Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.677
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 172(5): 952-965.e18, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29474921

RESUMEN

Viruses that are typically benign sometimes invade the brainstem in otherwise healthy children. We report bi-allelic DBR1 mutations in unrelated patients from different ethnicities, each of whom had brainstem infection due to herpes simplex virus 1 (HSV1), influenza virus, or norovirus. DBR1 encodes the only known RNA lariat debranching enzyme. We show that DBR1 expression is ubiquitous, but strongest in the spinal cord and brainstem. We also show that all DBR1 mutant alleles are severely hypomorphic, in terms of expression and function. The fibroblasts of DBR1-mutated patients contain higher RNA lariat levels than control cells, this difference becoming even more marked during HSV1 infection. Finally, we show that the patients' fibroblasts are highly susceptible to HSV1. RNA lariat accumulation and viral susceptibility are rescued by wild-type DBR1. Autosomal recessive, partial DBR1 deficiency underlies viral infection of the brainstem in humans through the disruption of tissue-specific and cell-intrinsic immunity to viruses.


Asunto(s)
Encefalopatías Metabólicas Innatas/genética , Tronco Encefálico/metabolismo , Tronco Encefálico/virología , ARN/química , ARN/metabolismo , Alelos , Secuencia de Aminoácidos , Animales , Encefalopatías Metabólicas Innatas/patología , Tronco Encefálico/patología , Encefalitis Viral/genética , Escherichia coli/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/virología , Herpesvirus Humano 1 , Humanos , Interferones/metabolismo , Intrones/genética , Masculino , Ratones , Proteínas Mutantes/metabolismo , Mutación/genética , Sistemas de Lectura Abierta/genética , Linaje , ARN Nucleotidiltransferasas/química , ARN Nucleotidiltransferasas/deficiencia , ARN Nucleotidiltransferasas/genética , Receptor Toll-Like 3/metabolismo , Replicación Viral
2.
Mol Cell ; 81(15): 3171-3186.e8, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34171297

RESUMEN

Accurate control of innate immune responses is required to eliminate invading pathogens and simultaneously avoid autoinflammation and autoimmune diseases. Here, we demonstrate that arginine monomethylation precisely regulates the mitochondrial antiviral-signaling protein (MAVS)-mediated antiviral response. Protein arginine methyltransferase 7 (PRMT7) forms aggregates to catalyze MAVS monomethylation at arginine residue 52 (R52), attenuating its binding to TRIM31 and RIG-I, which leads to the suppression of MAVS aggregation and subsequent activation. Upon virus infection, aggregated PRMT7 is disabled in a timely manner due to automethylation at arginine residue 32 (R32), and SMURF1 is recruited to PRMT7 by MAVS to induce proteasomal degradation of PRMT7, resulting in the relief of PRMT7 suppression of MAVS activation. Therefore, we not only reveal that arginine monomethylation by PRMT7 negatively regulates MAVS-mediated antiviral signaling in vitro and in vivo but also uncover a mechanism by which PRMT7 is tightly controlled to ensure the timely activation of antiviral defense.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Arginina/metabolismo , Interacciones Huésped-Patógeno/fisiología , Inmunidad Innata/fisiología , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Proteína 58 DEAD Box/metabolismo , Fibroblastos/virología , Células HEK293 , Herpes Simple/inmunología , Herpes Simple/metabolismo , Herpes Simple/virología , Humanos , Metilación , Ratones , Ratones Noqueados , Alcamidas Poliinsaturadas , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/inmunología , Receptores Inmunológicos/metabolismo , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/metabolismo , Infecciones por Respirovirus/virología , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Cell ; 151(7): 1569-80, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23260143

RESUMEN

Many signaling circuits face a fundamental tradeoff between accelerating their response speed while maintaining final levels below a cytotoxic threshold. Here, we describe a transcriptional circuitry that dynamically converts signaling inputs into faster rates without amplifying final equilibrium levels. Using time-lapse microscopy, we find that transcriptional activators accelerate human cytomegalovirus (CMV) gene expression in single cells without amplifying steady-state expression levels, and this acceleration generates a significant replication advantage. We map the accelerator to a highly self-cooperative transcriptional negative-feedback loop (Hill coefficient ∼7) generated by homomultimerization of the virus's essential transactivator protein IE2 at nuclear PML bodies. Eliminating the IE2-accelerator circuit reduces transcriptional strength through mislocalization of incoming viral genomes away from PML bodies and carries a heavy fitness cost. In general, accelerators may provide a mechanism for signal-transduction circuits to respond quickly to external signals without increasing steady-state levels of potentially cytotoxic molecules.


Asunto(s)
Infecciones por Citomegalovirus/virología , Citomegalovirus/genética , Redes Reguladoras de Genes , Aptitud Genética , Citomegalovirus/fisiología , Retroalimentación Fisiológica , Fibroblastos/virología , Regulación Viral de la Expresión Génica , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Cuerpos de Inclusión Viral/metabolismo , Imagen de Lapso de Tiempo , Transactivadores/metabolismo , Activación Transcripcional , Replicación Viral
4.
Mol Cell ; 76(3): 485-499.e8, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31495563

RESUMEN

Transcriptional responses to external stimuli remain poorly understood. Using global nuclear run-on followed by sequencing (GRO-seq) and precision nuclear run-on sequencing (PRO-seq), we show that CDK8 kinase activity promotes RNA polymerase II pause release in response to interferon-γ (IFN-γ), a universal cytokine involved in immunity and tumor surveillance. The Mediator kinase module contains CDK8 or CDK19, which are presumed to be functionally redundant. We implemented cortistatin A, chemical genetics, transcriptomics, and other methods to decouple their function while assessing enzymatic versus structural roles. Unexpectedly, CDK8 and CDK19 regulated different gene sets via distinct mechanisms. CDK8-dependent regulation required its kinase activity, whereas CDK19 governed IFN-γ responses through its scaffolding function (i.e., it was kinase independent). Accordingly, CDK8, not CDK19, phosphorylates the STAT1 transcription factor (TF) during IFN-γ stimulation, and CDK8 kinase inhibition blocked activation of JAK-STAT pathway TFs. Cytokines such as IFN-γ rapidly mobilize TFs to "reprogram" cellular transcription; our results implicate CDK8 and CDK19 as essential for this transcriptional reprogramming.


Asunto(s)
Quinasa 8 Dependiente de Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Fibroblastos/efectos de los fármacos , Interferón gamma/farmacología , Transcripción Genética/efectos de los fármacos , Animales , Quinasa 8 Dependiente de Ciclina/genética , Quinasas Ciclina-Dependientes/genética , Fibroblastos/enzimología , Fibroblastos/virología , Células HCT116 , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , ARN Polimerasa II/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Vesiculovirus/patogenicidad
5.
Proc Natl Acad Sci U S A ; 121(43): e2406773121, 2024 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-39401365

RESUMEN

Bats are tolerant to highly pathogenic viruses such as Marburg, Ebola, and Nipah, suggesting the presence of a unique immune tolerance toward viral infection. Here, we compared severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection of human and bat (Rhinolophus ferrumequinum) pluripotent cells and fibroblasts. Since bat cells do not express an angiotensin-converting enzyme 2 (ACE2) receptor that allows virus infection, we transduced the human ACE2 (hA) receptor into the cells and found that transduced cells can be infected with SARS-CoV-2. Compared to human embryonic stem cells-hA, infected bat induced Pluripotent Stem Cells (iPSCs)-hA produced about a 100-fold lower level of infectious virus and displayed lower toxicity. In contrast, bat embryonic fibroblast-hA produced no infectious virus while being infectable and synthesizing viral RNA and proteins, suggesting abortive infection. Indeed, electron microscopy failed to detect virus-like particles in infected bat fibroblasts in contrast to bat iPSCs or human cells, consistent with the latter producing infectious viruses. This suggests that bat somatic but not pluripotent cells have an effective mechanism to control virus replication. Consistent with previous results by others, we find that bat cells have a constitutively activated innate immune system, which might limit SARS-CoV-2 infection compared to human cells.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Quirópteros , Fibroblastos , SARS-CoV-2 , Quirópteros/virología , Fibroblastos/virología , Fibroblastos/metabolismo , Humanos , SARS-CoV-2/fisiología , SARS-CoV-2/inmunología , Animales , COVID-19/virología , COVID-19/inmunología , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Células Madre Pluripotentes Inducidas/virología , Células Madre Pluripotentes Inducidas/metabolismo , Replicación Viral
6.
Proc Natl Acad Sci U S A ; 121(39): e2408078121, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39292744

RESUMEN

The Pentamer complex of Human Cytomegalovirus (HCMV) consists of the viral glycoproteins gH, gL, UL128, UL130, and UL131 and is incorporated into infectious virions. HCMV strains propagated extensively in vitro in fibroblasts carry UL128, UL130, or UL131 alleles that do not make a functional complex and thus lack Pentamer function. Adding functional Pentamer to such strains decreases virus growth in fibroblasts. Here, we show that the Pentamer inhibits productive HCMV replication in fibroblasts by repressing viral Immediate Early (IE) transcription. We show that ectopic expression of the viral IE1 protein, a target of Pentamer-mediated transcriptional repression, complements the growth defect of a Pentamer-positive virus. Furthermore, we show that the Pentamer also represses viral IE transcription in cell types where HCMV in vitro latency is studied. Finally, we identify UL130 as a functional subunit of the Pentamer for IE transcriptional repression and demonstrate that cyclic AMP Response Element (CRE) and NFkB sites within the Major Immediate Early Promoter that drives IE1 transcription contribute to this repression. We conclude that the HCMV Pentamer represses viral IE transcription.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Fibroblastos , Proteínas Inmediatas-Precoces , Transcripción Genética , Proteínas del Envoltorio Viral , Humanos , Citomegalovirus/genética , Citomegalovirus/fisiología , Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Inmediatas-Precoces/genética , Fibroblastos/virología , Fibroblastos/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/genética , Regulación Viral de la Expresión Génica , Replicación Viral/genética , Glicoproteínas/metabolismo , Glicoproteínas/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Proteínas Virales/metabolismo , Proteínas Virales/genética , Genes Inmediatos-Precoces , Regiones Promotoras Genéticas
7.
PLoS Pathog ; 20(8): e1012468, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39146367

RESUMEN

Genetic editing of the germline using CRISPR/Cas9 technology has made it possible to alter livestock traits, including the creation of resistance to viral diseases. However, virus adaptability could present a major obstacle in this effort. Recently, chickens resistant to avian leukosis virus subgroup J (ALV-J) were developed by deleting a single amino acid, W38, within the ALV-J receptor NHE1 using CRISPR/Cas9 genome editing. This resistance was confirmed both in vitro and in vivo. In vitro resistance of W38-/- chicken embryonic fibroblasts to all tested ALV-J strains was shown. To investigate the capacity of ALV-J for further adaptation, we used a retrovirus reporter-based assay to select adapted ALV-J variants. We assumed that adaptive mutations overcoming the cellular resistance would occur within the envelope protein. In accordance with this assumption, we isolated and sequenced numerous adapted virus variants and found within their envelope genes eight independent single nucleotide substitutions. To confirm the adaptive capacity of these substitutions, we introduced them into the original retrovirus reporter. All eight variants replicated effectively in W38-/- chicken embryonic fibroblasts in vitro while in vivo, W38-/- chickens were sensitive to tumor induction by two of the variants. Importantly, receptor alleles with more extensive modifications have remained resistant to the virus. These results demonstrate an important strategy in livestock genome engineering towards antivirus resistance and illustrate that cellular resistance induced by minor receptor modifications can be overcome by adapted virus variants. We conclude that more complex editing will be necessary to attain robust resistance.


Asunto(s)
Virus de la Leucosis Aviar , Leucosis Aviar , Pollos , Animales , Virus de la Leucosis Aviar/genética , Virus de la Leucosis Aviar/fisiología , Pollos/virología , Leucosis Aviar/virología , Leucosis Aviar/genética , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/genética , Resistencia a la Enfermedad/genética , Sistemas CRISPR-Cas , Edición Génica , Embrión de Pollo , Evolución Molecular , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Fibroblastos/virología , Fibroblastos/metabolismo
8.
PLoS Pathog ; 20(8): e1012426, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39110744

RESUMEN

Merkel cell polyomavirus (MCPyV) is the causative agent of the majority of Merkel cell carcinomas (MCC). The virus has limited coding capacity, with its early viral proteins, large T (LT) and small T (sT), being multifunctional and contributing to infection and transformation. A fundamental difference in early viral gene expression between infection and MCPyV-driven tumorigenesis is the expression of a truncated LT (LTtr) in the tumor. In contrast, sT is expressed in both conditions and contributes significantly to oncogenesis. Here, we identified novel functions of early viral proteins by performing genome-wide transcriptome and chromatin studies in primary human fibroblasts. Due to current limitations in infection and tumorigenesis models, we mimic these conditions by ectopically expressing sT, LT or LTtr, individually or in combination, at different time points. In addition to its known function in cell cycle and inflammation modulation, we reveal a fundamentally new function of sT. We show that sT regulates the type I interferon (IFN) response downstream of the type I interferon receptor (IFNAR) by interfering with the interferon-stimulated gene factor 3 (ISGF3)-induced interferon-stimulated gene (ISG) response. Expression of sT leads to a reduction in the expression of interferon regulatory factor 9 (IRF9) which is a central component of the ISGF3 complex. We further show that this function of sT is conserved in BKPyV. We provide a first mechanistic understanding of which early viral proteins trigger and control the type I IFN response, which may influence MCPyV infection, persistence and, during MCC progression, regulation of the tumor microenvironment.


Asunto(s)
Carcinoma de Células de Merkel , Evasión Inmune , Interferón Tipo I , Poliomavirus de Células de Merkel , Infecciones por Polyomavirus , Transducción de Señal , Infecciones Tumorales por Virus , Humanos , Poliomavirus de Células de Merkel/inmunología , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Carcinoma de Células de Merkel/virología , Carcinoma de Células de Merkel/inmunología , Transducción de Señal/inmunología , Infecciones por Polyomavirus/inmunología , Infecciones por Polyomavirus/virología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología , Evasión Inmune/inmunología , Antígenos Virales de Tumores/metabolismo , Antígenos Virales de Tumores/inmunología , Antígenos Virales de Tumores/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/virología , Neoplasias Cutáneas/metabolismo , Fibroblastos/virología , Fibroblastos/metabolismo , Fibroblastos/inmunología
9.
J Immunol ; 213(2): 187-203, 2024 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-38829131

RESUMEN

The RING finger (RNF) family, a group of E3 ubiquitin ligases, plays multiple essential roles in the regulation of innate immunity and resistance to viral infection in mammals. However, it is still unclear whether RNF proteins affect the production of IFN-I and the replication of avian influenza virus (AIV) in ducks. In this article, we found that duck RNF216 (duRNF216) inhibited the duRIG-I signaling pathway. Conversely, duRNF216 deficiency enhanced innate immune responses in duck embryonic fibroblasts. duRNF216 did not interacted with duRIG-I, duMDA5, duMAVS, duSTING, duTBK1, or duIRF7 in the duck RIG-I pathway. However, duRNF216 targeted duTRAF3 and inhibited duMAVS in the recruitment of duTRAF3 in a dose-dependent manner. duRNF216 catalyzed K48-linked polyubiquitination of duck TRAF3, which was degraded by the proteasome pathway. Additionally, AIV PB1 protein competed with duTRAF3 for binding to duRNF216 to reduce degradation of TRAF3 by proteasomes in the cytoplasm, thereby slightly weakening duRNF216-mediated downregulation of IFN-I. Moreover, although duRNF216 downregulated the IFN-ß expression during virus infection, the expression level of IFN-ß in AIV-infected duck embryonic fibroblasts overexpressing duRNF216 was still higher than that in uninfected cells, which would hinder the viral replication. During AIV infection, duRNF216 protein targeted the core protein PB1 of viral polymerase to hinder viral polymerase activity and viral RNA synthesis in the nucleus, ultimately strongly restricting viral replication. Thus, our study reveals a new mechanism by which duRNF216 downregulates innate immunity and inhibits AIV replication in ducks. These findings broaden our understanding of the mechanisms by which the duRNF216 protein affects AIV replication in ducks.


Asunto(s)
Patos , Inmunidad Innata , Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Transducción de Señal , Ubiquitina-Proteína Ligasas , Replicación Viral , Animales , Patos/inmunología , Patos/virología , Replicación Viral/inmunología , Transducción de Señal/inmunología , Gripe Aviar/inmunología , Gripe Aviar/virología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/fisiología , Inmunidad Innata/inmunología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología , Fibroblastos/inmunología , Fibroblastos/virología , Proteínas Aviares/inmunología , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Ubiquitinación , Proteína 58 DEAD Box/metabolismo , Proteína 58 DEAD Box/inmunología
10.
Mol Cell ; 71(5): 825-840.e6, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30100266

RESUMEN

Virulent pathogens often cause the release of host-derived damage-associated molecular patterns (DAMPs) from infected cells. During encounters with immune-evasive viruses that block inflammatory gene expression, preformed DAMPs provide backup inflammatory signals that ensure protective immunity. Whether DAMPs exhibit additional backup defense activities is unknown. Herein, we report that viral infection of barrier epithelia (keratinocytes) elicits the release of preformed interleukin-1 (IL-1) family cytokines, including the DAMP IL-1α. Mechanistic studies revealed that IL-1 acts on skin fibroblasts to induce an interferon (IFN)-like state that restricts viral replication. We identified a branch in the IL-1 signaling pathway that induces IFN-stimulated gene expression in infected cells and found that IL-1 signaling is necessary to restrict viral replication in human skin explants. These activities are most important to control immune-evasive virus replication in fibroblasts and other barrier cell types. These findings highlight IL-1 as an important backup antiviral system to ensure barrier defense.


Asunto(s)
Evasión Inmune/inmunología , Interleucina-1/inmunología , Transducción de Señal/inmunología , Replicación Viral/inmunología , Animales , Línea Celular , Chlorocebus aethiops , Femenino , Fibroblastos/inmunología , Fibroblastos/virología , Expresión Génica/inmunología , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Piel/inmunología , Piel/virología , Células Vero
11.
Mol Cell ; 72(5): 862-874.e5, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30318442

RESUMEN

mRNAs carry two layers of information, the genetic code and the information that dictates their post-transcriptional fate. The latter function relies on a complex interplay between cis-elements and trans-regulators, and unbiased identification of these elements is still challenging. To identify cis-elements that control gene expression, we use dimethyl sulfate (DMS) mutational profiling with sequencing and map changes in mRNA secondary structure following viral infection. Our dynamic structural data reveal a major role for ribosomes in unwinding secondary structures, which is further supported by the relationship we uncover between structure and translation efficiency. Moreover, our analysis revealed dozens of regions in viral and cellular mRNAs that exhibit changes in secondary structure. In-depth analysis of these regions reveals cis-elements in 3' UTRs that regulate mRNA stability and elements within coding sequences that control translation. Overall, our study demonstrates how mapping dynamic changes in mRNA structure allows unbiased identification of functional regulatory elements.


Asunto(s)
Citomegalovirus/genética , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Fosfoproteínas/genética , Biosíntesis de Proteínas , ARN Mensajero/química , Proteínas de la Matriz Viral/genética , Línea Celular , Citomegalovirus/efectos de los fármacos , Citomegalovirus/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/virología , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Mutágenos/farmacología , Conformación de Ácido Nucleico , Fosfoproteínas/metabolismo , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Transducción de Señal , Ésteres del Ácido Sulfúrico/farmacología , Proteínas de la Matriz Viral/metabolismo
12.
J Virol ; 98(7): e0011024, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38837381

RESUMEN

We determined the transcription profile of adeno-associated virus type 2 (AAV2)-infected primary human fibroblasts. Subsequent analysis revealed that cells respond to AAV infection through changes in several significantly affected pathways, including cell cycle regulation, chromatin modulation, and innate immune responses. Various assays were performed to validate selected differentially expressed genes and to confirm not only the quality but also the robustness of the raw data. One of the genes upregulated in AAV2-infected cells was interferon-γ inducible factor 16 (IFI16). IFI16 is known as a multifunctional cytosolic and nuclear innate immune sensor for double-stranded as well as single-stranded DNA, exerting its effects through various mechanisms, such as interferon response, epigenetic modifications, or transcriptional regulation. IFI16 thereby constitutes a restriction factor for many different viruses among them, as shown here, AAV2 and thereof derived vectors. Indeed, the post-transcriptional silencing of IFI16 significantly increased AAV2 transduction efficiency, independent of the structure of the virus/vector genome. We also show that IFI16 exerts its inhibitory effect on AAV2 transduction in an immune-modulatory independent way by interfering with Sp1-dependent transactivation of wild-type AAV2 and AAV2 vector promoters. IMPORTANCE: Adeno-associated virus (AAV) vectors are among the most frequently used viral vectors for gene therapy. The lack of pathogenicity of the parental virus, the long-term persistence as episomes in non-proliferating cells, and the availability of a variety of AAV serotypes differing in their cellular tropism are advantageous features of this biological nanoparticle. To deepen our understanding of virus-host interactions, especially in terms of antiviral responses, we present here the first transcriptome analysis of AAV serotype 2 (AAV2)-infected human primary fibroblasts. Our findings indicate that interferon-γ inducible factor 16 acts as an antiviral factor in AAV2 infection and AAV2 vector-mediated cell transduction in an immune-modulatory independent way by interrupting the Sp1-dependent gene expression from viral or vector genomes.


Asunto(s)
Dependovirus , Fibroblastos , Proteínas Nucleares , Fosfoproteínas , Transducción Genética , Humanos , Dependovirus/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Fibroblastos/virología , Fibroblastos/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Inmunidad Innata , Vectores Genéticos/genética , Parvovirinae/genética , Células Cultivadas
13.
J Virol ; 98(7): e0079124, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38940584

RESUMEN

Fibrocytes were reported to be host cells for HIV-1, but the immunological recognition of HIV-1-infected fibrocytes has not been studied. Here, we investigated the recognition of HIV-1-infected fibrocytes by HIV-1-specific CD8+ T cells. CD8+ T cells specific for five HIV-1 epitopes (HLA-A*24:02-restricted, HLA-B*52:01-restricted, and HLA-C*12:02-restricted epitopes) produced IFN-γ and expressed CD107a after coculture with HIV-1-infected fibrocytes. HIV-1-infected fibrocytes were effectively killed by HIV-1-specific CD8+ T cells. Although it is well known that HIV-1 Nef-mediated downregulation of HLA-A and HLA-B critically affects the T cell recognition of HIV-1-infected CD4+ T cells and HIV-1-infected macrophages, Nef downregulated HLA-A, but not HLA-B, in HIV-1-infected fibrocytes. These findings suggested that HIV-1-specific CD8+ T cells could recognize HIV-1-infected fibrocytes more strongly than HIV-1-infected CD4+ T cells or HIV-1-infected macrophages. HIV-1-infected fibrocytes were also recognized by HIV-1-specific HLA-DR-restricted T cells, indicating that HIV-1-infected fibrocytes can present HIV-1 epitopes to helper T cells. Collectively, these findings suggest that fibrocytes have an important role as antigen-presenting cells during HIV-1 infection. The present study demonstrates effective recognition of HIV-1-infected fibrocytes by HIV-1-specific T cells and suggests possible roles of fibrocytes in the induction and maintenance of HIV-1-specific T cells. IMPORTANCE: Fibrocytes were identified as unique hematopoietic cells with the features of both macrophages and fibroblasts and were demonstrated to be host cells for HIV-1. However, T cell recognition of HIV-1-infected fibrocytes has not been studied. We investigated the recognition of HIV-1-infected fibrocytes by HIV-1-specific T cells. HIV-1-infected fibrocytes were effectively recognized and killed by CD8+ T cells specific for HIV-1 epitopes presented by HLA-A, HLA-B, or HLA-C and were recognized by HIV-1-specific HLA-DR-restricted CD4+ T cells. HIV-1 Nef-mediated downregulation of HLA-A and HLA-B was found in HIV-1-infected CD4+ T cells, whereas Nef did not downregulate HLA-B in HIV-1-infected fibrocytes. These results suggest that HIV-1-specific CD8+ T cells recognize HIV-1-infected fibrocytes more strongly than HIV-1-infected CD4+ T cells. The present study suggests the importance of fibrocytes in the induction and maintenance of HIV-1-specific T cells.


Asunto(s)
Linfocitos T CD8-positivos , Regulación hacia Abajo , Infecciones por VIH , VIH-1 , Antígenos HLA-B , Productos del Gen nef del Virus de la Inmunodeficiencia Humana , Humanos , VIH-1/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Antígenos HLA-B/inmunología , Antígenos HLA-B/metabolismo , Fibroblastos/virología , Fibroblastos/inmunología , Fibroblastos/metabolismo , Linfocitos T CD4-Positivos/inmunología , Epítopos de Linfocito T/inmunología , Macrófagos/inmunología , Macrófagos/virología , Macrófagos/metabolismo
14.
J Virol ; 98(3): e0127823, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38345384

RESUMEN

Cytomegalovirus (CMV), a type of herpes virus, is the predominant cause of congenital anomalies due to intrauterine infections in humans. Adverse outcomes related to intrauterine infections with human cytomegalovirus (HCMV) vary widely, depending on factors such as fetal infection timing, infection route, and viral virulence. The precise mechanism underlying HCMV susceptibility remains unclear. In this study, we compared the susceptibility of neonatal human dermal fibroblast cells (NHDFCs) and human induced pluripotent stem cells (hiPSCs) derived from NHDFCs, which are genetically identical to HCMV, using immunostaining, microarray, in situ hybridization, quantitative PCR, and scanning electron microscopy. These cells were previously used to compare CMV susceptibility, but the underlying mechanisms were not fully elucidated. HCMV susceptibility of hiPSCs was significantly lower in the earliest phase. No shared gene ontologies were observed immediately post-infection between the two cell types using microarray analysis. Early-stage expression of HCMV antigens and the HCMV genome was minimal in immunostaining and in in situ hybridization in hiPSCs. This strongly suggests that HCMV does not readily bind to hiPSC surfaces. Scanning electron microscopy performed using the NanoSuit method confirmed the scarcity of HCMV particles on hiPSC surfaces. The zeta potential and charge mapping of the charged surface in NHDFCs and hiPSCs exhibited minimal differences when assessed using zeta potential analyzer and scanning ion conductance microscopy; however, the expression of heparan sulfate (HS) was significantly lower in hiPSCs compared with that in NHDFCs. Thus, HS expression could be a primary determinant of HCMV resistance in hiPSCs at the attachment level. IMPORTANCE: Numerous factors such as attachment, virus particle entry, transcription, and virus particle egress can affect viral susceptibility. Since 1984, pluripotent cells are known to be CMV resistant; however, the exact mechanism underlying this resistance remains elusive. Some researchers suggest inhibition in the initial phase of HCMV binding, while others have suggested the possibility of a sufficient amount of HCMV entering the cells to establish latency. This study demonstrates that HCMV particles rarely attach to the surfaces of hiPSCs. This is not due to limitations in the electrostatic interactions between the surface of hiPSCs and HCMV particles, but due to HS expression. Therefore, HS expression should be recognized as a key factor in determining the susceptibility of HCMV in congenital infection in vitro and in vivo. In the future, drugs targeting HS may become crucial for the treatment of congenital CMV infections. Thus, further research in this area is warranted.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Heparitina Sulfato , Células Madre Pluripotentes Inducidas , Humanos , Recién Nacido , Membrana Celular/química , Membrana Celular/metabolismo , Citomegalovirus/fisiología , Heparitina Sulfato/análisis , Heparitina Sulfato/metabolismo , Infecciones por Herpesviridae , Células Madre Pluripotentes Inducidas/química , Células Madre Pluripotentes Inducidas/metabolismo , Fibroblastos/química , Fibroblastos/metabolismo , Fibroblastos/virología , Piel/citología
15.
J Virol ; 98(10): e0102424, 2024 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-39269177

RESUMEN

Currently, there are no specific antiviral therapeutic approaches targeting Human papillomaviruses (HPVs), which cause around 5% of all human cancers. Specific antiviral reagents are particularly needed for HPV-related oropharyngeal cancers (HPV+OPCs) whose incidence is increasing and for which there are no early diagnostic tools available. We and others have demonstrated that the estrogen receptor alpha (ERα) is overexpressed in HPV+OPCs, compared to HPV-negative cancers in this region, and that these elevated levels are associated with an improved disease outcome. Utilizing this HPV+-specific overexpression profile, we previously demonstrated that estrogen attenuates the growth and cell viability of HPV+ keratinocytes and HPV+ cancer cells in vitro. Expansion of this work in vivo failed to replicate this sensitization. The role of stromal support from the tumor microenvironment (TME) has previously been tied to both the HPV lifecycle and in vivo therapeutic responses. Our investigations revealed that in vitro co-culture with fibroblasts attenuated HPV+-specific estrogen growth responses. Continuing to monopolize on the HPV+-specific overexpression of ERα, our co-culture models then assessed the suitability of the selective estrogen receptor modulators (SERMs), raloxifene and tamoxifen, and showed growth attenuation in a variety of our models to one or both of these drugs in vitro. Utilization of these SERMs in vivo closely resembled the sensitization predicted by our co-culture models. Therefore, the in vitro fibroblast co-culture model better predicts in vivo responses. We propose that utilization of our co-culture in vitro model can accelerate cancer therapeutic drug discovery. IMPORTANCE: Human papillomavirus-related cancers (HPV+ cancers) remain a significant public health concern, and specific clinical approaches are desperately needed. In translating drug response data from in vitro to in vivo, the fibroblasts of the adjacent stromal support network play a key role. Our study presents the utilization of a fibroblast 2D co-culture system to better predict translational drug assessments for HPV+ cancers. We also suggest that this co-culture system should be considered for other translational approaches. Predicting even a portion of treatment paradigms that may fail in vivo with a co-culture model will yield significant time, effort, resource, and cost efficiencies.


Asunto(s)
Técnicas de Cocultivo , Receptor alfa de Estrógeno , Fibroblastos , Infecciones por Papillomavirus , Humanos , Receptor alfa de Estrógeno/metabolismo , Infecciones por Papillomavirus/virología , Infecciones por Papillomavirus/metabolismo , Fibroblastos/virología , Fibroblastos/metabolismo , Microambiente Tumoral , Neoplasias Orofaríngeas/virología , Neoplasias Orofaríngeas/metabolismo , Papillomaviridae/efectos de los fármacos , Papillomaviridae/fisiología , Queratinocitos/virología , Queratinocitos/metabolismo , Células del Estroma/metabolismo , Células del Estroma/virología , Estrógenos/metabolismo , Estrógenos/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Virus del Papiloma Humano
16.
J Virol ; 98(7): e0021324, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38832789

RESUMEN

Human cytomegalovirus (HCMV) is a ß-herpesvirus that poses severe disease risk for immunocompromised patients who experience primary infection or reactivation. Development and optimization of safe and effective anti-HCMV therapeutics is of urgent necessity for the prevention and treatment of HCMV-associated diseases in diverse populations. The use of neutralizing monoclonal antibodies (mAbs) to limit HCMV infection poses a promising therapeutic strategy, as anti-HCMV mAbs largely inhibit infection by targeting virion glycoprotein complexes. In contrast, the small-molecule compounds currently approved for patients (e.g., ganciclovir, letermovir, and maribavir) target later stages of the HCMV life cycle. Here, we present a broadly neutralizing human mAb, designated 1C10, elicited from a VelocImmune mouse immunized with infectious HCMV particles. Clone 1C10 neutralizes infection after virion binding to cells by targeting gH/gL envelope complexes and potently reduces infection of diverse HCMV strains in fibroblast, trophoblast, and epithelial cells. Antibody competition assays found that 1C10 recognizes a region of gH associated with broad neutralization and binds to soluble pentamer in the low nanomolar range. Importantly, 1C10 treatment significantly reduced virus proliferation in both fibroblast and epithelial cells. Further, the combination treatment of mAb 1C10 with ganciclovir reduced HCMV infection and proliferation in a synergistic manner. This work characterizes a neutralizing human mAb for potential use as a HCMV treatment, as well as a possible therapeutic strategy utilizing combination-based treatments targeting disparate steps of the viral life cycle. Collectively, the findings support an antibody-based therapy to effectively treat patients at risk for HCMV-associated diseases. IMPORTANCE: Human cytomegalovirus is a herpesvirus that infects a large proportion of the population and can cause significant disease in diverse patient populations whose immune systems are suppressed or compromised. The development and optimization of safe anti-HCMV therapeutics, especially those that have viral targets and inhibition mechanisms different from current HCMV treatments, are of urgent necessity to better public health. Human monoclonal antibodies (mAbs) that prevent HCMV entry of cells were identified by immunizing transgenic mice and screened for broad and effective neutralization capability. Here, we describe one such mAb, which was found to target gH/gL envelope complexes and effectively limit HCMV infection and dissemination. Further, administration of the antibody in combination with the antiviral drug ganciclovir inhibited HCMV in a synergistic manner, highlighting this approach and the use of anti-HCMV mAbs more broadly, as a potential therapeutic strategy for the treatment of diverse patient populations.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Antivirales , Infecciones por Citomegalovirus , Citomegalovirus , Ratones Transgénicos , Proteínas del Envoltorio Viral , Animales , Humanos , Citomegalovirus/inmunología , Citomegalovirus/efectos de los fármacos , Ratones , Anticuerpos Antivirales/inmunología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/prevención & control , Infecciones por Citomegalovirus/virología , Proteínas del Envoltorio Viral/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Virión/inmunología , Fibroblastos/virología , Replicación Viral/efectos de los fármacos , Anticuerpos ampliamente neutralizantes/inmunología , Antivirales/farmacología , Inmunización
17.
Nat Immunol ; 14(2): 179-85, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23263557

RESUMEN

The innate immune system senses viral DNA that enters mammalian cells, or in aberrant situations self-DNA, and triggers type I interferon production. Here we present an integrative approach that combines quantitative proteomics, genomics and small molecule perturbations to identify genes involved in this pathway. We silenced 809 candidate genes, measured the response to dsDNA and connected resulting hits with the known signaling network. We identified ABCF1 as a critical protein that associates with dsDNA and the DNA-sensing components HMGB2 and IFI204. We also found that CDC37 regulates the stability of the signaling molecule TBK1 and that chemical inhibition of the CDC37-HSP90 interaction and several other pathway regulators potently modulates the innate immune response to DNA and retroviral infection.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/inmunología , ADN Viral/inmunología , Células Dendríticas/inmunología , Fibroblastos/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Inmunidad Innata , Transportadoras de Casetes de Unión a ATP/genética , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/inmunología , Chaperoninas/antagonistas & inhibidores , Chaperoninas/genética , Chaperoninas/inmunología , Citosol/efectos de los fármacos , Citosol/metabolismo , Citosol/virología , ADN Viral/genética , Células Dendríticas/efectos de los fármacos , Células Dendríticas/virología , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Regulación de la Expresión Génica/inmunología , Silenciador del Gen , VIH-1/fisiología , Proteína HMGB2/genética , Proteína HMGB2/inmunología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/inmunología , Humanos , Ratones , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteómica , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Bibliotecas de Moléculas Pequeñas/farmacología , Vesiculovirus/fisiología
18.
Immunity ; 44(5): 1151-61, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27178468

RESUMEN

The transcription factor IRF-3 mediates cellular antiviral response by inducing the expression of interferon and other antiviral proteins. In RNA-virus infected cells, IRF-3's transcriptional activation is triggered primarily by RIG-I-like receptors (RLR), which can also activate the RLR-induced IRF-3-mediated pathway of apoptosis (RIPA). Here, we have reported that the pathway of IRF-3 activation in RIPA was independent of and distinct from the known pathway of transcriptional activation of IRF-3. It required linear polyubiquitination of two specific lysine residues of IRF-3 by LUBAC, the linear polyubiquitinating enzyme complex, which bound IRF-3 in signal-dependent fashion. To evaluate the role of RIPA in viral pathogenesis, we engineered a genetically targeted mouse, which expressed a mutant IRF-3 that was RIPA-competent but transcriptionally inert; this single-action IRF-3 could protect mice from lethal viral infection. Our observations indicated that IRF-3-mediated apoptosis of virus-infected cells could be an effective antiviral mechanism, without expression of the interferon-stimulated genes.


Asunto(s)
Apoptosis , Fibroblastos/inmunología , Factor 3 Regulador del Interferón/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Infecciones por Virus ARN/inmunología , Animales , Línea Celular , Clonación Molecular , Fibroblastos/virología , Humanos , Factor 3 Regulador del Interferón/genética , Ratones , Ratones Transgénicos , Mutación/genética , Receptores de Superficie Celular , Transducción de Señal , Activación Transcripcional , Ubiquitinación
19.
Nature ; 571(7765): 419-423, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31292545

RESUMEN

Single-cell RNA sequencing (scRNA-seq) has highlighted the important role of intercellular heterogeneity in phenotype variability in both health and disease1. However, current scRNA-seq approaches provide only a snapshot of gene expression and convey little information on the true temporal dynamics and stochastic nature of transcription. A further key limitation of scRNA-seq analysis is that the RNA profile of each individual cell can be analysed only once. Here we introduce single-cell, thiol-(SH)-linked alkylation of RNA for metabolic labelling sequencing (scSLAM-seq), which integrates metabolic RNA labelling2, biochemical nucleoside conversion3 and scRNA-seq to record transcriptional activity directly by differentiating between new and old RNA for thousands of genes per single cell. We use scSLAM-seq to study the onset of infection with lytic cytomegalovirus in single mouse fibroblasts. The cell-cycle state and dose of infection deduced from old RNA enable dose-response analysis based on new RNA. scSLAM-seq thereby both visualizes and explains differences in transcriptional activity at the single-cell level. Furthermore, it depicts 'on-off' switches and transcriptional burst kinetics in host gene expression with extensive gene-specific differences that correlate with promoter-intrinsic features (TBP-TATA-box interactions and DNA methylation). Thus, gene-specific, and not cell-specific, features explain the heterogeneity in transcriptomes between individual cells and the transcriptional response to perturbations.


Asunto(s)
Regulación de la Expresión Génica/genética , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual , Transcripción Genética/genética , Alquilación , Animales , Ciclo Celular , Citomegalovirus/fisiología , Metilación de ADN , Fibroblastos/metabolismo , Fibroblastos/virología , Cinética , Ratones , Regiones Promotoras Genéticas/genética , ARN/análisis , ARN/química , Compuestos de Sulfhidrilo/química
20.
Nature ; 574(7777): 259-263, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31554973

RESUMEN

Chikungunya virus (CHIKV) is a re-emerging alphavirus that is transmitted to humans by mosquito bites and causes musculoskeletal and joint pain1,2. Despite intensive investigations, the human cellular factors that are critical for CHIKV infection remain unknown, hampering the understanding of viral pathogenesis and the development of anti-CHIKV therapies. Here we identified the four-and-a-half LIM domain protein 1 (FHL1)3 as a host factor that is required for CHIKV permissiveness and pathogenesis in humans and mice. Ablation of FHL1 expression results in the inhibition of infection by several CHIKV strains and o'nyong-nyong virus, but not by other alphaviruses and flaviviruses. Conversely, expression of FHL1 promotes CHIKV infection in cells that do not normally express it. FHL1 interacts directly with the hypervariable domain of the nsP3 protein of CHIKV and is essential for the replication of viral RNA. FHL1 is highly expressed in CHIKV-target cells and is particularly abundant in muscles3,4. Dermal fibroblasts and muscle cells derived from patients with Emery-Dreifuss muscular dystrophy that lack functional FHL15 are resistant to CHIKV infection. Furthermore,  CHIKV infection  is undetectable in Fhl1-knockout mice. Overall, this study shows that FHL1 is a key factor expressed by the host that enables CHIKV infection and identifies the interaction between nsP3 and FHL1 as a promising target for the development of anti-CHIKV therapies.


Asunto(s)
Fiebre Chikungunya/virología , Virus Chikungunya/patogenicidad , Factores Celulares Derivados del Huésped/metabolismo , Interacciones Huésped-Patógeno , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas Musculares/metabolismo , Animales , Células Cultivadas , Fiebre Chikungunya/tratamiento farmacológico , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/genética , Virus Chikungunya/crecimiento & desarrollo , Femenino , Fibroblastos/virología , Células HEK293 , Factores Celulares Derivados del Huésped/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM/deficiencia , Proteínas con Dominio LIM/genética , Masculino , Ratones , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Mioblastos/virología , Virus O'nyong-nyong/crecimiento & desarrollo , Virus O'nyong-nyong/patogenicidad , Unión Proteica , ARN Viral/biosíntesis , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA