Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
PLoS Pathog ; 13(6): e1006455, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28628648

RESUMEN

The Adenovirus (Ad) genome within the capsid is tightly associated with a virus-encoded, histone-like core protein-protein VII. Two other Ad core proteins, V and X/µ, also are located within the virion and are loosely associated with viral DNA. Core protein VII remains associated with the Ad genome during the early phase of infection. It is not known if naked Ad DNA is packaged into the capsid, as with dsDNA bacteriophage and herpesviruses, followed by the encapsidation of viral core proteins, or if a unique packaging mechanism exists with Ad where a DNA-protein complex is simultaneously packaged into the virion. The latter model would require an entirely new molecular mechanism for packaging compared to known viral packaging motors. We characterized a virus with a conditional knockout of core protein VII. Remarkably, virus particles were assembled efficiently in the absence of protein VII. No changes in protein composition were evident with VII-virus particles, including the abundance of core protein V, but changes in the proteolytic processing of some capsid proteins were evident. Virus particles that lack protein VII enter the cell, but incoming virions did not escape efficiently from endosomes. This greatly diminished all subsequent aspects of the infectious cycle. These results reveal that the Ad major core protein VII is not required to condense viral DNA within the capsid, but rather plays an unexpected role during virus maturation and the early stages of infection. These results establish a new paradigm pertaining to the Ad assembly mechanism and reveal a new and important role of protein VII in early stages of infection.


Asunto(s)
Infecciones por Adenoviridae/virología , Adenoviridae/fisiología , Proteínas del Núcleo Viral/metabolismo , Ensamble de Virus , Adenoviridae/genética , Cápside/metabolismo , Genoma Viral , Humanos , Proteínas del Núcleo Viral/genética , Replicación Viral
2.
J Virol ; 87(22): 12367-79, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24027314

RESUMEN

Endocytosis is the most prevalent entry port for viruses into cells, but viruses must escape from the lumen of endosomes to ensure that viral genomes reach a site for replication and progeny formation. Endosomal escape also helps viruses bypass endolysosomal degradation and presentation to certain Toll-like intrinsic immunity receptors. The mechanisms for cytosolic delivery of nonenveloped viruses or nucleocapsids from enveloped viruses are poorly understood, in part because no quantitative assays are readily available which directly measure the penetration of viruses into the cytosol. Following uptake by clathrin-mediated endocytosis or macropinocytosis, the nonenveloped adenoviruses penetrate from endosomes to the cytosol, and they traffic with cellular motors on microtubules to the nucleus for replication. In this report, we present a novel single-cell imaging assay which quantitatively measures individual cytosolic viruses and distinguishes them from endosomal viruses or viruses at the plasma membrane. Using this assay, we showed that the penetration of human adenoviruses of the species C and B occurs rapidly after virus uptake. Efficient penetration does not require acidic pH in endosomes. This assay is versatile and can be adapted to other adenoviruses and members of other nonenveloped and enveloped virus families.


Asunto(s)
Infecciones por Adenoviridae/virología , Adenoviridae/fisiología , Bioensayo , Proteínas de la Cápside/metabolismo , Membrana Celular/virología , Endosomas/virología , Internalización del Virus , Infecciones por Adenoviridae/metabolismo , Membrana Celular/metabolismo , Clatrina/metabolismo , Citosol/metabolismo , Citosol/virología , Endocitosis , Endosomas/metabolismo , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Receptores Virales/metabolismo
3.
EMBO J ; 27(7): 956-69, 2008 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-18323776

RESUMEN

Endocytosis supports cell communication, growth, and pathogen infection. The species B human adenovirus serotype 3 (Ad3) is associated with epidemic conjunctivitis, and fatal respiratory and systemic disease. Here we show that Ad3 uses dynamin-independent endocytosis for rapid infectious entry into epithelial and haematopoietic cells. Unlike Ad5, which uses dynamin-dependent endocytosis, Ad3 endocytosis spatially and temporally coincided with enhanced fluid-phase uptake. It was sensitive to macropinocytosis inhibitors targeting F-actin, protein kinase C, the sodium-proton exchanger, and Rac1 but not Cdc42. Infectious Ad3 macropinocytosis required viral activation of p21-activated kinase 1 (PAK1) and the C-terminal binding protein 1 of E1A (CtBP1), recruited to macropinosomes. These macropinosomes also contained the Ad3 receptors CD46 and alpha v integrins. CtBP1 is a phosphorylation target of PAK1, and is bifunctionally involved in membrane traffic and transcriptional repression of cell cycle, cancer, and innate immunity pathways. Phosphorylation-defective S147A-CtBP1 blocked Ad3 but not Ad5 infection, providing a direct link between PAK1 and CtBP1. The data show that viruses induce macropinocytosis for infectious entry, a pathway used in antigen presentation and cell migration.


Asunto(s)
Adenovirus Humanos/clasificación , Adenovirus Humanos/metabolismo , Oxidorreductasas de Alcohol/metabolismo , Proteínas de Unión al ADN/metabolismo , Pinocitosis , Actinas/metabolismo , Infecciones por Adenoviridae/virología , Adenovirus Humanos/ultraestructura , Clatrina/metabolismo , Dinaminas/metabolismo , Endosomas/enzimología , Endosomas/ultraestructura , Endosomas/virología , Activación Enzimática , Células HeLa , Sistema Hematopoyético/citología , Sistema Hematopoyético/virología , Humanos , Integrinas/metabolismo , Células K562 , Modelos Biológicos , Proteína Quinasa C/metabolismo , Serotipificación , Internalización del Virus , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rac1/metabolismo
4.
J Virol ; 84(10): 5336-50, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20237079

RESUMEN

Human adenovirus serotype 35 (HAdV-35; here referred to as Ad35) causes kidney and urinary tract infections and infects respiratory organs of immunocompromised individuals. Unlike other adenoviruses, Ad35 has a low seroprevalence, which makes Ad35-based vectors promising candidates for gene therapy. Ad35 utilizes CD46 and integrins as receptors for infection of epithelial and hematopoietic cells. Here we show that infectious entry of Ad35 into HeLa cells, human kidney HK-2 cells, and normal human lung fibroblasts strongly depended on CD46 and integrins but not heparan sulfate and variably required the large GTPase dynamin. Ad35 infections were independent of expression of the carboxy-terminal domain of AP180, which effectively blocks clathrin-mediated uptake. Ad35 infections were inhibited by small chemicals against serine/threonine kinase Pak1 (p21-activated kinase), protein kinase C (PKC), sodium-proton exchangers, actin, and acidic organelles. Remarkably, the F-actin inhibitor jasplakinolide, the Pak1 inhibitor IPA-3, or the sodium-proton exchange inhibitor 5-(N-ethyl-N-isopropyl) amiloride (EIPA) blocked endocytic uptake of Ad35. Dominant-negative proteins or small interfering RNAs against factors driving macropinocytosis, including the small GTPase Rac1, Pak1, or the Pak1 effector C-terminal binding protein 1 (CtBP1), potently inhibited Ad35 infection. Confocal laser scanning microscopy, electron microscopy, and live cell imaging showed that Ad35 colocalized with fluid-phase markers in large endocytic structures that were positive for CD46, alphanu integrins, and also CtBP1. Our results extend earlier observations with HAdV-3 (Ad3) and establish macropinocytosis as an infectious pathway for species B human adenoviruses in epithelial and hematopoietic cells.


Asunto(s)
Adenovirus Humanos/fisiología , Células Epiteliales/virología , Pinocitosis , Internalización del Virus , Línea Celular , Fibroblastos/virología , Humanos , Integrinas/fisiología , Proteína Cofactora de Membrana/fisiología , Receptores Virales/fisiología
5.
Traffic ; 9(12): 2265-78, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18980614

RESUMEN

Receptor-mediated endocytosis is a major gate for pathogens into cells. In this study, we analyzed the trafficking of human adenovirus type 2 and 5 (Ad2/5) and the escape-defective temperature-sensitive Ad2-ts1 mutant in epithelial cancer cells. Ad2/5 and Ad2-ts1 uptake into endosomes containing transferrin, major histocompatibility antigen 1 and the Rab5 effector early endosome antigen 1 (EEA1) involved dynamin, amphiphysin, clathrin and Eps15. Cointernalization experiments showed that most of the Ad2/5 and Ad2-ts1 visited the same EEA1-positive endosomes. In contrast to Ad2/5, Ad2-ts1 required functional Rab5 for endocytosis and lysosomal transport and was sensitive to the phosphatidyl-inositol-3 (PI3)-kinase inhibitor wortmannin or the ubiquitin-binding protein Hrs for sorting from early to late endosomes. Endosomal escape of Ad2 was not affected by incubation at 19 degrees C, which blocked membrane sorting in early endosomes and inhibited Ad2-ts1 transport to lysosomes. Unlike Semliki Forest Virus (SFV), sorting of Ad2-ts1 to late endosomes was independent of Rab7 and Ad2/5 infection independent of EEA1. The data indicate that Ad2/5 and Ad2-ts1 use an invariant machinery for clathrin-mediated uptake to early endosomes. We suggest that the infectious Ad2 particles are either directly released from early endosomes to the cytosol or sorted by a temperature-insensitive and PI3-kinase-independent mechanism to an escape compartment different from late endosomes or lysosomes.


Asunto(s)
Adenoviridae/fisiología , Adenoviridae/ultraestructura , Línea Celular Tumoral , Clatrina/metabolismo , Dinaminas/metabolismo , Endocitosis , Endosomas/enzimología , Endosomas/ultraestructura , Humanos , Microscopía Inmunoelectrónica , Fosfatidilinositol 3-Quinasas/metabolismo , Factores de Tiempo , Internalización del Virus , Proteínas de Unión al GTP rab5/metabolismo
6.
Environ Microbiol ; 12(5): 1243-59, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20148929

RESUMEN

The amoebae-resistant opportunistic pathogen Legionella pneumophila employs a biphasic life cycle to replicate in host cells and spread to new niches. Upon entering the stationary growth phase, the bacteria switch to a transmissive (virulent) state, which involves a complex regulatory network including the lqs gene cluster (lqsA-lqsR-hdeD-lqsS). LqsR is a putative response regulator that promotes host-pathogen interactions and represses replication. The autoinducer synthase LqsA catalyses the production of the diffusible signalling molecule 3-hydroxypentadecan-4-one (LAI-1) that is presumably recognized by the sensor kinase LqsS. Here, we analysed L. pneumophila strains lacking lqsA or lqsS. Compared with wild-type L. pneumophila, the DeltalqsS strain was more salt-resistant and impaired for the Icm/Dot type IV secretion system-dependent uptake by phagocytes. Legionella pneumophila strains lacking lqsS, lqsR or the alternative sigma factor rpoS sedimented more slowly and produced extracellular filaments. Deletion of lqsA moderately reduced the uptake of L. pneumophila by phagocytes, and the defect was complemented by expressing lqsA in trans. Unexpectedly, the overexpression of lqsA also restored the virulence defect and reduced filament production of L. pneumophila mutant strains lacking lqsS or lqsR, but not the phenotypes of strains lacking rpoS or icmT. These results suggest that LqsA products also signal through sensors not encoded by the lqs gene cluster. A transcriptome analysis of the DeltalqsA and DeltalqsS mutant strains revealed that under the conditions tested, lqsA regulated only few genes, whereas lqsS upregulated the expression of 93 genes at least twofold. These include 52 genes clustered in a 133 kb high plasticity genomic island, which is flanked by putative DNA-mobilizing genes and encodes multiple metal ion efflux pumps. Upon overexpression of lqsA, a cluster of 19 genes in the genomic island was also upregulated, suggesting that LqsA and LqsS participate in the same regulatory circuit.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Islas Genómicas/fisiología , Legionella pneumophila/fisiología , Fagocitos/microbiología , Proteínas Quinasas/metabolismo , Factores de Transcripción/metabolismo , Acanthamoeba castellanii/crecimiento & desarrollo , Acanthamoeba castellanii/microbiología , Animales , Proteínas Bacterianas/genética , Línea Celular , Perfilación de la Expresión Génica , Células HL-60 , Histidina Quinasa , Interacciones Huésped-Patógeno , Humanos , Filamentos Intermedios , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Legionella pneumophila/patogenicidad , Ratones , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Fagocitos/inmunología , Proteínas Quinasas/genética , Factores de Transcripción/genética , Virulencia
7.
J Cell Biol ; 158(6): 1119-31, 2002 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-12221069

RESUMEN

Adenovirus type 2 (Ad2) binds the coxsackie B virus Ad receptor and is endocytosed upon activation of the alphav integrin coreceptors. Here, we demonstrate that expression of dominant negative clathrin hub, eps15, or K44A-dynamin (dyn) inhibited Ad2 uptake into epithelial cells, indicating clathrin-dependent viral endocytosis. Surprisingly, Ad strongly stimulated the endocytic uptake of fluid phase tracers, coincident with virus internalization but without affecting receptor-mediated transferrin uptake. A large amount of the stimulated endocytic activity was macropinocytosis. Macropinocytosis depended on alphav integrins, PKC, F-actin, and the amiloride-sensitive Na+/H+ exchanger, which are all required for Ad escape from endosomes and infection. Macropinocytosis stimulation was not a consequence of viral escape, since it occurred in K44A-dyn-expressing cells. Surprisingly, 30-50% of the endosomal contents were released into the cytosol of control and also K44A-dyn-expressing cells, and the number of fluid phase-positive endosomes dropped below the levels of noninfected cells, indicating macropinosomal lysis. The release of macropinosomal contents was Ad dose dependent, but the presence of Ad particles on macropinosomal membranes was not sufficient for contents release. We conclude that Ad signaling from the cell surface controls the induction of macropinosome formation and leakage, and this correlates with viral exit to the cytosol and infection.


Asunto(s)
Adenoviridae/metabolismo , Adenoviridae/patogenicidad , Clatrina/fisiología , Endosomas/metabolismo , Pinocitosis , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Adenoviridae/ultraestructura , Infecciones por Adenovirus Humanos/virología , Proteínas de Unión al Calcio/metabolismo , Invaginaciones Cubiertas de la Membrana Celular/ultraestructura , Vesículas Cubiertas/ultraestructura , Citosol/virología , Dinaminas , Células Epiteliales/virología , GTP Fosfohidrolasas/metabolismo , Células HeLa , Humanos , Integrinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Fosfoproteínas/metabolismo , Proteína Quinasa C/metabolismo , Intercambiadores de Sodio-Hidrógeno/fisiología , Transferrina/metabolismo , Células Tumorales Cultivadas
8.
Cell Rep ; 29(12): 3785-3795.e8, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31851912

RESUMEN

Adenoviruses (AdVs) cause respiratory, ocular, and gastrointestinal tract infection and inflammation in immunocompetent people and life-threatening disease upon immunosuppression. AdV vectors are widely used in gene therapy and vaccination. Incoming particles attach to nuclear pore complexes (NPCs) of post-mitotic cells, then rupture and deliver viral DNA (vDNA) to the nucleus or misdeliver to the cytosol. Our genome-wide RNAi screen in AdV-infected cells identified the RING-type E3 ubiquitin ligase Mind bomb 1 (Mib1) as a proviral host factor for AdV infection. Mib1 is implicated in Notch-Delta signaling, ciliary biogenesis, and RNA innate immunity. Mib1 depletion arrested incoming AdVs at NPCs. Induced expression of full-length but not ligase-defective Mib1 in knockout cells triggered vDNA uncoating from NPC-tethered virions, nuclear import, misdelivery of vDNA, and vDNA expression. Mib1 is an essential host factor for AdV uncoating in human cells, and it provides a new concept for licensing virion DNA delivery through the NPC.


Asunto(s)
Infecciones por Adenoviridae/virología , Adenoviridae/genética , Genoma Viral , Poro Nuclear/virología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Replicación Viral , Transporte Activo de Núcleo Celular , Adenoviridae/inmunología , Infecciones por Adenoviridae/genética , Infecciones por Adenoviridae/inmunología , ADN Viral/genética , Células HEK293 , Células HeLa , Humanos , Poro Nuclear/genética , Unión Proteica , Interferencia de ARN , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Virión
9.
Mol Biol Cell ; 16(6): 2999-3009, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15814838

RESUMEN

Incoming adenovirus type 2 (Ad2) and Ad5 shuttle bidirectionally along microtubules, biased to the microtubule-organizing center by the dynein/dynactin motor complex. It is unknown how the particles reach the nuclear pore complex, where capsids disassemble and viral DNA enters the nucleus. Here, we identified a novel link between nuclear export and microtubule-mediated transport. Two distinct inhibitors of the nuclear export factor CRM1, leptomycin B (LMB) and ratjadone A (RJA) or CRM1-siRNAs blocked adenovirus infection, arrested cytoplasmic transport of viral particles at the microtubule-organizing center or in the cytoplasm and prevented capsid disassembly and nuclear import of the viral genome. In mitotic cells where CRM1 is in the cytoplasm, adenovirus particles were not associated with microtubules but upon LMB treatment, they enriched at the spindle poles implying that CRM1 inhibited microtubule association of adenovirus. We propose that CRM1, a nuclear factor exported by CRM1 or a protein complex containing CRM1 is part of a sensor mechanism triggering the unloading of the incoming adenovirus particles from microtubules proximal to the nucleus of interphase cells.


Asunto(s)
Adenoviridae/metabolismo , Núcleo Celular/metabolismo , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transporte Activo de Núcleo Celular , Adenoviridae/ultraestructura , Animales , Western Blotting , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Endotelio Vascular/citología , Ácidos Grasos Insaturados/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Carioferinas/antagonistas & inhibidores , Microscopía Confocal , Centro Organizador de los Microtúbulos/efectos de los fármacos , Centro Organizador de los Microtúbulos/metabolismo , Mitosis , Pironas/farmacología , ARN Interferente Pequeño/farmacología , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Huso Acromático/virología , Transferrina/farmacocinética , Venas Umbilicales/citología , Proteína Exportina 1
10.
Cell Death Dis ; 9(3): 272, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29449668

RESUMEN

Apoptosis and programmed necrosis (necroptosis) determine cell fate, and antagonize infection. Execution of these complementary death pathways involves the formation of receptor-interacting protein kinase 1 (RIPK1) containing complexes. RIPK1 binds to adaptor proteins, such as TRIF (Toll-IL-1 receptor-domain-containing-adaptor-inducing interferon-beta factor), FADD (Fas-associated-protein with death domain), NEMO (NF-κB regulatory subunit IKKγ), SQSTM1 (sequestosome 1/p62), or RIPK3 (receptor-interacting protein kinase 3), which are involved in RNA sensing, NF-κB signaling, autophagosome formation, apoptosis, and necroptosis. We report that a range of rhinoviruses impair apoptosis and necroptosis in epithelial cells late in infection. Unlike the double-strand (ds) RNA mimetic poly I:C (polyinosinic:polycytidylic acid), the exposure of dsRNA to toll-like receptor 3 (TLR3) in rhinovirus-infected cells did not lead to apoptosis execution. Accordingly, necroptosis and the production of ROS (reactive oxygen species) were not observed late in infection, when RIPK3 was absent. Instead, a virus-induced alternative necrotic cell death pathway proceeded, which led to membrane rupture, indicated by propidium iodide staining. The impairment of dsRNA-induced apoptosis late in infection was controlled by the viral 3C-protease (3Cpro), which disrupted RIPK1-TRIF/FADD /SQSTM1 immune-complexes. 3Cpro and 3C precursors were found to coimmuno-precipitate with RIPK1, cleaving the RIPK1 death-domain, and generating N-terminal RIPK1 fragments. The depletion of RIPK1 or chemical inhibition of its kinase at the N-terminus did not interfere with virus progeny formation or cell fate. The data show that rhinoviruses suppress apoptosis and necroptosis, and release progeny by an alternative cell death pathway, which is controlled by viral proteases modifying innate immune complexes.


Asunto(s)
Apoptosis , Cisteína Endopeptidasas/metabolismo , Células Epiteliales/virología , Mucosa Nasal/virología , Necroptosis , Rhinovirus/enzimología , Neoplasias del Cuello Uterino/virología , Proteínas Virales/metabolismo , Proteasas Virales 3C , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Complejo Antígeno-Anticuerpo/metabolismo , Células Epiteliales/enzimología , Células Epiteliales/inmunología , Células Epiteliales/ultraestructura , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Femenino , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Mucosa Nasal/enzimología , Mucosa Nasal/inmunología , Mucosa Nasal/ultraestructura , Especies Reactivas de Oxígeno/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Rhinovirus/inmunología , Rhinovirus/patogenicidad , Proteína Sequestosoma-1/metabolismo , Transducción de Señal , Neoplasias del Cuello Uterino/enzimología , Neoplasias del Cuello Uterino/ultraestructura
11.
Nat Commun ; 9(1): 1980, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29773792

RESUMEN

The type I interferon (IFN) system plays an important role in controlling herpesvirus infections, but it is unclear which IFN-mediated effectors interfere with herpesvirus replication. Here we report that human myxovirus resistance protein B (MxB, also designated Mx2) is a potent human herpesvirus restriction factor in the context of IFN. We demonstrate that ectopic MxB expression restricts a range of herpesviruses from the Alphaherpesvirinae and Gammaherpesvirinae, including herpes simplex virus 1 and 2 (HSV-1 and HSV-2), and Kaposi's sarcoma-associated herpesvirus (KSHV). MxB restriction of HSV-1 and HSV-2 requires GTPase function, in contrast to restriction of lentiviruses. MxB inhibits the delivery of incoming HSV-1 DNA to the nucleus and the appearance of empty capsids, but not the capsid delivery to the cytoplasm or tegument dissociation from the capsid. Our study identifies MxB as a potent pan-herpesvirus restriction factor which blocks the uncoating of viral DNA from the incoming viral capsid.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Herpesviridae/fisiología , Interferón Tipo I/inmunología , Proteínas de Resistencia a Mixovirus/inmunología , Replicación Viral/inmunología , Cápside/inmunología , Proteínas de la Cápside/inmunología , Línea Celular Tumoral , Núcleo Celular/inmunología , Núcleo Celular/virología , Citoplasma , ADN Viral/inmunología , Células HEK293 , Herpesviridae/patogenicidad , Infecciones por Herpesviridae/virología , Humanos , Proteínas de Resistencia a Mixovirus/genética , ARN Interferente Pequeño/metabolismo , Desencapsidación Viral/inmunología
12.
Curr Biol ; 13(10): 828-32, 2003 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-12747830

RESUMEN

Photoreceptors (R cells) in the Drosophila retina connect to targets in three distinct layers of the optic lobe of the brain: R1-R6 connect to the lamina, and R7 and R8 connect to distinct layers in the medulla. In each of these layers, R axon termini are arranged in evenly spaced topographic arrays. In a genetic screen for mutants with abnormal R cell connectivity, we recovered mutations in flamingo (fmi). fmi encodes a seven-transmembrane cadherin, previously shown to function in planar cell polarity and in dendritic patterning. Here, we show that fmi has two specific functions in R8 axon targeting: it facilitates competitive interactions between adjacent R8 axons to ensure their correct spacing, and it promotes the formation of stable connections between R8 axons and their target cells in the medulla. The former suggests a general role for Fmi in establishing nonoverlapping dendritic and axonal target fields. The latter, together with the finding that N-Cadherin has an analogous role in R7 axon-target interactions, points to a cadherin-based system for target layer specificity in the Drosophila visual system.


Asunto(s)
Axones/fisiología , Cadherinas/metabolismo , Drosophila/fisiología , Células Fotorreceptoras de Invertebrados/metabolismo , Vías Visuales/metabolismo , Alelos , Animales , Axones/ultraestructura , Encéfalo/metabolismo , Encéfalo/ultraestructura , Cadherinas/genética , Drosophila/metabolismo , Drosophila/ultraestructura , Proteínas de Drosophila , Regulación de la Expresión Génica/fisiología , Conos de Crecimiento/metabolismo , Conos de Crecimiento/ultraestructura , Heterocigoto , Larva/crecimiento & desarrollo , Mosaicismo , Mutación , Células Fotorreceptoras de Invertebrados/crecimiento & desarrollo , Células Fotorreceptoras de Invertebrados/ultraestructura , Pupa/crecimiento & desarrollo , Retina/fisiología , Retina/ultraestructura , Vías Visuales/crecimiento & desarrollo , Vías Visuales/ultraestructura
13.
Cell Host Microbe ; 18(1): 75-85, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26159720

RESUMEN

During cell entry, non-enveloped viruses undergo partial uncoating to expose membrane lytic proteins for gaining access to the cytoplasm. We report that adenovirus uses membrane piercing to induce and hijack cellular wound removal processes that facilitate further membrane disruption and infection. Incoming adenovirus stimulates calcium influx and lysosomal exocytosis, a membrane repair mechanism resulting in release of acid sphingomyelinase (ASMase) and degradation of sphingomyelin to ceramide lipids in the plasma membrane. Lysosomal exocytosis is triggered by small plasma membrane lesions induced by the viral membrane lytic protein-VI, which is exposed upon mechanical cues from virus receptors, followed by virus endocytosis into leaky endosomes. Chemical inhibition or RNA interference of ASMase slows virus endocytosis, inhibits virus escape to the cytosol, and reduces infection. Ceramide enhances binding of protein-VI to lipid membranes and protein-VI-induced membrane rupture. Thus, adenovirus uses a positive feedback loop between virus uncoating and lipid signaling for efficient membrane penetration.


Asunto(s)
Adenoviridae/fisiología , Proteínas de la Cápside/metabolismo , Membrana Celular/fisiología , Interacciones Huésped-Patógeno , Internalización del Virus , Adenoviridae/enzimología , Biotransformación , Membrana Celular/metabolismo , Ceramidas/metabolismo , Endocitosis , Exocitosis , Células HeLa , Humanos , Lisosomas/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Esfingomielinas/metabolismo
14.
Nat Commun ; 2: 391, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21750545

RESUMEN

Mucosal epithelia provide strong barriers against pathogens. For instance, the outward facing apical membrane of polarized epithelial cells lacks receptors for agents, such as hepatitis C virus, herpesvirus, reovirus, poliovirus or adenovirus. In addition, macrophages eliminate pathogens from the luminal space. Here we show that human adenovirus type 5 engages an antiviral immune response to enter polarized epithelial cells. Blood-derived macrophages co-cultured apically on polarized epithelial cells facilitate epithelial infection. Infection also occurs in the absence of macrophages, if virus-conditioned macrophage-medium containing the chemotactic cytokine CXCL8 (interleukin-8), or recombinant CXCL8 are present. In polarized cells, CXCL8 activates a Src-family tyrosine kinase via the apical CXCR1 and CXCR2 receptors. This activation process relocates the viral co-receptor ανß3 integrin to the apical surface, and enables apical binding and infection with adenovirus depending on the primary adenovirus receptor CAR. This paradigm may explain how other mucosal pathogens enter epithelial cells.


Asunto(s)
Adenovirus Humanos/fisiología , Células Epiteliales/virología , Interleucina-8/inmunología , Internalización del Virus , Adenovirus Humanos/inmunología , Western Blotting , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Integrina alfaVbeta3/metabolismo , Interleucina-8/metabolismo , Macrófagos/inmunología , Microscopía Confocal
15.
Cell Host Microbe ; 10(2): 105-17, 2011 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-21843868

RESUMEN

Viral particle binding to plasma membrane receptors elicits virus motions, recruits signaling proteins, and triggers membrane bending and fission, finally resulting in endocytic virus uptake. Here we analyze how human adenovirus engages its receptor coxsackievirus adenovirus receptor (CAR) and coreceptor αv integrin to move on the plasma membrane. Virus binding to CAR through fiber knobs gave rise to diffusive motions and actomyosin-2-dependent drifts, while integrin-targeted viruses were spatially more confined. Diffusions, drifts, and confined motions were specifically observed with viral particles that were subsequently internalized. CAR-mediated drifts together with integrin binding supported fiber shedding from adenovirus particles, leading to exposure of the membrane-lytic internal virion protein VI and enhanced viral escape from endosomes. Our results show that adenovirus uncoating is initiated at the plasma membrane by CAR drifting motion and binding to immobile integrins.


Asunto(s)
Adenovirus Humanos/patogenicidad , Integrina alfaV/metabolismo , Receptores Virales/metabolismo , Desencapsidación Viral , Actinas/metabolismo , Actomiosina/antagonistas & inhibidores , Actomiosina/metabolismo , Adenovirus Humanos/metabolismo , Animales , Proteínas de la Cápside/metabolismo , Membrana Celular/metabolismo , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Microscopía por Crioelectrón , Endocitosis , Técnica del Anticuerpo Fluorescente , Células HeLa , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Ratones , Microscopía Confocal , Seudópodos/metabolismo , Acoplamiento Viral , Internalización del Virus , Liberación del Virus
16.
Cell Host Microbe ; 10(3): 210-23, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21925109

RESUMEN

Many viruses deliver their genomes into the host cell nucleus for replication. However, the size restrictions of the nuclear pore complex (NPC), which regulates the passage of proteins, nucleic acids, and solutes through the nuclear envelope, require virus capsid uncoating before viral DNA can access the nucleus. We report a microtubule motor kinesin-1-mediated and NPC-supported mechanism of adenovirus uncoating. The capsid binds to the NPC filament protein Nup214 and kinesin-1 light-chain Klc1/2. The nucleoporin Nup358, which is bound to Nup214/Nup88, interacts with the kinesin-1 heavy-chain Kif5c to indirectly link the capsid to the kinesin motor. Kinesin-1 disrupts capsids docked at Nup214, which compromises the NPC and dislocates nucleoporins and capsid fragments into the cytoplasm. NPC disruption increases nuclear envelope permeability as indicated by the nuclear influx of large cytoplasmic dextran polymers. Thus, kinesin-1 uncoats viral DNA and compromises NPC integrity, allowing viral genomes nuclear access to promote infection.


Asunto(s)
Infecciones por Adenoviridae/metabolismo , Adenoviridae/fisiología , Cápside/metabolismo , Cinesinas/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Poro Nuclear/metabolismo , Ensamble de Virus , Adenoviridae/genética , Infecciones por Adenoviridae/virología , Línea Celular , Células HeLa , Humanos , Cinesinas/genética , Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/genética , Unión Proteica
17.
Autophagy ; 5(7): 980-90, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19587536

RESUMEN

Autophagy is a lysosomal-mediated degradation process that promotes cell survival during nutrient-limiting conditions. However, excessive autophagy results in cell death. In Drosophila, autophagy is regulated nutritionally, hormonally and developmentally in several tissues, including the fat body, a nutrient-storage organ. Here we use a proteomics approach to identify components of starvation-induced autophagic responses in the Drosophila fat body. Using cICAT labeling and mass spectrometry, differences in protein expression levels of normal compared to starved fat bodies were determined. Candidates were analyzed genetically for their involvement in autophagy in fat bodies deficient for the respective genes. One of these genes, Desat1, encodes a lipid desaturase. Desat1 mutant cells fail to induce autophagy upon starvation. The desat1 protein localizes to autophagic structures after nutrient depletion and is required for fly development. Lipid analyses revealed that Desat1 regulates the composition of lipids in Drosophila. We propose that Desat1 exerts its role in autophagy by controlling lipid biosynthesis and/or signaling necessary for autophagic responses.


Asunto(s)
Autofagia/fisiología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Drosophila melanogaster/fisiología , Ácido Graso Desaturasas/metabolismo , Proteómica/métodos , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/anatomía & histología , Cuerpo Adiposo/citología , Cuerpo Adiposo/metabolismo , Ácido Graso Desaturasas/genética , Humanos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
18.
J Virol ; 79(4): 2604-13, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15681460

RESUMEN

Adenovirus type 2 (Ad2) and Ad5 enter epithelial cells via the coxsackievirus B Ad receptor (CAR) and alpha(v) integrin coreceptors. In the absence of CAR, they can be directed to the Fcgamma receptor 1 of hematopoietic cells by an adaptor comprising the extracellular CAR domain and the Fc portion of a human immunoglobulin G (CARex-Fc). This gives rise to Ad aggregates and single particles which together enhance gene delivery up to 250-fold compared to adaptor-less viruses. A small interfering RNA knockdown of the clathrin heavy chain and quantitative electron microscopy of hematopoietic leukemia cells showed that the majority of Ads were phagocytosed as clusters of 1 to 3 microm in diameter and that about 10% of the particles entered cells by clathrin-mediated endocytosis. The clathrin knockdown did not affect phagocytosis but, surprisingly, inhibited viral escape from phagosomes. Similarly, blocking an early stage of clathrin-coated pit assembly inhibited phagosomal escape and infection but not aggregate uptake, unlike blocking of a late stage of clathrin-coated pit formation. We propose a cooperative interaction of clathrin-mediated endocytosis and phagocytosis triggering phagosomal lysis and infection.


Asunto(s)
Adenovirus Humanos/fisiología , Endocitosis/fisiología , Receptores de IgG/fisiología , Receptores Virales/fisiología , Línea Celular , Clatrina/fisiología , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Humanos , Fagocitosis/fisiología
19.
J Virol ; 78(6): 3089-98, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14990728

RESUMEN

The species C adenovirus type 2 (Ad2) and Ad5 bind the coxsackievirus B Ad receptor and alphav integrin coreceptors and enter epithelial cells by clathrin-mediated endocytosis. This pathway is rapid and efficient. It leads to cell activation and the cholesterol-dependent formation of macropinosomes. Macropinosomes are triggered to release their contents when incoming Ad2 escapes from endosomes. Here, we show that cholesterol extraction of epithelial cells by methyl-beta-cyclodextrin (mbetaCD) treatment reduced Ad5-mediated luciferase expression approximately 4-fold. The addition of cholesterol to normal cells increased gene expression in a dose-dependent manner up to threefold, but it did not restore gene expression in mbetaCD-treated cells. mbetaCD had no effect in the presence of excess cholesterol, indicating that the inhibition of gene expression was due specifically to cholesterol depletion. Cholesterol depletion inhibited rapid Ad2 endocytosis, endosomal escape, and nuclear targeting, consistent with the notion that clathrin-dependent endocytosis of Ad2 is cholesterol dependent. In cholesterol-reduced cells, Ad2 internalized at a low rate, suggestive of an alternative, clathrin-independent, low-capacity entry pathway. While exogenous cholesterol completely restored rapid Ad2 endocytosis, macropinocytosis, and macropinosome disruption, it did not, surprisingly, restore viral escape from endosomes. Our results indicate that macropinosome disruption and endosomal escape of Ad2 are independent events in cells depleted of and then refilled with cholesterol, suggesting that viral escape from endosomes requires lipid-controlled membrane homeostasis, trafficking, or signaling.


Asunto(s)
Adenovirus Humanos/patogenicidad , Colesterol/metabolismo , Endocitosis , Endosomas/virología , Adenovirus Humanos/genética , Adenovirus Humanos/metabolismo , Citosol/virología , Células HeLa , Humanos , Microscopía Electrónica , Pinocitosis
20.
J Virol ; 78(9): 4454-62, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15078926

RESUMEN

Many human adenovirus (Ad) serotypes use the coxsackie B virus-Ad receptor (CAR). Recently, CD46 was suggested to be a receptor of species B Ad serotype 11 (Ad11), Ad14, Ad16, Ad21, Ad35, and Ad50. Using Sindbis virus-mediated cDNA library expression, we identify here the membrane cofactor protein CD46 as a surface receptor of species B Ad3. All four major CD46 transcripts and one minor CD46 transcript expressed in nucleated human cells were isolated. Rodent BHK cells stably expressing the BC1 form of CD46 bound radiolabeled Ad3 with a dissociation constant of 0.3 nM, identical to that of CD46-positive HeLa cells expressing twice as many Ad3 binding sites. Pull-down experiments with recombinant Ad3 fibers and a soluble form of the CD46 extracellular domain linked to the Fc portion of human immunoglobulin G (CD46ex-Fc) indicated direct interactions of the Ad3 fiber knob with CD46ex-Fc but not CARex-Fc (Fc-linked extracellular domain of CAR). Ad3 colocalized with cell surface CD46 in both rodent and human cells at the light and electron microscopy levels. Anti-CD46 antibodies and CD46ex-Fc inhibited Ad3 binding to CD46-expressing BHK cells more than 10-fold and to human cells 2-fold. In CD46-expressing BHK cells, wild-type Ad3 and a chimeric Ad consisting of the Ad5 capsid and the Ad3 fiber elicited dose-dependent cytopathic effects and transgene expression, albeit less efficiently than in human cells. Together, our results show that all of the major splice forms of CD46 are predominant and functional binding sites of Ad3 on CD46-expressing rodent and human cells but may not be the sole receptor of species B Ads on human cells. These results have implications for understanding viral pathogenesis and therapeutic gene delivery.


Asunto(s)
Adenovirus Humanos/metabolismo , Adenovirus Humanos/patogenicidad , Antígenos CD/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Virales/metabolismo , Adenovirus Humanos/clasificación , Adenovirus Humanos/genética , Animales , Antígenos CD/genética , Sitios de Unión , Proteínas de la Cápside/metabolismo , Línea Celular , Cricetinae , Efecto Citopatogénico Viral , Células HeLa , Humanos , Proteína Cofactora de Membrana , Glicoproteínas de Membrana/genética , Microscopía Confocal , Serotipificación , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA