Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Pediatr Hematol Oncol ; 45(8): e972-e975, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37669118

RESUMEN

Kaposi's sarcoma (KS) is a vascular / mesenchymal tumor with an indefinite degree of malignancy, caused by complex etiopathogenetic factors including Human Herpes Virus-8 infection of immunocompromised patients. For example, KS is more common in adult men with HIV. We describe 2 very rare cases of iatrogenic KS in children after hematopoietic stem cell transplant with isolated organ damage (case 1: lung; case 2: inguinal lymph node). KS is a potential complication of bone marrow transplant in pediatric patients and can occur in different age groups and at atypical sites.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Herpesvirus Humano 8 , Sarcoma de Kaposi , Niño , Humanos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Huésped Inmunocomprometido , Pulmón , Ganglios Linfáticos , Sarcoma de Kaposi/etiología
2.
PLoS Pathog ; 18(10): e1010906, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36306280

RESUMEN

As ultimate parasites, viruses depend on host factors for every step of their life cycle. On the other hand, cells evolved multiple mechanisms of detecting and interfering with viral replication. Yet, our understanding of the complex ensembles of pro- and anti-viral factors is very limited in virtually every virus-cell system. Here we investigated the proteins recruited to the replication organelles of poliovirus, a representative of the genus Enterovirus of the Picornaviridae family. We took advantage of a strict dependence of enterovirus replication on a host protein GBF1, and established a stable cell line expressing a truncated GBF1 fused to APEX2 peroxidase that effectively supported viral replication upon inhibition of the endogenous GBF1. This construct biotinylated multiple host and viral proteins on the replication organelles. Among the viral proteins, the polyprotein cleavage intermediates were overrepresented, suggesting that the GBF1 environment is linked to viral polyprotein processing. The proteomics characterization of biotinylated host proteins identified multiple proteins previously associated with enterovirus replication, as well as more than 200 new factors recruited to the replication organelles. RNA metabolism proteins, many of which normally localize in the nucleus, constituted the largest group, underscoring the massive release of nuclear factors into the cytoplasm of infected cells and their involvement in viral replication. Functional analysis of several newly identified proteins revealed both pro- and anti-viral factors, including a novel component of infection-induced stress granules. Depletion of these proteins similarly affected the replication of diverse enteroviruses indicating broad conservation of the replication mechanisms. Thus, our data significantly expand the knowledge of the composition of enterovirus replication organelles, provide new insights into viral replication, and offer a novel resource for identifying targets for anti-viral interventions.


Asunto(s)
Infecciones por Enterovirus , Enterovirus , Poliovirus , Humanos , Enterovirus/metabolismo , Biotinilación , Poliovirus/fisiología , Replicación Viral , Proteínas Virales/metabolismo , Poliproteínas/metabolismo , Antivirales/farmacología , Factores de Intercambio de Guanina Nucleótido/metabolismo
3.
Viruses ; 13(8)2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34452452

RESUMEN

The capsid precursor P1 constitutes the N-terminal part of the enterovirus polyprotein. It is processed into VP0, VP3, and VP1 by the viral proteases, and VP0 is cleaved autocatalytically into VP4 and VP2. We observed that poliovirus VP0 is recognized by an antibody against a cellular autophagy protein, LC3A. The LC3A-like epitope overlapped the VP4/VP2 cleavage site. Individually expressed VP0-EGFP and P1 strongly colocalized with a marker of selective autophagy, p62/SQSTM1. To assess the role of capsid proteins in autophagy development we infected different cells with poliovirus or encapsidated polio replicon coding for only the replication proteins. We analyzed the processing of LC3B and p62/SQSTM1, markers of the initiation and completion of the autophagy pathway and investigated the association of the viral antigens with these autophagy proteins in infected cells. We observed cell-type-specific development of autophagy upon infection and found that only the virion signal strongly colocalized with p62/SQSTM1 early in infection. Collectively, our data suggest that activation of autophagy is not required for replication, and that capsid proteins contain determinants targeting them to p62/SQSTM1-dependent sequestration. Such a strategy may control the level of capsid proteins so that viral RNAs are not removed from the replication/translation pool prematurely.


Asunto(s)
Autofagia , Proteínas de la Cápside/metabolismo , Interacciones Microbiota-Huesped , Poliovirus/química , Poliovirus/metabolismo , Proteínas Virales/metabolismo , Cápside/metabolismo , Proteínas de la Cápside/clasificación , Proteínas de la Cápside/genética , Células HEK293 , Células HeLa , Humanos , Poliovirus/genética , Procesamiento Proteico-Postraduccional , ARN Viral/metabolismo , Replicón , Proteínas Virales/genética , Virión/metabolismo
4.
Front Pediatr ; 8: 577, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33042920

RESUMEN

Objectives: Mutations in the neuroblastoma-amplified sequence (NBAS) gene were originally described in patients with skeletal dysplasia or isolated liver disease of variable severity. Subsequent publications reported a more complex phenotype. Among multisystemic clinical symptoms, we were particularly interested in the immunological consequences of the NBAS deficiency. Methods: Clinical and laboratory data of 3 patients ages 13, 6, and 5 in whom bi-allelic NBAS mutations had been detected via next-generation sequencing were characterized. Literature review of 23 publications describing 74 patients was performed. Results: We report three Russian patients with compound heterozygous mutations of the NBAS gene who had combined immunodeficiency characterized by hypogammaglobulinemia, low T-cells, and near-absent B-cells, along with liver disease, skeletal dysplasia, optic-nerve atrophy, and dysmorphic features. Analysis of the data of 74 previously reported patients who carried various NBAS mutations demonstrated that although the most severe form of liver disease seems to require disruption of the N-terminal or middle part of NBAS, mutations of variable localizations in the gene have been associated with some form of liver disease, as well as immunological disorders. Conclusions: NBAS deficiency has a broad phenotype, and referral to an immunologist should be made in order to screen for immunodeficiency.

5.
J Virol ; 93(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31375590

RESUMEN

The replication of many positive-strand RNA viruses [(+)RNA viruses] depends on the cellular protein GBF1, but its role in the replication process is not clear. In uninfected cells, GBF1 activates small GTPases of the Arf family and coordinates multiple steps of membrane metabolism, including functioning of the cellular secretory pathway. The nonstructural protein 3A of poliovirus and related viruses has been shown to directly interact with GBF1, likely mediating its recruitment to the replication complexes. Surprisingly, viral mutants with a severely reduced level of 3A-GBF1 interaction demonstrate minimal replication defects in cell culture. Here, we systematically investigated the conserved elements of GBF1 to understand which determinants are important to support poliovirus replication. We demonstrate that multiple GBF1 mutants inactive in cellular metabolism could still be fully functional in the replication complexes. Our results show that the Arf-activating property, but not the primary structure of the Sec7 domain, is indispensable for viral replication. They also suggest a redundant mechanism of recruitment of GBF1 to the replication sites, which is dependent not only on direct interaction of the protein with the viral protein 3A but also on determinants located in the noncatalytic C-terminal domains of GBF1. Such a double-targeting mechanism explains the previous observations of the remarkable tolerance of different levels of GBF1-3A interaction by the virus and likely constitutes an important element of the resilience of viral replication.IMPORTANCE Enteroviruses are a vast group of viruses associated with diverse human diseases, but only two of them could be controlled with vaccines, and effective antiviral therapeutics are lacking. Here, we investigated in detail the contribution of a cellular protein, GBF1, in the replication of poliovirus, a representative enterovirus. GBF1 supports the functioning of cellular membrane metabolism and is recruited to viral replication complexes upon infection. Our results demonstrate that the virus requires a limited subset of the normal GBF1 functions and reveal the elements of GBF1 essential to support viral replication under different conditions. Since diverse viruses often rely on the same cellular proteins for replication, understanding the mechanisms by which these proteins support infection is essential for the development of broad-spectrum antiviral therapeutics.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Poliovirus/fisiología , Replicación Viral , Factor 1 de Ribosilacion-ADP/metabolismo , Proteínas Activadoras de GTPasa/genética , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/genética , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Mutación , Poliomielitis/metabolismo , Poliomielitis/virología , Poliovirus/metabolismo , Unión Proteica , Dominios Proteicos , Proteínas del Núcleo Viral/metabolismo
6.
Am J Physiol Cell Physiol ; 310(6): C456-69, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26718629

RESUMEN

Members of the large Sec7 domain-containing Arf guanine nucleotide exchange factor (GEF) family have been shown to dimerize through their NH2-terminal dimerization and cyclophilin binding (DCB) and homology upstream of Sec7 (HUS) domains. However, the importance of dimerization in GEF localization and function has not been assessed. We generated a GBF1 mutant (91/130) in which two residues required for oligomerization (K91 and E130 within the DCB domain) were replaced with A and assessed the effects of these mutations on GBF1 localization and cellular functions. We show that 91/130 is compromised in oligomerization but that it targets to the Golgi in a manner indistinguishable from wild-type GBF1 and that it rapidly exchanges between the cytosolic and membrane-bound pools. The 91/130 mutant appears active as it integrates within the functional network at the Golgi, supports Arf activation and COPI recruitment, and sustains Golgi homeostasis and cargo secretion when provided as a sole copy of functional GBF1 in cells. In addition, like wild-type GBF1, the 91/130 mutant supports poliovirus RNA replication, a process requiring GBF1 but believed to be independent of GBF1 catalytic activity. However, oligomerization appears to stabilize GBF1 in cells, and the 91/130 mutant is degraded faster than the wild-type GBF1. Our data support a model in which oligomerization is not a key regulator of GBF1 activity but impacts its function by regulating the cellular levels of GBF1.


Asunto(s)
Aparato de Golgi/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Línea Celular Tumoral , Proteína Coat de Complejo I/metabolismo , Citosol/metabolismo , Células HeLa , Humanos , Membranas Intracelulares/metabolismo , Unión Proteica/fisiología , Proteolisis
7.
J Virol ; 88(19): 11091-107, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25008939

RESUMEN

UNLABELLED: Few drugs targeting picornaviruses are available, making the discovery of antivirals a high priority. Here, we identified and characterized three compounds from a library of kinase inhibitors that block replication of poliovirus, coxsackievirus B3, and encephalomyocarditis virus. Using an in vitro translation-replication system, we showed that these drugs inhibit different stages of the poliovirus life cycle. A4(1) inhibited both the formation and functioning of the replication complexes, while E5(1) and E7(2) were most effective during the formation but not the functioning step. Neither of the compounds significantly inhibited VPg uridylylation. Poliovirus resistant to E7(2) had a G5318A mutation in the 3A protein. This mutation was previously found to confer resistance to enviroxime-like compounds, which target a phosphatidylinositol 4-kinase IIIß (PI4KIIIß)-dependent step in viral replication. Analysis of host protein recruitment showed that E7(2) reduced the amount of GBF1 on the replication complexes; however, the level of PI4KIIIß remained intact. E7(2) as well as another enviroxime-like compound, GW5074, interfered with viral polyprotein processing affecting both 3C- and 2A-dependent cleavages, and the resistant G5318A mutation partially rescued this defect. Moreover, E7(2) induced abnormal recruitment to membranes of the viral proteins; thus, enviroxime-like compounds likely severely compromise the interaction of the viral polyprotein with membranes. A4(1) demonstrated partial protection from paralysis in a murine model of poliomyelitis. Multiple attempts to isolate resistant mutants in the presence of A4(1) or E5(1) were unsuccessful, showing that effective broad-spectrum antivirals could be developed on the basis of these compounds. IMPORTANCE: Diverse picornaviruses can trigger multiple human maladies, yet currently, only hepatitis A virus and poliovirus can be controlled with vaccination. The development of antipicornavirus therapeutics is also facing significant difficulties because these viruses readily generate resistance to compounds targeting either viral or cellular factors. Here, we describe three novel compounds that effectively block replication of distantly related picornaviruses with minimal toxicity to cells. The compounds prevent viral RNA replication after the synthesis of the uridylylated VPg primer. Importantly, two of the inhibitors are strongly refractory to the emergence of resistant mutants, making them promising candidates for further broad-spectrum therapeutic development. Evaluation of one of the compounds in an in vivo model of poliomyelitis demonstrated partial protection from the onset of paralysis.


Asunto(s)
Antivirales/farmacología , Poliomielitis/tratamiento farmacológico , Poliovirus/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , 1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , 1-Fosfatidilinositol 4-Quinasa/genética , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Animales , Antivirales/química , Sistema Libre de Células , Modelos Animales de Enfermedad , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/metabolismo , Enterovirus Humano B/efectos de los fármacos , Enterovirus Humano B/genética , Enterovirus Humano B/metabolismo , Regulación Viral de la Expresión Génica , Células HeLa , Humanos , Ratones , Mutación , Poliomielitis/virología , Poliovirus/genética , Poliovirus/crecimiento & desarrollo , Poliproteínas/antagonistas & inhibidores , Poliproteínas/genética , Poliproteínas/metabolismo , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Proteínas Virales/genética , Proteínas Virales/metabolismo
8.
Cell Host Microbe ; 14(3): 281-93, 2013 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-24034614

RESUMEN

Cholesterol is a critical component of cellular membranes, regulating assembly and function of membrane-based protein/lipid complexes. Many RNA viruses, including enteroviruses, remodel host membranes to generate organelles with unique lipid blueprints on which they assemble replication complexes and synthesize viral RNA. Here we find that clathrin-mediated endocytosis (CME) is harnessed by enteroviruses to traffic cholesterol from the plasma membrane (PM) and extracellular medium to replication organelles, where cholesterol then regulates viral polyprotein processing and facilitates genome synthesis. When CME is disrupted, cellular cholesterol pools are instead stored in lipid droplets, cholesterol cannot be trafficked to replication organelles, and replication is inhibited. In contrast, replication is stimulated in cholesterol-elevated cells like those lacking caveolins or those from Niemann-Pick disease patients. Our findings indicate cholesterol as a critical determinant for enteroviral replication and outline roles for the endocytic machinery in both the enteroviral life cycle and host cell cholesterol homeostasis.


Asunto(s)
Colesterol/metabolismo , Endocitosis , Enterovirus/fisiología , Interacciones Huésped-Patógeno , Replicación Viral , Membrana Celular/metabolismo , Membrana Celular/virología , Endosomas/metabolismo , Células HeLa , Humanos , Membranas Intracelulares/metabolismo
9.
PLoS Pathog ; 9(6): e1003401, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23762027

RESUMEN

All positive strand (+RNA) viruses of eukaryotes replicate their genomes in association with membranes. The mechanisms of membrane remodeling in infected cells represent attractive targets for designing future therapeutics, but our understanding of this process is very limited. Elements of autophagy and/or the secretory pathway were proposed to be hijacked for building of picornavirus replication organelles. However, even closely related viruses differ significantly in their requirements for components of these pathways. We demonstrate here that infection with diverse picornaviruses rapidly activates import of long chain fatty acids. While in non-infected cells the imported fatty acids are channeled to lipid droplets, in infected cells the synthesis of neutral lipids is shut down and the fatty acids are utilized in highly up-regulated phosphatidylcholine synthesis. Thus the replication organelles are likely built from de novo synthesized membrane material, rather than from the remodeled pre-existing membranes. We show that activation of fatty acid import is linked to the up-regulation of cellular long chain acyl-CoA synthetase activity and identify the long chain acyl-CoA syntheatse3 (Acsl3) as a novel host factor required for polio replication. Poliovirus protein 2A is required to trigger the activation of import of fatty acids independent of its protease activity. Shift in fatty acid import preferences by infected cells results in synthesis of phosphatidylcholines different from those in uninfected cells, arguing that the viral replication organelles possess unique properties compared to the pre-existing membranes. Our data show how poliovirus can change the overall cellular membrane homeostasis by targeting one critical process. They explain earlier observations of increased phospholipid synthesis in infected cells and suggest a simple model of the structural development of the membranous scaffold of replication complexes of picorna-like viruses, that may be relevant for other (+)RNA viruses as well.


Asunto(s)
Coenzima A Ligasas/biosíntesis , Ácidos Grasos/metabolismo , Regulación Enzimológica de la Expresión Génica , Poliomielitis/enzimología , Poliovirus/fisiología , Replicación Viral/fisiología , Transporte Biológico Activo , Cisteína Endopeptidasas/metabolismo , Células HeLa , Humanos , Poliomielitis/genética , Poliomielitis/metabolismo , Regulación hacia Arriba , Proteínas Virales/metabolismo
10.
J Virol ; 84(3): 1348-54, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19923182

RESUMEN

The DA strain and other members of the TO subgroup of Theiler's murine encephalomyelitis virus (TMEV) induce a persistent central nervous system infection associated with an inflammatory white matter demyelinating disease. TO subgroup strains synthesize an 18-kDa protein, L*, out of frame with the polyprotein from an initiation codon 13 nucleotides downstream from the polyprotein's AUG codon. We previously generated a mutant virus from our infectious DA full-length clone that has a change of the L* AUG codon to ACG (with no change in the polyprotein's amino acid sequence). Studies of this mutant virus showed that L* was key to the TO subgroup phenotype because the mutant had a decreased ability to persist and demyelinate. This work was initially called into question because a similar mutant derived from a different full-length DA infectious clone persisted and demyelinated similarly to wild-type DA virus (O. van Eyll and T. Michiels, J. Virol. 74:9071-9077, 2000). We now report that (i) the sequence of the L* coding region differs in the two infectious clones, resulting in a Ser or Leu as the predicted amino acid at position 93 of L* (with no change in the polyprotein's amino acid sequence), (ii) the difference in this amino acid is key to the phenotypic differences between the two mutants, and (iii) the change in amino acid 93 may affect L* phosphorylation. It is of interest that this amino acid only appears critical in determining the virus phenotype when L* is present in a significantly reduced amount (i.e., following translation from an ACG initiating codon).


Asunto(s)
Enfermedades Desmielinizantes/virología , Theilovirus/fisiología , Proteínas Virales/fisiología , Animales , Secuencia de Bases , Western Blotting , Línea Celular , Codón , Cricetinae , Cartilla de ADN , Ratones , Theilovirus/química , Proteínas Virales/química , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA