links tetraspanin-enriched microdomains to the actin cytoskeleton. We found that EWI-2 colocalizes with CD3 and CD81 at the central supramolecular activation cluster of the T cell immune synapse. Silencing of the endogenous expression or overexpression of a cytoplasmic truncated mutant of EWI-2 in T cells increases IL-2 secretion upon Ag stimulation. Mass spectrometry experiments of pull-downs with the C-term intracellular domain of EWI-2 revealed the specific association of EWI-2 with the actin-binding protein α-actinin; this association was regulated by PIP2. α-Actinin regulates the immune synapse formation and is required for efficient T cell activation. We extended these observations to virological synapses induced by HIV and found that silencing of either EWI-2 or α-actinin-4 increased cell infectivity. Our data suggest that the EWI-2-α-actinin complex is involved in the regulation of the actin cytoskeleton at T cell immune and virological synapses, providing a link between membrane microdomains and the formation of polarized membrane structures involved in T cell recognition.


Asunto(s)
Actinina/metabolismo , Antígenos CD/metabolismo , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Sinapsis Inmunológicas/metabolismo , Sinapsis Inmunológicas/virología , Proteínas de la Membrana/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/virología , Actinina/fisiología , Secuencia de Aminoácidos , Presentación de Antígeno/inmunología , Antígenos CD/fisiología , Línea Celular Transformada , Citoesqueleto/inmunología , Citoesqueleto/patología , Citoesqueleto/virología , Infecciones por VIH/patología , VIH-1/inmunología , Humanos , Sinapsis Inmunológicas/patología , Células Jurkat , Activación de Linfocitos/inmunología , Microdominios de Membrana/inmunología , Microdominios de Membrana/patología , Microdominios de Membrana/virología , Proteínas de la Membrana/fisiología , Datos de Secuencia Molecular , Subgrupos de Linfocitos T/patología , Células Tumorales Cultivadas
13.
Viruses ; 16(5)2024 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-38793550

RESUMEN

Rotavirus (RV) replicates within viroplasms, membraneless electron-dense globular cytosolic inclusions with liquid-liquid phase properties. In these structures occur the virus transcription, replication, and packaging of the virus genome in newly assembled double-layered particles. The viroplasms are composed of virus proteins (NSP2, NSP5, NSP4, VP1, VP2, VP3, and VP6), single- and double-stranded virus RNAs, and host components such as microtubules, perilipin-1, and chaperonins. The formation, coalescence, maintenance, and perinuclear localization of viroplasms rely on their association with the cytoskeleton. A stabilized microtubule network involving microtubules and kinesin Eg5 and dynein molecular motors is associated with NSP5, NSP2, and VP2, facilitating dynamic processes such as viroplasm coalescence and perinuclear localization. Key post-translation modifications, particularly phosphorylation events of RV proteins NSP5 and NSP2, play pivotal roles in orchestrating these interactions. Actin filaments also contribute, triggering the formation of the viroplasms through the association of soluble cytosolic VP4 with actin and the molecular motor myosin. This review explores the evolving understanding of RV replication, emphasizing the host requirements essential for viroplasm formation and highlighting their dynamic interplay within the host cell.


Asunto(s)
Citoesqueleto , Rotavirus , Replicación Viral , Rotavirus/fisiología , Rotavirus/metabolismo , Rotavirus/genética , Citoesqueleto/metabolismo , Citoesqueleto/virología , Humanos , Animales , Microtúbulos/metabolismo , Microtúbulos/virología , Proteínas Virales/metabolismo , Proteínas Virales/genética , Interacciones Huésped-Patógeno , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Compartimentos de Replicación Viral/metabolismo , Infecciones por Rotavirus/virología , ARN Viral/genética , ARN Viral/metabolismo
14.
Traffic ; 12(12): 1702-13, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21917091

RESUMEN

Dendritic cells (DCs) capture human immunodeficiency virus (HIV) through a non-fusogenic mechanism that enables viral transmission to CD4(+) T cells, contributing to in vivo viral dissemination. Although previous studies have provided important clues to cell-free viral capture by mature DCs (mDCs), dynamic and kinetic insight on this process is still missing. Here, we used three-dimensional video microscopy and single-particle tracking approaches to dynamically dissect both cell-free and cell-associated viral capture by living mDCs. We show that cell-free virus capture by mDCs operates through three sequential phases: virus binding through specific determinants expressed in the viral particle, polarized or directional movements toward concrete regions of the cell membrane and virus accumulation in a sac-like structure where trapped viral particles display a hindered diffusive behavior. Moreover, real-time imaging of cell-associated viral transfer to mDCs showed a similar dynamics to that exhibited by cell-free virus endocytosis leading to viral accumulation in compartments. However, cell-associated HIV type 1 transfer to mDCs was the most effective pathway, boosted throughout enhanced cellular contacts with infected CD4(+) T cells. Our results suggest that in lymphoid tissues, mDC viral uptake could occur either by encountering cell-free or cell-associated virus produced by infected cells generating the perfect scenario to promote HIV pathogenesis and impact disease progression.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/virología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/inmunología , Virión/inmunología , Transporte Biológico , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Membrana Celular/inmunología , Membrana Celular/virología , Sistema Libre de Células , Células Cultivadas , Citoesqueleto/inmunología , Citoesqueleto/virología , Endocitosis/inmunología , Humanos , Imagenología Tridimensional/métodos , Microscopía por Video/métodos , Acoplamiento Viral
15.
Gene Ther ; 20(3): 308-17, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22622241

RESUMEN

We investigated the transduction of HEK293T cells permissive to adeno-associated virus serotype 8 (AAV8) to understand the mechanisms underlying its endocytic processing. Results showed that AAV8 enters cells through clathrin-mediated endocytosis followed by trafficking through various endosomal compartments. Interestingly, compared to the relatively well-characterized AAV2, a distinct involvement of late endosomes was observed for AAV8 trafficking within the target cell. AAV8 particles were also shown to exploit the cytoskeleton network to facilitate their transport within cells. Moreover, the cellular factors involved during endosomal escape were examined by an in vitro membrane permeabilization assay. Our data demonstrated that an acidic endosomal environment was required for AAV2 penetration through endosomal membranes and that the cellular endoprotease furin could promote AAV2 escape from the early endosomes. In contrast, these factors were not sufficient for AAV8 penetration through endosomal membranes. We further found that the ubiquitin-proteasome system is likely involved in the intracellular transport of AAV8 to nucleus. Taken together, our data have shed some light on the intracellular trafficking pathways of AAV8, which, in turn, could provide insight for potentializing AAV-mediated gene delivery.


Asunto(s)
Clatrina/metabolismo , Dependovirus/metabolismo , Endocitosis , Vectores Genéticos/farmacocinética , Transporte Biológico , Núcleo Celular/metabolismo , Núcleo Celular/virología , Citoesqueleto/metabolismo , Citoesqueleto/virología , Dependovirus/clasificación , Dependovirus/genética , Endosomas/metabolismo , Endosomas/virología , Furina/metabolismo , Vectores Genéticos/genética , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Microscopía Confocal , Microtúbulos/metabolismo , Microtúbulos/virología , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal , Especificidad de la Especie , Transducción Genética/métodos , Ubiquitina/metabolismo , Red trans-Golgi/metabolismo , Red trans-Golgi/virología
16.
Postepy Hig Med Dosw (Online) ; 66: 810-7, 2012 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-23175336

RESUMEN

Cytoskeleton, composed of actin filaments, microtubules and intermediate filaments, regulates many processes in the cell, e.g. intracellular transport. Actin and microtubules are polarized structures, along which bidirectional transport of motor proteins occurs: myosins along actin and the dynein/dynactin complex and kinesins along microtubules. Viruses interact with the cytoskeleton and motor proteins at different stages during their replication cycle. When entering and egressing the cell, viruses must penetrate the cortical layer of microfilaments, which usually takes place with the contribution of myosin. In the cytoplasm, retrograde transport involving dynein is used to move viruses to the microtubule organizing center. After replication, kinesins participate in anterograde transport of newly produced virions to the peripheral region, close to the plasma membrane. Some families of viruses have developed alternate routes of intracellular transport. The aim of this study is to describe the interactions between virus and cytoskeletal motor proteins and to determine their role in viral infection according to the current literature data.  


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/virología , Proteínas Motoras Moleculares/metabolismo , Virosis/metabolismo , Virus/patogenicidad , Citoesqueleto de Actina , Actinas/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Citoplasma/metabolismo , Humanos , Cinesinas/metabolismo , Centro Organizador de los Microtúbulos/metabolismo , Microtúbulos/metabolismo , Microtúbulos/fisiología , Miosinas/metabolismo
17.
Microbiol Spectr ; 10(1): e0226521, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35138149

RESUMEN

Avian hepatitis E virus (HEV) causes liver diseases and multiple extrahepatic disorders in chickens. However, the mechanisms involved in avian HEV entry remain elusive. Herein, we identified the RAS-related protein 1b (Rap1b) as a potential HEV-ORF2 protein interacting candidate. Experimental infection of chickens and cells with an avian HEV isolate from China (CaHEV) led to upregulated expression and activation of Rap1b both in vivo and in vitro. By using CaHEV capsid as mimic of virion to treat cell in vitro, it appears that the interaction between the viral capsid and Rap1b promoted cell membrane recruitment of the downstream effector Rap1-interacting molecule (RIAM). In turn, RIAM further enhanced Talin-1 membrane recruitment and retention, which led to the activation of integrin α5/ß1, as well as integrin-associated membrane protein kinases, including focal adhesion kinase (FAK). Meanwhile, FAK activation triggered activation of downstream signaling molecules, such as Ras-related C3 botulinum toxin substrate 1 RAC1 cell division cycle 42 (CDC42), p21-activated kinase 1 (PAK1), and LIM domain kinase 1 (LIMK1). Finally, F-actin rearrangement induced by Cofilin led to the formation of lamellipodia, filopodia, and stress fibers, contributes to plasma membrane remodeling, and might enhance CaHEV virion internalization. In conclusion, our data suggested that Rap1b activation was triggered during CaHEV infection and appeared to require interaction between CaHEV-ORF2 and Rap1b, thereby further inducing membrane recruitment of Talin-1. Membrane-bound Talin-1 then activates key Integrin-FAK-Cofilin cascades involved in modulation of actin kinetics, and finally leads to F-actin rearrangement and membrane remodeling to potentially facilitate internalization of CaHEV virions into permissive cells. IMPORTANCE Rap1b is a multifunctional protein that is responsible for cell adhesion, growth, and differentiation. The inactive form of Rap1b is phosphorylated and distributed in the cytoplasm, while active Rap1b is prenylated and loaded with GTP to the cell membrane. In this study, the activation of Rap1b was induced during the early stage of avian HEV infection under the regulation of PKA and SmgGDS. Continuously activated Rap1b recruited its effector RIAM to the membrane, thereby inducing the membrane recruitment of Talin-1 that led to the activation of membrane α5/ß1 integrins. The triggering of the signaling pathway-associated Integrin α5/ß1-FAK-CDC42&RAC1-PAK1-LIMK1-Cofilin culminated in F-actin polymerization and membrane remodeling that might promote avian HEV virion internalization. These findings suggested a novel mechanism that is potentially utilized by avian HEV to invade susceptible cells.


Asunto(s)
Citoesqueleto/metabolismo , Hepatitis Viral Animal/metabolismo , Hepevirus/patogenicidad , Enfermedades de las Aves de Corral/metabolismo , Proteínas Virales/metabolismo , Internalización del Virus , Proteínas de Unión al GTP rap/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Pollos , Citoesqueleto/genética , Citoesqueleto/virología , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/virología , Hepevirus/genética , Interacciones Huésped-Patógeno , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/virología , Unión Proteica , Proteínas Virales/genética , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rap/genética
18.
PLoS Pathog ; 5(10): e1000623, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19834551

RESUMEN

Silencing of T cell activation and function is a highly efficient strategy of immunosuppression induced by pathogens. By promoting formation of membrane microdomains essential for clustering of receptors and signalling platforms in the plasma membrane, ceramides accumulating as a result of membrane sphingomyelin breakdown are not only essential for assembly of signalling complexes and pathogen entry, but also act as signalling modulators, e. g. by regulating relay of phosphatidyl-inositol-3-kinase (PI3K) signalling. Their role in T lymphocyte functions has not been addressed as yet. We now show that measles virus (MV), which interacts with the surface of T cells and thereby efficiently interferes with stimulated dynamic reorganisation of their actin cytoskeleton, causes ceramide accumulation in human T cells in a neutral (NSM) and acid (ASM) sphingomyelinase-dependent manner. Ceramides induced by MV, but also bacterial sphingomyelinase, efficiently interfered with formation of membrane protrusions and T cell spreading and front/rear polarisation in response to beta1 integrin ligation or alphaCD3/CD28 activation, and this was rescued upon pharmacological or genetic ablation of ASM/NSM activity. Moreover, membrane ceramide accumulation downmodulated chemokine-induced T cell motility on fibronectin. Altogether, these findings highlight an as yet unrecognised concept of pathogens able to cause membrane ceramide accumulation to target essential processes in T cell activation and function by preventing stimulated actin cytoskeletal dynamics.


Asunto(s)
Membrana Celular/metabolismo , Ceramidas/metabolismo , Citoesqueleto/metabolismo , Tolerancia Inmunológica , Linfocitos T/virología , Animales , Células Cultivadas , Citoesqueleto/virología , Humanos , Tolerancia Inmunológica/inmunología , Células Jurkat , Activación de Linfocitos/inmunología , Virus del Sarampión/inmunología , Virus del Sarampión/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/fisiología , Linfocitos T/inmunología , Linfocitos T/metabolismo
19.
PLoS Pathog ; 5(7): e1000512, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19593382

RESUMEN

The role of actin dynamics in clathrin-mediated endocytosis in mammalian cells is unclear. In this study, we define the role of actin cytoskeleton in Kaposi's sarcoma-associated herpesvirus (KSHV) entry and trafficking in endothelial cells using an immunofluorescence-based assay to visualize viral capsids and the associated cellular components. In contrast to infectivity or reporter assays, this method does not rely on the expression of any viral and reporter genes, but instead directly tracks the accumulation of individual viral particles at the nuclear membrane as an indicator of successful viral entry and trafficking in cells. Inhibitors of endosomal acidification reduced both the percentage of nuclei with viral particles and the total number of viral particles docking at the perinuclear region, indicating endocytosis, rather than plasma membrane fusion, as the primary route for KSHV entry into endothelial cells. Accordingly, a viral envelope protein was only detected on internalized KSHV particles at the early but not late stage of infection. Inhibitors of clathrin- but not caveolae/lipid raft-mediated endocytosis blocked KSHV entry, indicating that clathrin-mediated endocytosis is the major route of KSHV entry into endothelial cells. KSHV particles were colocalized not only with markers of early and recycling endosomes, and lysosomes, but also with actin filaments at the early time points of infection. Consistent with these observations, transferrin, which enters cells by clathrin-mediated endocytosis, was found to be associated with actin filaments together with early and recycling endosomes, and to a lesser degree, with late endosomes and lysosomes. KSHV infection induced dynamic actin cytoskeleton rearrangements. Disruption of the actin cytoskeleton and inhibition of regulators of actin nucleation such as Rho GTPases and Arp2/3 complex profoundly blocked KSHV entry and trafficking. Together, these results indicate an important role for actin dynamics in the internalization and endosomal sorting/trafficking of KSHV and clathrin-mediated endocytosis in endothelial cells.


Asunto(s)
Actinas/metabolismo , Endosomas/metabolismo , Herpesvirus Humano 8/fisiología , Transporte de Proteínas/fisiología , Internalización del Virus , Actinas/antagonistas & inhibidores , Transporte Activo de Núcleo Celular , Caveolas/metabolismo , Línea Celular , Núcleo Celular/virología , Clorpromazina/farmacología , Vesículas Cubiertas por Clatrina/metabolismo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Citoesqueleto/virología , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Endosomas/virología , Células Endoteliales/metabolismo , Inhibidores Enzimáticos/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Microdominios de Membrana/metabolismo , Microscopía Fluorescente , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal , Transferrina/metabolismo , Proteínas Virales/metabolismo
20.
Curr Opin Cell Biol ; 13(1): 97-105, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11163140

RESUMEN

In the past decade, studies into the way in which intracellular bacterial pathogens hijack and subvert their hosts have provided many important insights into regulation of the actin cytoskeleton and cell motility, in addition to increasing our understanding of the infection process. Viral pathogens, however, may ultimately unlock more cellular secrets as they are even more dependent on their hosts during their life cycle.


Asunto(s)
Citoesqueleto/virología , Fenómenos Fisiológicos de los Virus , Virus/patogenicidad , Animales , Transporte Biológico Activo , Células HeLa , Humanos , Virosis/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA