Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int J Mol Sci ; 23(23)2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36499391

RESUMEN

The SH-SY5Y neuroblastoma cells are a widely used in vitro model approximating neurons for testing the target engagement of therapeutics designed for neurodegenerative diseases and pain disorders. However, their potential as a model for receptor-mediated delivery and uptake of novel modalities, such as antibody-drug conjugates, remains understudied. Investigation of the SH-SY5Y cell surfaceome will aid in greater in vitro to in vivo correlation of delivery and uptake, thereby accelerating drug discovery. So far, the majority of studies have focused on total cell proteomics from undifferentiated and differentiated SH-SY5Y cells. While some studies have investigated the expression of specific proteins in neuroblastoma tissue, a global approach for comparison of neuroblastoma cell surfaceome to the brain and dorsal root ganglion (DRG) neurons remains uninvestigated. Furthermore, an isoform-specific evaluation of cell surface proteins expressed on neuroblastoma cells remains unexplored. In this study, we define a bioinformatic workflow for the identification of high-confidence surface proteins expressed on brain and DRG neurons using tissue proteomic and transcriptomic data. We then delineate the SH-SY5Y cell surfaceome by surface proteomics and show that it significantly overlaps with the human brain and DRG neuronal surface proteome. We find that, for 32% of common surface proteins, SH-SY5Y-specific major isoforms are alternatively spliced, maintaining their protein-coding ability, and are predicted to localize to the cell surface. Validation of these isoforms using surface proteomics confirms a SH-SY5Y-specific alternative NRCAM (neuron-glia related cell adhesion molecule) isoform, which is absent in typical brain neurons, but present in neuroblastomas, making it a receptor of interest for neuroblastoma-specific therapeutics.


Asunto(s)
Neuroblastoma , Humanos , Neuroblastoma/terapia , Neuroblastoma/tratamiento farmacológico , Línea Celular Tumoral , Proteómica , Neuronas/metabolismo , Diferenciación Celular/fisiología , Proteínas de la Membrana/metabolismo
2.
PLoS Pathog ; 15(6): e1007790, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31194854

RESUMEN

Bourbon virus (BRBV) is an emerging tick-borne RNA virus in the orthomyxoviridae family that was discovered in 2014. Although fatal human cases of BRBV have been described, little is known about its pathogenesis, and no antiviral therapies or vaccines exist. We obtained serum from a fatal case in 2017 and successfully recovered the second human infectious isolate of BRBV. Next-generation sequencing of the St. Louis isolate of BRBV (BRBV-STL) showed >99% nucleotide identity to the original reference isolate. Using BRBV-STL, we developed a small animal model to study BRBV-STL tropism in vivo and evaluated the prophylactic and therapeutic efficacy of the experimental antiviral drug favipiravir against BRBV-induced disease. Infection of Ifnar1-/- mice lacking the type I interferon receptor, but not congenic wild-type animals, resulted in uniformly fatal disease 6 to 10 days after infection. RNA in situ hybridization and viral yield assays demonstrated a broad tropism of BRBV-STL with highest levels detected in liver and spleen. In vitro replication and polymerase activity of BRBV-STL were inhibited by favipiravir. Moreover, administration of favipiravir as a prophylaxis or as post-exposure therapy three days after infection prevented BRBV-STL-induced mortality in immunocompromised Ifnar1-/- mice. These results suggest that favipiravir may be a candidate treatment for humans who become infected with BRBV.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Infecciones por Orthomyxoviridae/prevención & control , Pirazinas/farmacología , Thogotovirus/inmunología , Animales , Chlorocebus aethiops , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/inmunología , Thogotovirus/patogenicidad , Células Vero , Tropismo Viral/efectos de los fármacos , Tropismo Viral/genética , Tropismo Viral/inmunología
3.
J Biol Chem ; 294(39): 14231-14240, 2019 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-31375559

RESUMEN

Innate immune detection of viral nucleic acids during viral infection activates a signaling cascade that induces type I and type III IFNs as well as other cytokines, to generate an antiviral response. This signaling is initiated by pattern recognition receptors, such as the RNA helicase retinoic acid-inducible gene I (RIG-I), that sense viral RNA. These sensors then interact with the adaptor protein mitochondrial antiviral signaling protein (MAVS), which recruits additional signaling proteins, including TNF receptor-associated factor 3 (TRAF3) and TANK-binding kinase 1 (TBK1), to form a signaling complex that activates IFN regulatory factor 3 (IRF3) for transcriptional induction of type I IFNs. Here, using several immunological and biochemical approaches in multiple human cell types, we show that the GTPase-trafficking protein RAB1B up-regulates RIG-I pathway signaling and thereby promotes IFN-ß induction and the antiviral response. We observed that RAB1B overexpression increases RIG-I-mediated signaling to IFN-ß and that RAB1B deletion reduces signaling of this pathway. Additionally, loss of RAB1B dampened the antiviral response, indicated by enhanced Zika virus infection of cells depleted of RAB1B. Importantly, we identified the mechanism of RAB1B action in the antiviral response, finding that it forms a protein complex with TRAF3 to facilitate the interaction of TRAF3 with mitochondrial antiviral signaling protein. We conclude that RAB1B regulates TRAF3 and promotes the formation of innate immune signaling complexes in response to nucleic acid sensing during RNA virus infection.


Asunto(s)
Inmunidad Innata , Factor 3 Asociado a Receptor de TNF/metabolismo , Infección por el Virus Zika/inmunología , Proteínas de Unión al GTP rab1/metabolismo , Animales , Chlorocebus aethiops , Proteína 58 DEAD Box/metabolismo , Células HEK293 , Humanos , Interferón beta/metabolismo , Unión Proteica , Receptores Inmunológicos , Transducción de Señal , Células Vero
4.
J Virol ; 90(9): 4796-4806, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26937034

RESUMEN

UNLABELLED: Reassortment between H5 or H9 subtype avian and mammalian influenza A viruses (IAV) can generate a novel virus that causes disease and transmits between mammals. Such information is currently not available for H7 subtype viruses. We evaluated the ability of a low-pathogenicity North American avian H7N3 virus (A/shorebird/Delaware/22/2006) to reassort with mammalian or avian viruses using a plasmid-based competition assay. In addition to genome segments derived from an avian H7N9 virus, the H7N3 virus reassorted efficiently with the PB2, NA, and M segments from the 2009 pandemic H1N1 (PH1N1) virus.In vitro and in vivo evaluation of the H7N3:PH1N1 (7 + 1) reassortant viruses revealed that the PB2, NA, or M segments from PH1N1 largely do not attenuate the H7N3 virus, whereas the PB1, PA, NP, or NS genome segments from PH1N1 do. Additionally, we assessed the functionality of the H7N3:PH1N1 7 + 1 reassortant viruses by measuring the inflammatory response in vivo We found that infection with wild-type H7N3 resulted in increased inflammatory cytokine production relative to that seen with the PH1N1 strain and that the increase was further exacerbated by substitution of PH1N1 PB2 but not NA or M. Finally, we assessed if any adaptations occurred in the individually substituted segments after in vivo inoculation and found no mutations, suggesting that PH1N1 PB2, NA, and M are genetically stable in the background of this H7N3 virus. Taking the data together, we demonstrate that a North American avian H7N3 IAV is genetically and functionally compatible with multiple gene segments from the 2009 pandemic influenza virus strain without prior adaptation. IMPORTANCE: The 2009 pandemic H1N1 virus continues to circulate and reassort with other influenza viruses, creating novel viruses with increased replication and transmission potential in humans. Previous studies have found that this virus can also reassort with H5N1 and H9N2 avian influenza viruses. We now show that several genome segments of the 2009 H1N1 virus are also highly compatible with a low-pathogenicity avian H7N3 virus and that these reassortant viruses are stable and not attenuated in an animal model. These results highlight the potential for reassortment of H1N1 viruses with avian influenza virus and emphasize the need for continued surveillance of influenza viruses in areas of cocirculation between avian, human, and swine viruses.


Asunto(s)
Adaptación Biológica , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H7N3 del Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/virología , Virus Reordenados/fisiología , Animales , Línea Celular , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Variación Genética , Genoma Viral , Humanos , Mediadores de Inflamación/metabolismo , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H7N3 del Virus de la Influenza A/genética , Ratones , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/mortalidad , Filogenia , Virus Reordenados/genética , Virus Reordenados/patogenicidad , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
5.
J Virol ; 89(18): 9465-76, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26157117

RESUMEN

UNLABELLED: Interferon-induced protein with tetratricopeptide repeats 1 (IFIT1) is a host protein with reported cell-intrinsic antiviral activity against several RNA viruses. The proposed basis for the activity against negative-sense RNA viruses is the binding to exposed 5'-triphosphates (5'-ppp) on the genome of viral RNA. However, recent studies reported relatively low binding affinities of IFIT1 for 5'-ppp RNA, suggesting that IFIT1 may not interact efficiently with this moiety under physiological conditions. To evaluate the ability of IFIT1 to have an impact on negative-sense RNA viruses, we infected Ifit1(-/-) and wild-type control mice and primary cells with four negative-sense RNA viruses (influenza A virus [IAV], La Crosse virus [LACV], Oropouche virus [OROV], and Ebola virus) corresponding to three distinct families. Unexpectedly, a lack of Ifit1 gene expression did not result in increased infection by any of these viruses in cell culture. Analogously, morbidity, mortality, and viral burdens in tissues were identical between Ifit1(-/-) and control mice after infection with IAV, LACV, or OROV. Finally, deletion of the human IFIT1 protein in A549 cells did not affect IAV replication or infection, and reciprocally, ectopic expression of IFIT1 in HEK293T cells did not inhibit IAV infection. To explain the lack of antiviral activity against IAV, we measured the binding affinity of IFIT1 for RNA oligonucleotides resembling the 5' ends of IAV gene segments. The affinity for 5'-ppp RNA was approximately 10-fold lower than that for non-2'-O-methylated (cap 0) RNA oligonucleotides. Based on this analysis, we conclude that IFIT1 is not a dominant restriction factor against negative-sense RNA viruses. IMPORTANCE: Negative-sense RNA viruses, including influenza virus and Ebola virus, have been responsible for some of the most deadly outbreaks in recent history. The host interferon response and induction of antiviral genes contribute to the control of infections by these viruses. IFIT1 is highly induced after virus infection and reportedly has antiviral activity against several RNA and DNA viruses. However, its role in restricting infection by negative-sense RNA viruses remains unclear. In this study, we evaluated the ability of IFIT1 to inhibit negative-sense RNA virus replication and pathogenesis both in vitro and in vivo. Detailed cell culture and animal studies demonstrated that IFIT1 is not a dominant restriction factor against three different families of negative-sense RNA viruses.


Asunto(s)
Proteínas Portadoras/metabolismo , Ebolavirus/metabolismo , Virus de la Influenza A/metabolismo , Virus La Crosse/metabolismo , Infecciones por Virus ARN/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/genética , Línea Celular , Ebolavirus/genética , Humanos , Virus de la Influenza A/genética , Virus La Crosse/genética , Ratones , Ratones Noqueados , Infecciones por Virus ARN/genética , Proteínas de Unión al ARN
6.
mSphere ; 5(3)2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32404510

RESUMEN

Type I interferons (IFN) initiate an antiviral state through a signal transduction cascade that leads to the induction of hundreds of IFN-stimulated genes (ISGs) to restrict viral infection. Recently, RNA modifications on both host and viral RNAs have been described as regulators of infection. However, the impact of host mRNA cap modifications on the IFN response and how this regulates viral infection are unknown. Here, we reveal that CMTR1, an ISG that catalyzes 2'-O-methylation of the first transcribed nucleotide in cellular mRNA (Cap 1), promotes the protein expression of specific ISGs that contribute to the antiviral response. Depletion of CMTR1 reduces the IFN-induced protein levels of ISG15, MX1, and IFITM1, without affecting their transcript abundance. However, CMTR1 depletion does not significantly affect the IFN-induced protein or transcript abundance of IFIT1 and IFIT3. Importantly, knockdown of IFIT1, which acts with IFIT3 to inhibit the translation of RNAs lacking Cap 1 2'-O-methylation, restores protein expression of ISG15, MX1, and IFITM1 in cells depleted of CMTR1. Finally, we found that CMTR1 plays a role in restricting RNA virus replication, likely by ensuring the expression of specific antiviral ISGs. Taken together, these data reveal that CMTR1 is required to establish an antiviral state by ensuring the protein expression of a subset of ISGs during the type I IFN response.IMPORTANCE Induction of an efficient type I IFN response is important to control viral infection. We show that the host 2'-O-methyltransferase CMTR1 facilitates the protein expression of ISGs in human cells by preventing IFIT1 from inhibiting the translation of those mRNAs lacking cap 2'-O-methylation. Thus, CMTR1 promotes the IFN-mediated antiviral response.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Interferón Tipo I/inmunología , Metiltransferasas/genética , ARN Mensajero/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular , Células HEK293 , Humanos , Inmunidad Innata , Péptidos y Proteínas de Señalización Intracelular/genética , Metilación , Biosíntesis de Proteínas , Virus ARN/inmunología , Virus ARN/fisiología , Proteínas de Unión al ARN/genética , Transducción de Señal , Células THP-1 , Replicación Viral/inmunología
7.
Annu Rev Virol ; 6(1): 235-253, 2019 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-31283446

RESUMEN

In recent years, the RNA modification N6-methyladenosine (m6A) has been found to play a role in the life cycles of numerous viruses and also in the cellular response to viral infection. m6A has emerged as a regulator of many fundamental aspects of RNA biology. Here, we highlight recent advances in techniques for the study of m6A, as well as advances in our understanding of the cellular machinery that controls the addition, removal, recognition, and functions of m6A. We then summarize the many newly discovered roles of m6A during viral infection, including how it regulates innate and adaptive immune responses to infection. Overall, the goals of this review are to summarize the roles of m6A on both cellular and viral RNAs and to describe future directions for uncovering new functions of m6A during infection.


Asunto(s)
Adenosina/análogos & derivados , Regulación de la Expresión Génica , Interacciones Microbiota-Huesped , ARN Viral/química , Inmunidad Adaptativa , Adenosina/química , Animales , Humanos , Inmunidad Innata , Ratones , Procesamiento Postranscripcional del ARN , ARN Viral/genética , Virosis/inmunología , Replicación Viral
8.
J Exp Med ; 216(1): 117-132, 2019 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-30559127

RESUMEN

Tissue-resident memory CD8+ T cells (TRMs) confer rapid protection and immunity against viral infections. Many viruses have evolved mechanisms to inhibit MHCI presentation in order to evade CD8+ T cells, suggesting that these mechanisms may also apply to TRM-mediated protection. However, the effects of viral MHCI inhibition on the function and generation of TRMs is unclear. Herein, we demonstrate that viral MHCI inhibition reduces the abundance of CD4+ and CD8+ TRMs, but its effects on the local microenvironment compensate to promote antigen-specific CD8+ TRM formation. Unexpectedly, local cognate antigen enhances CD8+ TRM development even in the context of viral MHCI inhibition and CD8+ T cell evasion, strongly suggesting a role for in situ cross-presentation in local antigen-driven TRM differentiation. However, local cognate antigen is not required for CD8+ TRM maintenance. We also show that viral MHCI inhibition efficiently evades CD8+ TRM effector functions. These findings indicate that viral evasion of MHCI antigen presentation has consequences on the development and response of antiviral TRMs.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Evasión Inmune , Memoria Inmunológica , Virosis/inmunología , Animales , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Chlorocebus aethiops , Perros , Células de Riñón Canino Madin Darby , Ratones , Ratones Transgénicos , Células Vero , Virosis/genética , Virosis/patología
9.
Nat Commun ; 9(1): 465, 2018 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-29386621

RESUMEN

Influenza A virus nucleoprotein (NP) association with viral RNA (vRNA) is essential for packaging, but the pattern of NP binding to vRNA is unclear. Here we applied photoactivatable ribonucleoside enhanced cross-linking and immunoprecipitation (PAR-CLIP) to assess the native-state of NP-vRNA interactions in infected human cells. NP binds short fragments of RNA (~12 nucleotides) non-uniformly and without apparent sequence specificity. Moreover, NP binding is reduced at specific locations within the viral genome, including regions previously identified as required for viral genome segment packaging. Synonymous mutations designed to alter the predicted RNA structures in these low-NP-binding regions impact genome packaging and result in virus attenuation, whereas control mutations or mutagenesis of NP-bound regions have no effect. Finally, we demonstrate that the sequence conservation of low-NP-binding regions is required in multiple genome segments for propagation of diverse mammalian and avian IAV in host cells.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/genética , ARN Viral/genética , Proteínas de Unión al ARN/genética , Proteínas del Núcleo Viral/genética , Replicación Viral/genética , Animales , Secuencia Conservada , Perros , Genoma Viral , Células HEK293 , Humanos , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Células de Riñón Canino Madin Darby , Proteínas de la Nucleocápside , Mapeo Nucleótido , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas del Núcleo Viral/metabolismo
10.
Cell Host Microbe ; 24(2): 308-323.e6, 2018 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-30092202

RESUMEN

Pathogens have been a strong driving force for natural selection. Therefore, understanding how human genetic differences impact infection-related cellular traits can mechanistically link genetic variation to disease susceptibility. Here we report the Hi-HOST Phenome Project (H2P2): a catalog of cellular genome-wide association studies (GWAS) comprising 79 infection-related phenotypes in response to 8 pathogens in 528 lymphoblastoid cell lines. Seventeen loci surpass genome-wide significance for infection-associated phenotypes ranging from pathogen replication to cytokine production. We combined H2P2 with clinical association data from patients to identify a SNP near CXCL10 as a risk factor for inflammatory bowel disease. A SNP in the transcriptional repressor ZBTB20 demonstrated pleiotropy, likely through suppression of multiple target genes, and was associated with viral hepatitis. These data are available on a web portal to facilitate interpreting human genome variation through the lens of cell biology and should serve as a rich resource for the research community.


Asunto(s)
Biología Computacional/métodos , Predisposición Genética a la Enfermedad , Variación Genética , Genoma Humano , Estudio de Asociación del Genoma Completo/métodos , Infecciones , Fenotipo , Anticuerpos Monoclonales , Línea Celular , Quimiocina CXCL10/genética , Citocinas/genética , Citocinas/metabolismo , Análisis Mutacional de ADN , Replicación del ADN , Recolección de Datos , Bases de Datos Genéticas , Registros Electrónicos de Salud , Pleiotropía Genética , Estudio de Asociación del Genoma Completo/instrumentación , Hepatitis Viral Humana , Humanos , Enfermedades Inflamatorias del Intestino , Proteínas del Tejido Nervioso/genética , Factores de Riesgo , Factores de Transcripción/genética , Navegador Web
11.
Virology ; 494: 89-99, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27105450

RESUMEN

Replication and transmission of avian influenza virus in humans poses a pandemic threat. The molecular determinants that facilitate this process are not well understood. We used DBA/2 mice to identify viral factors that mediate the difference in pathogenesis between a virulent (H7N3) and a non-virulent (H7N9) avian influenza virus from North America. In vitro and in vivo characterization of reassortant viruses identified the PB2 and PA polymerase genes as major determinants of H7N3 pathogenesis. Analysis of individual residues in the PB2 and PA genes identified position 358 (E358V) in PB2 and positions 190 (P190S) and 400 (Q400P) in PA that reduced the virulence of H7N3 virus. The E358V and P190S substitutions also caused reduced inflammation after infection. Our results suggest that specific residues in the polymerase proteins PB2 and PA are important for replication and virulence of avian influenza viruses in a mammalian host.


Asunto(s)
Subtipo H7N3 del Virus de la Influenza A/genética , Subtipo H7N3 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/virología , ARN Polimerasa Dependiente del ARN/genética , Proteínas Virales/genética , Secuencia de Aminoácidos , Aminoácidos , Animales , Secuencia Conservada , Subtipo H7N9 del Virus de la Influenza A/genética , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Ratones , Ratones Endogámicos DBA , Infecciones por Orthomyxoviridae/mortalidad , ARN Polimerasa Dependiente del ARN/química , Virus Reordenados/genética , Virus Reordenados/patogenicidad , Índice de Severidad de la Enfermedad , Carga Viral , Proteínas Virales/química , Factores de Virulencia/genética , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA