Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
mBio ; : e0191523, 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37962360

RESUMEN

IMPORTANCE: Multiple pattern recognition receptors sense vRNAs and initiate downstream innate signaling: endosomal Toll-like receptors (TLRs) 3, 7, and 8 and cytoplasmic RIG-I-like receptors (RLRs) RIG-I, and MDA5. They engage distinct signaling scaffolds: mitochondrial antiviral signaling protein (RLR), MyD88, and TLR-adaptor interacting with SLC15A4 on the lysosome (TLR7 and TLR8) and toll/IL-1R domain-containing adaptor inducing IFN (TLR3). By virtue of their unusual vRNA structure and direct host cell entry path, the innate response to EVs uniquely is orchestrated by MDA5. We reported that PVSRIPO's profound attenuation and loss of cytopathogenicity triggers MDA5-directed polar TBK1-IRF3 signaling that generates priming of polyfunctional antitumor CD8+ T-cell responses and durable antitumor surveillance in vivo. Here we unraveled EV-host relations that control suppression of host type-I IFN responses and show that PVSRIPO's deficient immediate host eIF4G cleavage generates unopposed MDA5-directed downstream signaling cascades resulting in sustained type-I IFN release.

2.
mBio ; 13(3): e0085422, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35652592

RESUMEN

For RNA virus families except Picornaviridae, viral RNA sensing includes Toll-like receptors and/or RIG-I. Picornavirus RNAs, whose 5' termini are shielded by a genome-linked protein, are predominately recognized by MDA5. This has important ramifications for adaptive immunity, as MDA5-specific patterns of type-I interferon (IFN) release are optimal for CD4+T cell TH1 polarization and CD8+T cell priming. We are exploiting this principle for cancer immunotherapy with recombinant poliovirus (PV), PVSRIPO, the type 1 (Sabin) PV vaccine containing a rhinovirus type 2 internal ribosomal entry site (IRES). Here we show that PVSRIPO-elicited MDA5 signaling is preceded by early sensing of the IRES by the double-stranded (ds)RNA-activated protein kinase (PKR). PKR binding to IRES stem-loop domains 5-6 led to dimerization and autoactivation, displaced host translation initiation factors, and suppressed viral protein synthesis. Early PKR-mediated antiviral responses tempered incipient viral translation and the activity of cytopathogenic viral proteinases, setting up accentuated MDA5 innate inflammation in response to PVSRIPO infection. IMPORTANCE Among the RIG-I-like pattern recognition receptors, MDA5 stands out because it senses long dsRNA duplexes independent of their 5' features (RIG-I recognizes viral [v]RNA 5'-ppp blunt ends). Uniquely among RNA viruses, the innate defense against picornaviruses is controlled by MDA5. We show that prior to engaging MDA5, recombinant PV RNA is sensed upon PKR binding to the viral IRES at a site that overlaps with the footprint for host translation factors mediating 40S subunit recruitment. Our study demonstrates that innate antiviral type-I IFN responses orchestrated by MDA5 involve separate innate modules that recognize distinct vRNA features and interfere with viral functions at multiple levels.


Asunto(s)
Enterovirus , Poliovirus , Virus ARN , Antivirales , Enterovirus/genética , Inmunidad Innata , Sitios Internos de Entrada al Ribosoma , Poliovirus/genética , Virus ARN/genética , ARN Bicatenario , ARN Viral/genética , Receptores de Reconocimiento de Patrones/genética
3.
mBio ; 12(2)2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33849973

RESUMEN

Enteroviruses (EV) deploy two proteases that mediate viral polyprotein cleavage and host cell manipulation. Here, we report that EV 2A proteases cleave all three members of the YTHDF protein family, cytosolic N6-methyladenosine (m6A) "readers" that regulate target mRNA fate. YTHDF protein cleavage occurs very early during infection, before viral translation is detected or cytopathogenic effects are observed. Preemptive YTHDF protein depletion enhanced viral translation and replication but only in cells with restrained viral translation, signs of inefficient 2A protease activity, and protective innate host immune responses. This effect corresponded with repression of interferon (IFN)-stimulated gene (ISG) induction, while type I/III IFN production was not significantly altered. Moreover, YTHDF3 depletion impaired JAK/STAT signaling in cells treated with type I, but not type II, IFN. YTHDF3 depletion's stimulatory effect on viral dynamics was dampened by JAK/STAT blockade and enhanced by type I IFN pretreatment of cells. We propose that EV 2A proteases cleave YTHDF proteins to antagonize ISG induction in infected cells.IMPORTANCE It is believed that ∼7,000 messenger RNAs (mRNAs) are subject to N6-methyladenosine modification. The biological significance of this remains mysterious. The YTHDF m6A readers are three related proteins with high affinity for m6A-modified mRNA, yet their biological functions remain obscure. We discovered that polio/enteroviruses elicit very early proteolysis of YTHDF1 to 3 in infected cells. Our research demonstrates that YTHDF3 acts as a positive regulator of antiviral JAK/STAT signaling in response to positive single-strand RNA virus infection, enabling type I interferon (IFN)-mediated gene regulatory programs to unfurl in infected cells. Our observation of viral degradation of the YTHDF proteins demonstrates that they are key response modifiers in the innate antiviral immune response.


Asunto(s)
Enterovirus/genética , Interferón Tipo I/metabolismo , Quinasas Janus/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Virales/metabolismo , Línea Celular , Enterovirus/enzimología , Células HeLa , Humanos , Inmunidad Innata , Interferón Tipo I/inmunología , Quinasas Janus/genética , Proteolisis , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Factores de Transcripción STAT/genética , Proteínas Virales/genética
4.
Curr Opin Virol ; 44: 73-80, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32771959

RESUMEN

Dendritic cells (DCs) are pivotal stimulators of T cell responses. They provide essential signals (epitope presentation, proinflammatory cytokines, co-stimulation) to T cells and prime adaptive immunity. Therefore, they are paramount to immunization strategies geared to generate T cell immunity. The inflammatory signals DCs respond to, classically occur in the context of acute virus infection. Yet, enlisting viruses for engaging DCs is hampered by their penchant for targeting DCs with sophisticated immune evasive and suppressive ploys. In this review, we discuss our work on devising vectors based on a recombinant polio:rhinovirus chimera for effectively targeting and engaging DCs. We are juxtaposing this approach with commonly used, recently studied dsDNA virus vector platforms.


Asunto(s)
Células Dendríticas/inmunología , Vectores Genéticos , Linfocitos T/inmunología , Tropismo Viral , Vacunas Virales/inmunología , Inmunidad Adaptativa , Animales , Presentación de Antígeno/inmunología , Células Dendríticas/virología , Humanos , Evasión Inmune , Inmunidad Innata , Ratones , Poliovirus/genética , Poliovirus/inmunología
5.
Sci Adv ; 6(23): eaba0745, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32537501

RESUMEN

Eukaryotic protein synthesis control at multiple levels allows for dynamic, selective responses to diverse conditions, but spatial organization of translation initiation machinery as a regulatory principle has remained largely unexplored. Here we report on a role of constitutive repressor of eIF2α phosphorylation (CReP) in translation of poliovirus and the endoplasmic reticulum (ER)-resident chaperone binding immunoglobulin protein (BiP) at the ER. Functional, proximity-dependent labeling and cell fractionation studies revealed that CReP, through binding eIF2α, anchors translation initiation machinery at the ER and enables local protein synthesis in this compartment. This ER site was protected from the suppression of cytoplasmic protein synthesis by acute stress responses, e.g., phosphorylation of eIF2α(S51) or mTOR blockade. We propose that partitioning of translation initiation machinery at the ER enables cells to maintain active translation during stress conditions associated with global protein synthesis suppression.

6.
Nat Commun ; 11(1): 524, 2020 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-31988324

RESUMEN

Viruses naturally engage innate immunity, induce antigen presentation, and mediate CD8 T cell priming against foreign antigens. Polioviruses can provide a context optimal for generating antigen-specific CD8 T cells, as they have natural tropism for dendritic cells, preeminent inducers of CD8 T cell immunity; elicit Th1-promoting inflammation; and lack interference with innate or adaptive immunity. However, notorious genetic instability and underlying neuropathogenicity has hampered poliovirus-based vector applications. Here we devised a strategy based on the polio:rhinovirus chimera PVSRIPO, devoid of viral neuropathogenicity after intracerebral inoculation in human subjects, for stable expression of exogenous antigens. PVSRIPO vectors infect, activate, and induce epitope presentation in DCs in vitro; they recruit and activate DCs with Th1-dominant cytokine profiles at the injection site in vivo. They efficiently prime tumor antigen-specific CD8 T cells in vivo, induce CD8 T cell migration to the tumor site, delay tumor growth and enhance survival in murine tumor models.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Células Dendríticas/inmunología , Poliovirus/inmunología , Animales , Vacunas contra el Cáncer , Vectores Genéticos/inmunología , Glioma/inmunología , Células HEK293 , Células HeLa , Humanos , Inmunidad Innata , Inmunoterapia/métodos , Interferón Tipo I/inmunología , Melanoma/inmunología , Ratones , Ratones Endogámicos C57BL , Poliovirus/genética
7.
Mol Cell Biol ; 38(19)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30012863

RESUMEN

Eukaryotic ribosomes contain the high-affinity protein kinase C ßII (PKCßII) scaffold, receptor for activated C kinase (RACK1), but its role in protein synthesis control remains unclear. We found that RACK1:PKCßII phosphorylates eukaryotic initiation factor 4G1 (eIF4G1) at S1093 and eIF3a at S1364. We showed that reversible eIF4G(S1093) phosphorylation is involved in a global protein synthesis surge upon PKC-Raf-extracellular signal-regulated kinase 1/2 (ERK1/2) activation and in induction of phorbol ester-responsive transcripts, such as cyclooxygenase 2 (Cox-2) and cyclin-dependent kinase inhibitor (p21Cip1), or in 5' 7-methylguanosine (m7G) cap-independent enterovirus translation. Comparison of mRNA and protein levels revealed that eIF4G1 or RACK1 depletion blocked phorbol ester-induced Cox-2 or p21Cip1 expression mostly at the translational level, whereas PKCß inhibition reduced them both at the translational and transcript levels. Our findings reveal a physiological role for ribosomal RACK1 in providing the molecular scaffold for PKCßII and its role in coordinating the translational response to PKC-Raf-ERK1/2 activation.


Asunto(s)
Factor 4G Eucariótico de Iniciación/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína Quinasa C beta/metabolismo , Receptores de Cinasa C Activada/metabolismo , Sustitución de Aminoácidos , Sitios de Unión/genética , Línea Celular , Factor 3 de Iniciación Eucariótica/química , Factor 3 de Iniciación Eucariótica/genética , Factor 3 de Iniciación Eucariótica/metabolismo , Factor 4G Eucariótico de Iniciación/química , Factor 4G Eucariótico de Iniciación/genética , Células HEK293 , Células HeLa , Humanos , Sistema de Señalización de MAP Quinasas , Modelos Moleculares , Proteínas de Neoplasias/genética , Iniciación de la Cadena Peptídica Traduccional/genética , Fosforilación/efectos de los fármacos , Proteína Quinasa C beta/genética , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo , Receptores de Cinasa C Activada/genética , Ribosomas/metabolismo , Acetato de Tetradecanoilforbol/farmacología
8.
Mol Cell Biol ; 38(19)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30012864

RESUMEN

The receptor for activated C kinase (RACK1), a conserved constituent of eukaryotic ribosomes, mediates phosphorylation of eukaryotic initiation factor 4G1(S1093) [eIF4G1(S1093)] and eIF3a(S1364) by protein kinase C ßII (PKCßII) (M. I. Dobrikov, E. Y. Dobrikova, and M. Gromeier, Mol Cell Biol 38:e00304-18, 2018, https://doi.org/10.1128/MCB.00304-18). RACK1:PKCßII activation drives a phorbol ester-induced surge of global protein synthesis and template-specific translation induction of PKC-Raf-extracellular signal-regulated kinase 1/2 (ERK1/2)-responsive genes. For unraveling mechanisms of RACK1:PKCßII-mediated translation stimulation, we used sequentially truncated eIF4G1 in coimmunoprecipitation analyses to delineate a set of autoinhibitory elements in the N-terminal unstructured region (surrounding the eIF4E-binding motif) and the interdomain linker (within the eIF3-binding site) of eIF4G1. Computer-based predictions of secondary structure, mutational analyses, and fluorescent titration with the ß-sheet dye thioflavin T suggest that eIF4G1(S1093) modulates a 4-stranded ß-sheet composed of antiparallel ß-hairpins formed by the autoinhibitory elements in eIF4G1's unstructured regions. The intact ß-sheet "locks" the eIF4G configuration, preventing assembly with eIF3/40S ribosomal subunits. Upon PKC stimulation, activated RACK1:PKCßII phosphorylates eIF4G(S1093) in the tight 48S initiation complex, possibly facilitating dissociation/recycling of eIF4F.


Asunto(s)
Factor 3 de Iniciación Eucariótica/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Factor 4G Eucariótico de Iniciación/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína Quinasa C beta/metabolismo , Receptores de Cinasa C Activada/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Benzotiazoles/metabolismo , Factor 4G Eucariótico de Iniciación/química , Factor 4G Eucariótico de Iniciación/genética , Células HEK293 , Humanos , Modelos Moleculares , Fosforilación , Unión Proteica , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Ribosomas/metabolismo
9.
Mol Cell Biol ; 38(11)2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29530922

RESUMEN

Death-associated protein 5 (DAP5) is an atypical isoform of the translation initiation scaffolds eukaryotic initiation factor 4GI (eIF4GI) and eIF4GII (eIF4GI/II), which recruit mRNAs to ribosomes in mammals. Unlike eIF4GI/II, DAP5 binds eIF2ß, a subunit of the eIF2 complex that delivers methionyl-tRNA to ribosomes. We discovered that DAP5:eIF2ß binding depends on specific stimuli, e.g., protein kinase C (PKC)-Raf-extracellular signal-regulated kinase 1/2 (ERK1/2) signals, and determines DAP5's influence on global and template-specific translation. DAP5 depletion caused an unanticipated surge of hypoxia-inducible factor 1α (HIF-1α), the transcription factor and master switch of the hypoxia response. Physiologically, the hypoxia response is tempered through HIF-1α hydroxylation by the oxygen-sensing prolyl hydroxylase-domain protein 2 (PHD2) and subsequent ubiquitination and degradation. We found that DAP5 regulates HIF-1α abundance through DAP5:eIF2ß-dependent translation of PHD2. DAP5:eIF2-induced PHD2 translation occurred during hypoxia-associated protein synthesis repression, indicating a role as a safeguard to reverse HIF-1α accumulation and curb the hypoxic response.


Asunto(s)
Factor 4G Eucariótico de Iniciación/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Hipoxia de la Célula , Regulación de la Expresión Génica/fisiología , Células HEK293 , Humanos , Hidroxilación/fisiología , Transducción de Señal/fisiología
10.
J Virol ; 88(22): 13135-48, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25187541

RESUMEN

UNLABELLED: Protein synthesis, the most energy-consuming process in cells, responds to changing physiologic priorities, e.g., upon mitogen- or stress-induced adaptations signaled through the mitogen-activated protein kinases (MAPKs). The prevailing status of protein synthesis machinery is a viral pathogenesis factor, particularly for plus-strand RNA viruses, where immediate translation of incoming viral RNAs shapes host-virus interactions. In this study, we unraveled signaling pathways centered on the ERK1/2 and p38α MAPK-interacting kinases MNK1/2 and their role in controlling 7-methyl-guanosine (m(7)G) "cap"-independent translation at enterovirus type 1 internal ribosomal entry sites (IRESs). Activation of Raf-MEK-ERK1/2 signals induced viral IRES-mediated translation in a manner dependent on MNK1/2. This effect was not due to MNK's known functions as eukaryotic initiation factor (eIF) 4G binding partner or eIF4E(S209) kinase. Rather, MNK catalytic activity enabled viral IRES-mediated translation/host cell cytotoxicity through negative regulation of the Ser/Arg (SR)-rich protein kinase (SRPK). Our investigations suggest that SRPK activity is a major determinant of type 1 IRES competency, host cell cytotoxicity, and viral proliferation in infected cells. IMPORTANCE: We are targeting unfettered enterovirus IRES activity in cancer with PVSRIPO, the type 1 live-attenuated poliovirus (PV) (Sabin) vaccine containing a human rhinovirus type 2 (HRV2) IRES. A phase I clinical trial of PVSRIPO with intratumoral inoculation in patients with recurrent glioblastoma (GBM) is showing early promise. Viral translation proficiency in infected GBM cells is a core requirement for the antineoplastic efficacy of PVSRIPO. Therefore, it is critically important to understand the mechanisms controlling viral cap-independent translation in infected host cells.


Asunto(s)
Enterovirus/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Virales/biosíntesis , Línea Celular , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos
11.
Cancer ; 120(21): 3277-86, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24939611

RESUMEN

Recently, the century-old idea of targeting cancer with viruses (oncolytic viruses) has come of age, and promise has been documented in early stage and several late-stage clinical trials in a variety of cancers. Although originally prized for their direct tumor cytotoxicity (oncolytic virotherapy), recently, the proinflammatory and immunogenic effects of viral tumor infection (oncolytic immunotherapy) have come into focus. Indeed, a capacity for eliciting broad, sustained antineoplastic effects stemming from combined direct viral cytotoxicity, innate antiviral activation, stromal proinflammatory stimulation, and recruitment of adaptive immune effector responses is the greatest asset of oncolytic viruses. However, it also is the source for enormous mechanistic complexity that must be considered for successful clinical translation. Because of fundamentally different relationships with their hosts (malignant or not), diverse replication strategies, and distinct modes of tumor cytotoxicity/killing, oncolytic viruses should not be referred to collectively. These agents must be evaluated based on their individual merits. In this review, the authors highlight key mechanistic principles of cancer treatment with the polio:rhinovirus chimera PVSRIPO and their implications for oncolytic immunotherapy in the clinic.


Asunto(s)
Inmunidad Innata/genética , Neoplasias/genética , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Humanos , Inmunoterapia , Neoplasias/virología , Poliomielitis/genética , Receptores Virales/inmunología , Internalización del Virus
12.
Mol Cell Biol ; 33(5): 937-46, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23263986

RESUMEN

Eukaryotic translation initiation factor 4F (eIF4F), comprising the cap-binding protein eIF4E, the helicase eIF4A, and the central scaffold eIF4G, is a convergence node for a complex signaling network that controls protein synthesis. Together with eIF3 and eIF4A/4B, eIF4G recruits ribosomal subunits to mRNAs and facilitates 5' untranslated region unwinding. Mammalian eIF4G contains three HEAT domains and unstructured regions involved in protein-protein interactions. Despite detailed eIF4G structure data, the mechanisms controlling initiation scaffold formation remain obscure. We found a new, highly regulated eIF4B/-3 binding site within the HEAT-1/-2 interdomain linker, harboring two phosphorylation sites that we identified as substrates for Erk1/2 and casein kinase 2. Phorbol ester-induced sequential phosphorylation of both sites detached HEAT-2 from the complex with eIF4A/-4B/-3 and stimulated the association of HEAT-3 with the mitogen-activated protein kinase signal integrating kinase Mnk1. Our results provide a mechanistic link between intracellular signal transduction and dynamic initiation complex formation coordinated by flexible eIF4G structure.


Asunto(s)
Factor 4G Eucariótico de Iniciación/química , Factor 4G Eucariótico de Iniciación/metabolismo , Secuencia de Aminoácidos , Quinasa de la Caseína II/metabolismo , Factor 3 de Iniciación Eucariótica/metabolismo , Factor 4A Eucariótico de Iniciación/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Mapeo de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína
13.
Mol Cell Biol ; 33(1): 127-35, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23109423

RESUMEN

The p38α to p38δ mitogen-activated protein kinases (MAPKs) are central regulatory nodes coordinating acute stress and inflammatory responses. Their activation leads to rapid adjustment of protein synthesis, for instance translational induction of proinflammatory cytokines. The only known direct link of p38 to translation machinery is the MAPK signal-integrating kinase Mnk. Only p38α and p38ß transcripts are ubiquitously expressed. These mRNAs encode highly conserved proteins that equally phosphorylate recombinant Mnk1 in vitro. We discovered that expression of the p38α protein, but not the p38ß isoform, is suppressed in the brain. This is due to p38α depletion by two neuron-selective microRNAs (miRNAs), miR-124 and -128. Suppression of p38α protein was reversed by miR-124/-128 antisense oligonucleotides in primary explant neuronal cultures. Targeted p38α depletion reduced Mnk1 activation, which cannot be compensated by p38ß. Our research shows that p38α alone controls acute stress and cytokine signaling from p38 MAPK to translation machinery. This regulatory axis is greatly diminished in neurons, which may insulate brain physiology and function from p38α-Mnk1-mediated signaling.


Asunto(s)
MicroARNs/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Neuronas/fisiología , Animales , Secuencia de Bases , Cerebelo/citología , Cerebelo/fisiología , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Datos de Secuencia Molecular , Neuronas/metabolismo , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Técnicas de Cultivo de Órganos , Fosforilación , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal
14.
J Virol ; 86(5): 2750-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22171271

RESUMEN

A dependence of poliovirus on an unorthodox translation initiation mode can be targeted selectively to drive viral protein synthesis and cytotoxicity in malignant cells. Transformed cells are naturally susceptible to poliovirus, due to widespread ectopic upregulation of the poliovirus receptor, Necl-5, in ectodermal/neuroectodermal cancers. Viral tumor cell killing and the host immunologic response it engenders produce potent, lasting antineoplastic effects in animal tumor models. Clinical application of this principle depends on unequivocal demonstration of safety in primate models for paralytic poliomyelitis. We conducted extensive dose-range-finding, toxicity, biodistribution, shedding, and neutralizing antibody studies of the prototype oncolytic poliovirus recombinant, PVS-RIPO, after intrathalamic inoculation in Macaca fascicularis. These studies suggest that intracerebral PVS-RIPO inoculation does not lead to viral propagation in the central nervous system (CNS), does not cause histopathological CNS lesions or neurological symptoms that can be attributed to the virus, is not associated with extraneural virus dissemination or replication and does not induce shedding of virus with stool. Intrathalamic PVS-RIPO inoculation induced neutralizing antibody responses against poliovirus serotype 1 in all animals studied.


Asunto(s)
Quimera/fisiología , Modelos Animales de Enfermedad , Macaca fascicularis , Poliomielitis/virología , Poliovirus/fisiología , Poliovirus/patogenicidad , Rhinovirus/genética , Esparcimiento de Virus , Animales , Anticuerpos Antivirales/inmunología , Línea Celular , Quimera/genética , Humanos , Macaca fascicularis/inmunología , Macaca fascicularis/virología , Poliomielitis/inmunología , Poliovirus/genética , Rhinovirus/fisiología , Distribución Tisular , Virulencia
15.
PLoS One ; 5(7): e11710, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20668518

RESUMEN

BACKGROUND: RNA-binding proteins accompany all steps in the life of mRNAs and provide dynamic gene regulatory functions for rapid adjustment to changing extra- or intracellular conditions. The association of RNA-binding proteins with their targets is regulated through changing subcellular distribution, post-translational modification or association with other proteins. METHODOLOGY: We demonstrate that the dsRNA binding protein 76 (DRBP76), synonymous with nuclear factor 90, displays inherently distinct tissue type-specific subcellular distribution in the normal human central nervous system and in malignant brain tumors of glial origin. Altered subcellular localization and isoform distribution in malignant glioma indicate that tumor-specific changes in DRBP76-related gene products and their regulatory functions may contribute to the formation and/or maintenance of these tumors. To identify endogenous mRNA targets of DRBP76, we performed RNA-immunoprecipitation and genome-wide microarray analyses in HEK293 cells, and identified specific classes of transcripts encoding critical functions in cellular metabolism. SIGNIFICANCE: Our data suggest that physiologic DRBP76 expression, isoform distribution and subcellular localization are profoundly altered upon malignant transformation. Thus, the functional role of DRBP76 in co- or post-transcriptional gene regulation may contribute to the neoplastic phenotype.


Asunto(s)
Proteínas del Factor Nuclear 90/metabolismo , ARN Mensajero/genética , Animales , Encéfalo/metabolismo , Línea Celular , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Técnicas In Vitro , Macaca , Ratones , Proteínas del Factor Nuclear 90/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Mol Ther ; 16(11): 1865-72, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18766173

RESUMEN

Many viruses, either naturally occurring or as a result of genetic manipulation, exhibit conditional replication in transformed cells. This principle is the basis for experimental therapeutic approaches exploiting the oncolytic potential of such agents without the danger of collateral damage to resistant normal tissues. One of the potential obstacles to these approaches is the possibility of genetic adaptation of oncolytic viruses upon replication in susceptible tumor tissues. Genetic variation can reverse genetic manipulations of parental viral genomes that determine attenuation of virulence, selective tumor cell tropism or other desirable traits. Alternatively, it may convey new properties not originally associated with parental strains, e.g., adaptation to a human host range. We examined genetic stability of an oncolytic nonpathogenic poliovirus recombinant considered for therapy of recurrent glioblastoma multiforme (GBM). This was done by serial passage experiments in glioma xenografts in vivo and investigation of phenotypic and genotypic markers of attenuation. Intratumoral inoculation of oncolytic poliovirus produced efficient tumor regress and elimination without altering temperature-sensitive growth, selective cytotoxicity, or genetic markers of attenuation of virus recovered from inoculated animals. Our studies demonstrate that active viral oncolysis of malignant glioma does not alter the conditional replication properties of oncolytic nonpathogenic poliovirus recombinants.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Virus Oncolíticos , Poliovirus , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Viroterapia Oncolítica , Trasplante Heterólogo
17.
RNA ; 14(10): 2170-82, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18755839

RESUMEN

Protein synthesis is tightly controlled by assembly of an intricate ribonucleoprotein complex at the m(7)GTP-cap on eukaryotic mRNAs. Ensuing linear scanning of the 5' untranslated region (UTR) is believed to transfer the preinitiation complex to the initiation codon. Eukaryotic mRNAs are characterized by significant 5' UTR heterogeneity, raising the possibility of differential control of translation initiation rate at individual mRNAs. Curiously, many mRNAs with unconventional, highly structured 5' UTRs encode proteins with central biological roles in growth control, metabolism, or stress response. The 5' UTRs of such mRNAs may influence protein synthesis rate in multiple ways, but most significantly they have been implicated in mediating alternative means of translation initiation. Cap-independent initiation bypasses strict control over the formation of initiation intermediates at the m(7)GTP cap. However, the molecular mechanisms that favor alternative means of ribosome recruitment are not understood. Here we provide evidence that eukaryotic initiation factor (eIF) 4G controls cap-independent translation initiation at the c-myc and vascular endothelial growth factor (VEGF) 5' UTRs in vivo. Cap-independent translation was investigated in tetracycline-inducible cell lines expressing either full-length eIF4G or a C-terminal fragment (Ct) lacking interaction with eIF4E and poly(A) binding protein. Expression of Ct, but not intact eIF4G, potently stimulated cap-independent initiation at the c-myc/VEGF 5' UTRs. In vitro RNA-binding assays suggest that stimulation of cap-independent translation initiation by Ct is due to direct association with the c-myc/VEGF 5' UTR, enabling 43S preinitiation complex recruitment. Our work demonstrates that variant translation initiation factors enable unconventional translation initiation at mRNA subsets with distinct structural features.


Asunto(s)
Regiones no Traducidas 5'/metabolismo , Factor 4G Eucariótico de Iniciación/metabolismo , Iniciación de la Cadena Peptídica Traduccional , Análogos de Caperuza de ARN/metabolismo , Línea Celular , Factor 4G Eucariótico de Iniciación/genética , Humanos , Poliadenilación , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Factor A de Crecimiento Endotelial Vascular/biosíntesis
18.
J Med Virol ; 80(2): 352-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18098139

RESUMEN

The use of oncolytic recombinant polioviruses has an important therapeutic potential in the treatment of human gliomas. This study was carried out to assess parameters of the utility of the oncolytic poliovirus/human rhinovirus type 2 chimeras (PV/HRV2). The prototype PV/HRV2 chimera was constructed containing the complete genome of wild-type PV type 1 (Mahoney) [PV1(M)] in which the cognate IRES was replaced with that of HRV2 [called PV1(RIPO)]. A derivative of PV1(RIPO) is PV1(RIPOS) in which the capsid coding region (P1) was replaced with the capsid-coding region of the PV type 1 (Sabin) [PV1(S)] vaccine strain. In addition, a third PV/HRV2 chimera was constructed containing the complete genome of PV1(S) in which the cognate IRES was replaced with that of HRV2 [termed PVS(RIPO)]. To analyze the growth phenotypes of PV/HRV2 recombinants [PV1(RIPO), PV1(RIPOS), PVS(RIPO)], one-step growth experiments were performed in four human cell lines at three different temperatures. To address the safety profile, PVS(RIPO) was injected into the brain of CD155 tg mice at the dose 10(7) PFU. Then, clinical signs, persistence of the virus in the CNS and genetic stability of PVS(RIPO) replicating in the CNS were evaluated. The data obtained in the present study suggest (i) a correlation between temperature-sensitive (ts) phenotype in both neuronal and non-neuronal cell lines and neuroattenuation in experimental animals, (ii) that PVS (RIPO) is genetically stable on replication in the CNS of poliovirus-susceptible mice. These findings highlight the safety of intracerebral inoculation of PVS(RIPO) for the treatment of human glioma.


Asunto(s)
Proteínas de la Membrana/genética , Viroterapia Oncolítica , Poliovirus/crecimiento & desarrollo , Receptores Virales/genética , Rhinovirus/crecimiento & desarrollo , Animales , Encéfalo/virología , Línea Celular , Genoma Viral , Humanos , Ratones , Ratones Transgénicos , Poliovirus/genética , Poliovirus/patogenicidad , Recombinación Genética , Rhinovirus/genética , Rhinovirus/patogenicidad
19.
RNA ; 13(9): 1582-93, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17652408

RESUMEN

The 3' poly(A) tail present on the majority of mature eukaryotic mRNAs is an important regulator of protein synthesis and mRNA stability. The poly(A) tail improves the efficiency of translation initiation through recruitment of PABP, enabling its interaction with eIF4F located at the mRNA 5'-end. Recent evidence has also implicated a possible role for PABP and the poly(A) tract in translation control at steps beyond the initiation phase. Similar to conventional mRNAs, plus-strand RNA virus genomes that utilize internal ribosome entry sites (IRESes) to promote cap-independent translation are influenced by PABP and poly(A) status. However, the relative contribution of these factors to translation initiation mediated by distinct IRESes is unclear. We have investigated cis- and trans-acting effects of poly(A) and PABP, respectively, on RNAs harboring IRESes from three diverse viruses: encephalomyocarditis virus (EMCV), hepatitis C virus (HCV), and coxsackievirus B3 (CBV3). A 3' poly(A) tract enhanced translation of both capped and IRES-containing reporter RNAs. However, only CBV3 and capped transcripts were stabilized as a result of polyadenylation. Correspondingly, translation of polyadenylated CBV3 and capped RNAs displayed heightened sensitivity to the PABP inhibitor Paip2 compared with EMCV and HCV. Sucrose density gradient analyses suggested a stimulatory role for PABP and 3' poly(A) in the CBV3 initiation phase, while assembly of HCV and EMCV RNAs into ribosomal complexes was little affected by either factor. Collectively, the observed differential effects of PABP and poly(A) on translation imply mechanistic differences between viral IRES elements and suggest modulating roles for PABP and the poly(A) tail at multiple phases of translation.


Asunto(s)
Proteínas de Unión a Poli(A)/fisiología , Biosíntesis de Proteínas/genética , ARN Viral/genética , Ribosomas/fisiología , Virus de la Encefalomiocarditis/genética , Enterovirus Humano B/genética , Escherichia coli , Células HeLa , Hepacivirus/genética , Humanos , Poli A/genética , Proteínas de Unión a Poli(A)/química , Proteínas de Unión a Poli(A)/genética , Estabilidad del ARN/genética , Ribosomas/química , Ribosomas/genética
20.
J Virol ; 80(7): 3147-56, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16537583

RESUMEN

Translation of picornavirus plus-strand RNA genomes occurs via internal ribosomal entry at highly structured 5' untranslated regions. In addition to canonical translation factors, translation rate is likely influenced by supplementary host and viral trans-acting factors. We previously reported that insertion of a heterologous human rhinovirus type 2 internal ribosomal entry site (IRES) into the poliovirus (PV) genome, generating the chimeric virus PV-RIPO, selectively abrogates viral translation and propagation in neurons, which eliminate poliovirus's signature neuropathogenicity. While severely deficient in cells of neuronal lineage, the rhinovirus IRES promotes efficient propagation of PV-RIPO in cancer cells. Tumor-specific IRES function can be therapeutically exploited to direct viral cytotoxicity to cancer cells. Neuron-glioma heterokaryon analysis implicates neuronal trans-dominant inhibition in this effect, suggesting that host trans-acting factors repress IRES function in a cell-type-specific manner. We identified a set of proteins from neuronal cells with affinity for the rhinovirus IRES, including double-stranded RNA-binding protein 76 (DRBP76). DRBP76 associates with the IRES in neuronal but not in malignant glioma cells. Moreover, DRBP76 depletion in neuronal cells enhances rhinovirus IRES-driven translation and virus propagation. Our observations suggest that cell-type-specific association of DRBP76 with the rhinovirus IRES represses PV-RIPO translation and propagation in neuronal cells.


Asunto(s)
Regulación Viral de la Expresión Génica , Ribonucleoproteínas Nucleares Heterogéneas , Proteínas de Unión al ARN/metabolismo , Ribosomas/metabolismo , Western Blotting , Extractos Celulares/química , Línea Celular , Línea Celular Tumoral , Glioma/química , Humanos , Neuronas/química , Pruebas de Precipitina , Interferencia de ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/aislamiento & purificación , Rhinovirus/genética , Ribosomas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...