Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nat Commun ; 15(1): 4696, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38824133

RESUMEN

Age-related microangiopathy, also known as small vessel disease (SVD), causes damage to the brain, retina, liver, and kidney. Based on the DNA damage theory of aging, we reasoned that genomic instability may underlie an SVD caused by dominant C-terminal variants in TREX1, the most abundant 3'-5' DNA exonuclease in mammals. C-terminal TREX1 variants cause an adult-onset SVD known as retinal vasculopathy with cerebral leukoencephalopathy (RVCL or RVCL-S). In RVCL, an aberrant, C-terminally truncated TREX1 mislocalizes to the nucleus due to deletion of its ER-anchoring domain. Since RVCL pathology mimics that of radiation injury, we reasoned that nuclear TREX1 would cause DNA damage. Here, we show that RVCL-associated TREX1 variants trigger DNA damage in humans, mice, and Drosophila, and that cells expressing RVCL mutant TREX1 are more vulnerable to DNA damage induced by chemotherapy and cytokines that up-regulate TREX1, leading to depletion of TREX1-high cells in RVCL mice. RVCL-associated TREX1 mutants inhibit homology-directed repair (HDR), causing DNA deletions and vulnerablility to PARP inhibitors. In women with RVCL, we observe early-onset breast cancer, similar to patients with BRCA1/2 variants. Our results provide a mechanistic basis linking aberrant TREX1 activity to the DNA damage theory of aging, premature senescence, and microvascular disease.


Asunto(s)
Daño del ADN , Exodesoxirribonucleasas , Fosfoproteínas , Animales , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Humanos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ratones , Reparación del ADN por Recombinación , Fenotipo , Mutación , Drosophila/genética , Envejecimiento/genética , Envejecimiento/metabolismo , Femenino , Drosophila melanogaster/genética , Masculino , Enfermedades de la Retina , Enfermedades Vasculares , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias
2.
J Virol ; 96(4): e0151021, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34935440

RESUMEN

Recent studies have demonstrated that the signaling activity of the cytosolic pathogen sensor retinoic acid-inducible gene-I (RIG-I) is modulated by a variety of posttranslational modifications (PTMs) to fine-tune the antiviral type I interferon (IFN) response. Whereas K63-linked ubiquitination of the RIG-I caspase activation and recruitment domains (CARDs) catalyzed by TRIM25 or other E3 ligases activates RIG-I, phosphorylation of RIG-I at S8 and T170 represses RIG-I signal transduction by preventing the TRIM25-RIG-I interaction and subsequent RIG-I ubiquitination. While strategies to suppress RIG-I signaling by interfering with its K63-polyubiquitin-dependent activation have been identified for several viruses, evasion mechanisms that directly promote RIG-I phosphorylation to escape antiviral immunity are unknown. Here, we show that the serine/threonine (Ser/Thr) kinase US3 of herpes simplex virus 1 (HSV-1) binds to RIG-I and phosphorylates RIG-I specifically at S8. US3-mediated phosphorylation suppressed TRIM25-mediated RIG-I ubiquitination, RIG-I-MAVS binding, and type I IFN induction. We constructed a mutant HSV-1 encoding a catalytically-inactive US3 protein (K220A) and found that, in contrast to the parental virus, the US3 mutant HSV-1 was unable to phosphorylate RIG-I at S8 and elicited higher levels of type I IFNs, IFN-stimulated genes (ISGs), and proinflammatory cytokines in a RIG-I-dependent manner. Finally, we show that this RIG-I evasion mechanism is conserved among the alphaherpesvirus US3 kinase family. Collectively, our study reveals a novel immune evasion mechanism of herpesviruses in which their US3 kinases phosphorylate the sensor RIG-I to keep it in the signaling-repressed state. IMPORTANCE Herpes simplex virus 1 (HSV-1) establishes lifelong latency in the majority of the human population worldwide. HSV-1 occasionally reactivates to produce infectious virus and to facilitate dissemination. While often remaining subclinical, both primary infection and reactivation occasionally cause debilitating eye diseases, which can lead to blindness, as well as life-threatening encephalitis and newborn infections. To identify new therapeutic targets for HSV-1-induced diseases, it is important to understand the HSV-1-host interactions that may influence infection outcome and disease. Our work uncovered direct phosphorylation of the pathogen sensor RIG-I by alphaherpesvirus-encoded kinases as a novel viral immune escape strategy and also underscores the importance of RNA sensors in surveilling DNA virus infection.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Herpesvirus Humano 1/inmunología , Evasión Inmune , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Inmunológicos/metabolismo , Proteínas Virales/metabolismo , Alphaherpesvirinae/genética , Alphaherpesvirinae/metabolismo , Alphaherpesvirinae/fisiología , Secuencia de Aminoácidos , Proteína 58 DEAD Box/química , Células HEK293 , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Receptores Inmunológicos/química , Proteínas Virales/genética
3.
mBio ; 12(2)2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33849980

RESUMEN

JC polyomavirus (JCV), a DNA virus that leads to persistent infection in humans, is the causative agent of progressive multifocal leukoencephalopathy, a lethal brain disease that affects immunocompromised individuals. Almost nothing is currently known about how JCV infection is controlled by the innate immune response and, further, whether JCV has evolved mechanisms to antagonize antiviral immunity. Here, we show that the innate immune sensors retinoic acid-inducible gene I (RIG-I) and cGMP-AMP synthase (cGAS) control JCV replication in human astrocytes. We further identify that the small t antigen (tAg) of JCV functions as an interferon (IFN) antagonist by suppressing RIG-I-mediated signal transduction. JCV tAg interacts with the E3 ubiquitin ligase TRIM25, thereby preventing its ability to bind RNA and to induce the K63-linked ubiquitination of RIG-I, which is known to facilitate RIG-I-mediated cytokine responses. Antagonism of RIG-I K63-linked ubiquitination and antiviral signaling is also conserved in the tAg of the related polyomavirus BK virus (BKV). These findings highlight how JCV and BKV manipulate a key innate surveillance pathway, which may stimulate research into designing novel therapies.IMPORTANCE The innate immune response is the first line of defense against viral pathogens, and in turn, many viruses have evolved strategies to evade detection by the host's innate immune surveillance machinery. Investigation of the interplay between viruses and the innate immune response provides valuable insight into potential therapeutic targets against viral infectious diseases. JC polyomavirus (JCV) is associated with a lifelong, persistent infection that can cause a rare neurodegenerative disease, called progressive multifocal leukoencephalopathy, in individuals that are immunosuppressed. The molecular mechanisms of JCV infection and persistence are not well understood, and very little is currently known about the relevance of innate immunity for the control of JCV replication. Here, we define the intracellular innate immune sensors responsible for controlling JCV infection and also demonstrate a novel mechanism by which a JCV-encoded protein acts as an antagonist of the type I interferon-mediated innate immune response.


Asunto(s)
Antígenos Virales de Tumores/inmunología , Proteína 58 DEAD Box/inmunología , Inmunidad Innata , Virus JC/inmunología , Proteínas de Unión al ARN/antagonistas & inhibidores , ARN/metabolismo , Receptores Inmunológicos/inmunología , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Antígenos Virales de Tumores/genética , Astrocitos/virología , Células Cultivadas , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Células HEK293 , Humanos , Virus JC/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética
4.
Nat Microbiol ; 6(4): 467-478, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33727702

RESUMEN

Activation of the RIG-I-like receptors, retinoic-acid inducible gene I (RIG-I) and melanoma differentiation-associated protein 5 (MDA5), establishes an antiviral state by upregulating interferon (IFN)-stimulated genes (ISGs). Among these is ISG15, the mechanistic roles of which in innate immunity still remain enigmatic. In the present study, we report that ISG15 conjugation is essential for antiviral IFN responses mediated by the viral RNA sensor MDA5. ISGylation of the caspase activation and recruitment domains of MDA5 promotes its oligomerization and thereby triggers activation of innate immunity against a range of viruses, including coronaviruses, flaviviruses and picornaviruses. The ISG15-dependent activation of MDA5 is antagonized through direct de-ISGylation mediated by the papain-like protease of SARS-CoV-2, a recently emerged coronavirus that has caused the COVID-19 pandemic. Our work demonstrates a crucial role for ISG15 in the MDA5-mediated antiviral response, and also identifies a key immune evasion mechanism of SARS-CoV-2, which may be targeted for the development of new antivirals and vaccines to combat COVID-19.


Asunto(s)
Proteasas Similares a la Papaína de Coronavirus/metabolismo , Citocinas/metabolismo , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , SARS-CoV-2/enzimología , SARS-CoV-2/inmunología , Ubiquitinas/metabolismo , Aedes , Animales , Chlorocebus aethiops , Cricetinae , Células HEK293 , Humanos , Helicasa Inducida por Interferón IFIH1/metabolismo , Leucocitos Mononucleares , Ratones , Células Vero
5.
Viruses ; 13(2)2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33530371

RESUMEN

Viral dysregulation or suppression of innate immune responses is a key determinant of virus-induced pathogenesis. Important sensors for the detection of virus infection are the RIG-I-like receptors (RLRs), which, in turn, are antagonized by many RNA viruses and DNA viruses. Among the different escape strategies are viral mechanisms to dysregulate the post-translational modifications (PTMs) that play pivotal roles in RLR regulation. In this review, we present the current knowledge of immune evasion by viral pathogens that manipulate ubiquitin- or ISG15-dependent mechanisms of RLR activation. Key viral strategies to evade RLR signaling include direct targeting of ubiquitin E3 ligases, active deubiquitination using viral deubiquitinating enzymes (DUBs), and the upregulation of cellular DUBs that regulate RLR signaling. Additionally, we summarize emerging new evidence that shows that enzymes of certain coronaviruses such as SARS-CoV-2, the causative agent of the current COVID-19 pandemic, actively deISGylate key molecules in the RLR pathway to escape type I interferon (IFN)-mediated antiviral responses. Finally, we discuss the possibility of targeting virally-encoded proteins that manipulate ubiquitin- or ISG15-mediated innate immune responses for the development of new antivirals and vaccines.


Asunto(s)
Citocinas/metabolismo , Proteína 58 DEAD Box/metabolismo , Evasión Inmune , Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Virus/inmunología , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/metabolismo , Humanos , Inmunidad Innata , Receptores Inmunológicos , SARS-CoV-2/inmunología , SARS-CoV-2/metabolismo , Transducción de Señal , Virosis/inmunología , Virosis/metabolismo , Virosis/virología , Virus/metabolismo
6.
Cell Host Microbe ; 26(4): 493-503.e6, 2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31600501

RESUMEN

14-3-3 protein family members facilitate the translocation of RIG-I-like receptors (RLRs) to organelles that mediate downstream RLR signaling, leading to interferon production. 14-3-3ϵ promotes the cytosolic-to-mitochondrial translocation of RIG-I, while 14-3-3η facilitates MDA5 translocation to mitochondria. We show that the NS3 protein of Zika virus (ZIKV) antagonizes antiviral gene induction by RIG-I and MDA5 by binding to and sequestering the scaffold proteins 14-3-3ϵ and 14-3-3η. 14-3-3-binding is mediated by a negatively charged RLDP motif in NS3 that is conserved in ZIKV strains of African and Asian lineages and is similar to the one found in dengue and West Nile viruses. ZIKV NS3 is sufficient to inhibit the RLR-14-3-3ϵ/η interaction and to suppress antiviral signaling. Mutational perturbation of 14-3-3ϵ/η binding in a recombinant ZIKV leads to enhanced innate immune responses and impaired growth kinetics. Our study provides molecular understanding of immune evasion functions of ZIKV, which may guide vaccine and anti-flaviviral therapy development.


Asunto(s)
Proteínas 14-3-3/metabolismo , Evasión Inmune/inmunología , Péptido Hidrolasas/metabolismo , Proteínas Virales/metabolismo , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Células A549 , Animales , Línea Celular , Chlorocebus aethiops , Proteína 58 DEAD Box/antagonistas & inhibidores , Células HEK293 , Células HeLa , Humanos , Inmunidad Innata/inmunología , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Interferón beta/inmunología , Mitocondrias/metabolismo , Péptido Hidrolasas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores Inmunológicos , Serina Endopeptidasas , Células Vero , Proteínas Virales/genética , Virus Zika/genética
7.
J Interferon Cytokine Res ; 39(8): 450-458, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31066607

RESUMEN

The innate immune defense of mammalian hosts relies on its capacity to detect invading pathogens and then directly eliminate them or help guide adaptive immune responses. Recognition of microbial DNA and RNA by pattern recognition receptors (PRRs) is central to the detection of pathogens by initiating cytokine-mediated innate immunity. In contrast, disturbance of this pathogen surveillance system can result in aberrant innate immune activation, leading to proinflammatory or autoimmune diseases. Among the many important PRRs are proteins of the retinoic acid-inducible gene-I (RIG-I)-like receptor (RLR) family as well as cyclic GMP-AMP synthase (cGAS), which detect viral RNA and DNA, respectively, within the host cell. Intriguingly, recent evidence has shown that "unmasked," misprocessed, or mislocalized host-derived RNA or DNA molecules can also be recognized by RLRs or cGAS, thereby triggering antiviral host defenses or causing inflammation. Here, we review recent advances of endogenous nucleic acid recognition by RLRs and cGAS during viral infection and systemic proinflammatory/autoimmune disorders.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Ácidos Nucleicos/inmunología , Nucleotidiltransferasas/metabolismo , Receptores de Reconocimiento de Patrones/inmunología , Animales , Humanos , Ácidos Nucleicos/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo
8.
Nat Microbiol ; 4(1): 164-176, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30420784

RESUMEN

Tripartite motif (TRIM) proteins mediate antiviral host defences by either directly targeting viral components or modulating innate immune responses. Here we identify a mechanism of antiviral restriction in which a TRIM E3 ligase controls viral replication by regulating the structure of host cell centrosomes and thereby nuclear lamina integrity. Through RNAi screening we identified several TRIM proteins, including TRIM43, that control the reactivation of Kaposi's sarcoma-associated herpesvirus. TRIM43 was distinguished by its ability to restrict a broad range of herpesviruses and its profound upregulation during herpesvirus infection as part of a germline-specific transcriptional program mediated by the transcription factor DUX4. TRIM43 ubiquitinates the centrosomal protein pericentrin, thereby targeting it for proteasomal degradation, which subsequently leads to alterations of the nuclear lamina that repress active viral chromatin states. Our study identifies a role of the TRIM43-pericentrin-lamin axis in intrinsic immunity, which may be targeted for therapeutic intervention against herpesviral infections.


Asunto(s)
Antígenos/metabolismo , Centrosoma/metabolismo , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 8/crecimiento & desarrollo , Proteínas de Motivos Tripartitos/metabolismo , Replicación Viral/fisiología , Células A549 , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Células HeLa , Infecciones por Herpesviridae/prevención & control , Infecciones por Herpesviridae/virología , Proteínas de Homeodominio/metabolismo , Humanos , Lámina Nuclear/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/fisiología , Ubiquitinación , Células Vero , Replicación Viral/genética
9.
Acad Pediatr ; 18(1): 59-65, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28870650

RESUMEN

OBJECTIVE: To examine the prevalence of positive screening scores, construct validity, and opportunities for follow-up in a large sample of adolescents who chose to fill out the Pediatric Symptom Checklist-Youth Form (PSC-Y) through the Mental Health America (MHA) Web site. METHODS: MHA sent researchers a deidentified data set of all PSC-Y data submitted to MHA from May 15, 2015 to May 14, 2016. The analytic data set contained 29,886 PSC-Y forms from youth aged 11 to 17 years who sought out the Web site and chose to fill out the PSC-Y anonymously and independently online. The prevalence of impairment on the PSC-Y was calculated overall and for various subgroups. Next steps reported by at-risk youth were also examined. RESULTS: Of all respondents, 77.4% of youth screened positive on the PSC-Y. Significant associations between positive screening and self-ratings of a need for help, previous history of mental health treatment, and low family income provided construct validation for the online PSC-Y. Almost two-thirds of positively screened youth stated that they planned to get some kind of help in the future and 10% indicated that they planned to seek professional treatment. CONCLUSIONS: The large number of respondents suggested that many adolescents use the Internet to learn about mental health and that a very high percentage of them might be at risk. The availability of brief, free Internet-based psychosocial screens might offer a viable way to identify at-risk youth and provide them with pathways to additional support and/or treatment.


Asunto(s)
Internet , Tamizaje Masivo , Trastornos Mentales/diagnóstico , Adolescente , Lista de Verificación , Niño , Autoevaluación Diagnóstica , Femenino , Humanos , Masculino , Trastornos Mentales/epidemiología , Prioridad del Paciente , Prevalencia , Derivación y Consulta , Minorías Sexuales y de Género/estadística & datos numéricos , Estados Unidos/epidemiología
10.
J Virol ; 92(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29263274

RESUMEN

Retinoic acid-inducible gene I (RIG-I) is a key pattern recognition receptor that senses viral RNA and interacts with the mitochondrial adaptor MAVS, triggering a signaling cascade that results in the production of type I interferons (IFNs). This signaling axis is initiated by K63-linked ubiquitination of RIG-I mediated by the E3 ubiquitin ligase TRIM25, which promotes the interaction of RIG-I with MAVS. USP15 was recently identified as an upstream regulator of TRIM25, stabilizing the enzyme through removal of degradative K48-linked polyubiquitin, ultimately promoting RIG-I-dependent cytokine responses. Here, we show that the E6 oncoprotein of human papillomavirus type 16 (HPV16) as well as of other HPV types form a complex with TRIM25 and USP15 in human cells. In the presence of E6, the K48-linked ubiquitination of TRIM25 was markedly increased, and in line with this, TRIM25 degradation was enhanced. Our results further showed that E6 inhibited the TRIM25-mediated K63-linked ubiquitination of RIG-I and its CARD-dependent interaction with MAVS. HPV16 E6, but not E7, suppressed the RIG-I-mediated induction of IFN-ß, chemokines, and IFN-stimulated genes (ISGs). Finally, CRISPR-Cas9 gene targeting in human keratinocytes showed that the TRIM25-RIG-I-MAVS triad is important for eliciting an antiviral immune response to HPV16 infection. Our study thus identifies a novel immune escape mechanism that is conserved among different HPV strains and further indicates that the RIG-I signaling pathway plays an important role in the innate immune response to HPV infection.IMPORTANCE Persistent infection and tumorigenesis by HPVs are known to require viral manipulation of a variety of cellular processes, including those involved in innate immune responses. Here, we show that the HPV E6 oncoprotein antagonizes the activation of the cytoplasmic innate immune sensor RIG-I by targeting its upstream regulatory enzymes TRIM25 and USP15. We further show that the RIG-I signaling cascade is important for an antiviral innate immune response to HPV16 infection, providing evidence that RIG-I, whose role in sensing RNA virus infections has been well characterized, also plays a crucial role in the antiviral host response to small DNA viruses of the Papillomaviridae family.


Asunto(s)
Proteína 58 DEAD Box/inmunología , Papillomavirus Humano 6/inmunología , Inmunidad Innata , Queratinocitos/inmunología , Proteínas Oncogénicas Virales/inmunología , Infecciones por Papillomavirus/inmunología , Transducción de Señal/inmunología , Factores de Transcripción/inmunología , Proteínas de Motivos Tripartitos/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Proteasas Ubiquitina-Específicas/inmunología , Proteína 58 DEAD Box/genética , Células HEK293 , Papillomavirus Humano 6/genética , Humanos , Queratinocitos/patología , Queratinocitos/virología , Proteínas Oncogénicas Virales/genética , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/patología , Receptores Inmunológicos , Transducción de Señal/genética , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética , Proteasas Ubiquitina-Específicas/genética
11.
Mol Ther ; 25(8): 1900-1916, 2017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28527723

RESUMEN

Oncolytic viruses (OVs) offer a promising therapeutic approach to treat multiple types of cancer. In this study, we show that the manipulation of the antioxidant network via transcription factor Nrf2 augments vesicular stomatitis virus Δ51 (VSVΔ51) replication and sensitizes cancer cells to viral oncolysis. Activation of Nrf2 signaling by the antioxidant compound sulforaphane (SFN) leads to enhanced VSVΔ51 spread in OV-resistant cancer cells and improves the therapeutic outcome in different murine syngeneic and xenograft tumor models. Chemoresistant A549 lung cancer cells that display constitutive dominant hyperactivation of Nrf2 signaling are particularly vulnerable to VSVΔ51 oncolysis. Mechanistically, enhanced Nrf2 signaling stimulated viral replication in cancer cells and disrupted the type I IFN response via increased autophagy. This study reveals a previously unappreciated role for Nrf2 in the regulation of autophagy and the innate antiviral response that complements the therapeutic potential of VSV-directed oncolysis against multiple types of OV-resistant or chemoresistant cancer.


Asunto(s)
Autofagia , Factor 2 Relacionado con NF-E2/metabolismo , Virus Oncolíticos/fisiología , Transducción de Señal , Estomatitis Vesicular/metabolismo , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/fisiología , Animales , Antineoplásicos/farmacología , Antioxidantes/farmacología , Autofagia/efectos de los fármacos , Línea Celular , Terapia Combinada , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Isotiocianatos/farmacología , Ratones , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Neoplasias/metabolismo , Neoplasias/mortalidad , Neoplasias/patología , Neoplasias/terapia , Viroterapia Oncolítica , Eliminación de Secuencia , Transducción de Señal/efectos de los fármacos , Sulfóxidos , Estomatitis Vesicular/inmunología , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Proteínas de la Matriz Viral/genética , Replicación Viral/efectos de los fármacos
12.
Trends Immunol ; 38(1): 39-52, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27863906

RESUMEN

Mammalian cells recognize virus-derived nucleic acids using a defined set of intracellular sensors including the DNA sensors cyclic GMP-AMP (cGAMP) synthase (cGAS) and interferon gamma (IFNγ)-inducible protein 16 (IFI16) as well as viral RNA receptors of the retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) family. Following innate immune recognition, these sensors launch an immune response that is characterized by the transcriptional upregulation of many antiviral molecules, including proinflammatory cytokines, chemokines, and IFN-stimulated genes. Recent studies have demonstrated that the signal transduction initiated by these sensors is sophisticatedly regulated by post-translational modifications (PTMs) resulting in a robust yet 'tunable' cytokine response to maintain immune homeostasis. Here we summarize recent advances in our understanding of how PTMs and regulatory enzymes control the signaling activity of RLRs, cGAS, and IFI16 as well as their proximal adaptor proteins.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Proteínas Nucleares/metabolismo , Nucleótidos Cíclicos/metabolismo , Fosfoproteínas/metabolismo , Procesamiento Proteico-Postraduccional , Virosis/inmunología , Animales , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Espacio Intracelular , Proteínas Nucleares/genética , Fosfoproteínas/genética , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal
13.
J Virol ; 89(20): 10612-24, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26269188

RESUMEN

UNLABELLED: The molecular interaction between viral RNA and the cytosolic sensor RIG-I represents the initial trigger in the development of an effective immune response against infection with RNA viruses, resulting in innate immune activation and subsequent induction of adaptive responses. In the present study, the adjuvant properties of a sequence-optimized 5'-triphosphate-containing RNA (5'pppRNA) RIG-I agonist (termed M8) were examined in combination with influenza virus-like particles (VLP) (M8-VLP) expressing H5N1 influenza virus hemagglutinin (HA) and neuraminidase (NA) as immunogens. In combination with VLP, M8 increased the antibody response to VLP immunization, provided VLP antigen sparing, and protected mice from a lethal challenge with H5N1 influenza virus. M8-VLP immunization also led to long-term protective responses against influenza virus infection in mice. M8 adjuvantation of VLP increased endpoint and antibody titers and inhibited influenza virus replication in lungs compared with approved or experimental adjuvants alum, AddaVax, and poly(I·C). Uniquely, immunization with M8-VLP stimulated a TH1-biased CD4 T cell response, as determined by increased TH1 cytokine levels in CD4 T cells and increased IgG2 levels in sera. Collectively, these data demonstrate that a sequence-optimized, RIG-I-specific agonist is a potent adjuvant that can be utilized to increase the efficacy of influenza VLP vaccination and dramatically improve humoral and cellular mediated protective responses against influenza virus challenge. IMPORTANCE: The development of novel adjuvants to increase vaccine immunogenicity is an important goal that seeks to improve vaccine efficacy and ultimately prevent infections that endanger human health. This proof-of-principle study investigated the adjuvant properties of a sequence-optimized 5'pppRNA agonist (M8) with enhanced capacity to stimulate antiviral and inflammatory gene networks using influenza virus-like particles (VLP) expressing HA and NA as immunogens. Vaccination with VLP in combination with M8 increased anti-influenza virus antibody titers and protected animals from lethal influenza virus challenge, highlighting the potential clinical use of M8 as an adjuvant in vaccine development. Altogether, the results describe a novel immunostimulatory agonist targeted to the cytosolic RIG-I sensor as an attractive vaccine adjuvant candidate that can be used to increase vaccine efficacy, a pressing issue in children and the elderly population.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Anticuerpos Antivirales/biosíntesis , ARN Helicasas DEAD-box/inmunología , Vacunas contra la Influenza/inmunología , Oligorribonucleótidos/administración & dosificación , Infecciones por Orthomyxoviridae/prevención & control , Vacunas de Partículas Similares a Virus/inmunología , Adyuvantes Inmunológicos/genética , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/genética , Células Dendríticas/inmunología , Células Dendríticas/virología , Femenino , Células HEK293 , Hemaglutininas Virales/química , Hemaglutininas Virales/genética , Hemaglutininas Virales/inmunología , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Inmunización , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Ratones , Ratones Endogámicos BALB C , Neuraminidasa/química , Neuraminidasa/genética , Neuraminidasa/inmunología , Oligorribonucleótidos/genética , Oligorribonucleótidos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/mortalidad , Infecciones por Orthomyxoviridae/virología , Cultivo Primario de Células , Receptores Inmunológicos , Análisis de Supervivencia , Balance Th1 - Th2/efectos de los fármacos , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/genética
14.
Biol Chem ; 396(12): 1269-80, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26068902

RESUMEN

Resistance to both cytotoxic and targeted therapies is a major problem facing cancer treatment. The mechanisms of resistance to unrelated drugs share many common features, including up-regulation of detoxifying pathways, activation of pro-survival mechanisms, and ineffective induction of cell death. Oncolytic viruses (OVs) are promising biotherapeutics for cancer treatment that specifically replicate in and lyse cancer cells. In addition to direct viral lysis, the anti-tumor effects of OVs are mediated via innate and adaptive immune responses, and several adaptation mechanisms such as autophagy appear to contribute to their anti-tumor properties. Autophagy is a versatile pathway that plays a key role in cancer survival during stressful conditions such as starvation or cytotoxic drug challenges. Autophagy also plays a role in mediating innate and adaptive immune responses by contributing to antigen presentation and cytokine secretion. This role of autophagy in regulation of immune responses can be utilized to design therapeutic combinations using approaches that either stimulate or block autophagy to potentiate therapeutic efficacy of OVs. Additional studies are needed to determine optimal multimodal combination approaches that will facilitate future successful clinical implementation of OV-based therapies.


Asunto(s)
Autofagia/fisiología , Resistencia a Medicamentos , Neoplasias/terapia , Neoplasias/virología , Virus Oncolíticos , Humanos , Neoplasias/tratamiento farmacológico , Linfocitos T/inmunología
15.
J Virol ; 89(15): 8011-25, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26018150

RESUMEN

UNLABELLED: The cytosolic RIG-I (retinoic acid-inducible gene I) receptor plays a pivotal role in the initiation of the immune response against RNA virus infection by recognizing short 5'-triphosphate (5'ppp)-containing viral RNA and activating the host antiviral innate response. In the present study, we generated novel 5'ppp RIG-I agonists of varieous lengths, structures, and sequences and evaluated the generation of the antiviral and inflammatory responses in human epithelial A549 cells, human innate immune primary cells, and murine models of influenza and chikungunya viral pathogenesis. A 99-nucleotide, uridine-rich hairpin 5'pppRNA termed M8 stimulated an extensive and robust interferon response compared to other modified 5'pppRNA structures, RIG-I aptamers, or poly(I·C). Interestingly, manipulation of the primary RNA sequence alone was sufficient to modulate antiviral activity and inflammatory response, in a manner dependent exclusively on RIG-I and independent of MDA5 and TLR3. Both prophylactic and therapeutic administration of M8 effectively inhibited influenza virus and dengue virus replication in vitro. Furthermore, multiple strains of influenza virus that were resistant to oseltamivir, an FDA-approved therapeutic treatment for influenza, were highly sensitive to inhibition by M8. Finally, prophylactic M8 treatment in vivo prolonged survival and reduced lung viral titers of mice challenged with influenza virus, as well as reducing chikungunya virus-associated foot swelling and viral load. Altogether, these results demonstrate that 5'pppRNA can be rationally designed to achieve a maximal RIG-I-mediated protective antiviral response against human-pathogenic RNA viruses. IMPORTANCE: The development of novel therapeutics to treat human-pathogenic RNA viral infections is an important goal to reduce spread of infection and to improve human health and safety. This study investigated the design of an RNA agonist with enhanced antiviral and inflammatory properties against influenza, dengue, and chikungunya viruses. A novel, sequence-dependent, uridine-rich RIG-I agonist generated a protective antiviral response in vitro and in vivo and was effective at concentrations 100-fold lower than prototype sequences or other RNA agonists, highlighting the robust activity and potential clinical use of the 5'pppRNA against RNA virus infection. Altogether, the results identify a novel, sequence-specific RIG-I agonist as an attractive therapeutic candidate for the treatment of a broad range of RNA viruses, a pressing issue in which a need for new and more effective options persists.


Asunto(s)
Virus Chikungunya/inmunología , ARN Helicasas DEAD-box/inmunología , Virus del Dengue/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , ARN Viral/agonistas , ARN Viral/inmunología , Virosis/inmunología , Animales , Línea Celular , Virus Chikungunya/química , Virus Chikungunya/genética , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Virus del Dengue/química , Virus del Dengue/genética , Humanos , Subtipo H1N1 del Virus de la Influenza A/química , Subtipo H1N1 del Virus de la Influenza A/genética , Ratones , Ratones Endogámicos BALB C , Conformación de Ácido Nucleico , ARN Viral/genética , Receptores Inmunológicos , Virosis/genética , Virosis/virología
16.
PLoS Pathog ; 10(12): e1004575, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25521510

RESUMEN

The mechanisms involved in the persistence of activated CD4+ T lymphocytes following primary human T leukemia/lymphoma virus type 1 (HTLV-1) infection remain unclear. Here, we demonstrate that the HTLV-1 Tax oncoprotein modulates phosphorylation and transcriptional activity of the FOXO3a transcription factor, via upstream activation of the AKT pathway. De novo HTLV-1 infection of CD4+ T cells or direct lentiviral-mediated introduction of Tax led to AKT activation and AKT-dependent inactivation of FOXO3a, via phosphorylation of residues Ser253 and Thr32. Inhibition of FOXO3a signalling led to the long-term survival of a population of highly activated, terminally differentiated CD4+Tax+CD27negCCR7neg T cells that maintained the capacity to disseminate infectious HTLV-1. CD4+ T cell persistence was reversed by chemical inhibition of AKT activity, lentiviral-mediated expression of a dominant-negative form of FOXO3a or by specific small interfering RNA (siRNA)-mediated silencing of FOXO3a. Overall this study provides new mechanistic insight into the strategies used by HTLV-1 to increase long-term maintenance of Tax+CD4+ T lymphocytes during the early stages of HTLV-1 pathogenesis.


Asunto(s)
Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/virología , Factores de Transcripción Forkhead/antagonistas & inhibidores , Productos del Gen tax/fisiología , Infecciones por HTLV-I/fisiopatología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Diferenciación Celular , Supervivencia Celular/fisiología , Células Cultivadas , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/efectos de los fármacos , Factores de Transcripción Forkhead/fisiología , Infecciones por HTLV-I/patología , Humanos , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , ARN Interferente Pequeño/farmacología , Transducción de Señal/fisiología , Proteínas Virales/fisiología
17.
PLoS One ; 9(4): e95927, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24759703

RESUMEN

Upon viral infections, pattern recognition receptors (PRRs) recognize pathogen-associated molecular patterns (PAMPs) and stimulate an antiviral state associated with the production of type I interferons (IFNs) and inflammatory markers. Type I IFNs play crucial roles in innate antiviral responses by inducing expression of interferon-stimulated genes and by activating components of the adaptive immune system. Although pegylated IFNs have been used to treat hepatitis B and C virus infections for decades, they exert substantial side effects that limit their use. Current efforts are directed toward the use of PRR agonists as an alternative approach to elicit host antiviral responses in a manner similar to that achieved in a natural infection. RIG-I is a cytosolic PRR that recognizes 5' triphosphate (5'ppp)-containing RNA ligands. Due to its ubiquitous expression profile, induction of the RIG-I pathway provides a promising platform for the development of novel antiviral agents and vaccine adjuvants. In this study, we investigated whether structured RNA elements in the genome of coxsackievirus B3 (CVB3), a picornavirus that is recognized by MDA5 during infection, could activate RIG-I when supplied with 5'ppp. We show here that a 5'ppp-containing cloverleaf (CL) RNA structure is a potent RIG-I inducer that elicits an extensive antiviral response that includes induction of classical interferon-stimulated genes, as well as type III IFNs and proinflammatory cytokines and chemokines. In addition, we show that prophylactic treatment with CVB3 CL provides protection against various viral infections including dengue virus, vesicular stomatitis virus and enterovirus 71, demonstrating the antiviral efficacy of this RNA ligand.


Asunto(s)
Antivirales/farmacología , Picornaviridae/genética , Infecciones por Virus ARN/inmunología , ARN Viral/química , Receptores de Ácido Retinoico/metabolismo , Animales , Antivirales/uso terapéutico , Línea Celular , Citocinas/metabolismo , ARN Helicasas DEAD-box/metabolismo , Fibroblastos/citología , Fibroblastos/inmunología , Fibroblastos/virología , Células HeLa , Humanos , Ratones , Modelos Moleculares , Conformación de Ácido Nucleico , Picornaviridae/química , Picornaviridae/inmunología , Polifosfatos/farmacología , Infecciones por Virus ARN/prevención & control , Infecciones por Virus ARN/virología , ARN Viral/inmunología , ARN Viral/metabolismo , ARN Viral/farmacología , ARN Viral/uso terapéutico , Receptores de Ácido Retinoico/genética
18.
J Virol ; 88(8): 4180-94, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24478443

RESUMEN

UNLABELLED: RIG-I is a cytosolic sensor critically involved in the activation of the innate immune response to RNA virus infection. In the present study, we evaluated the inhibitory effect of a RIG-I agonist on the replication of two emerging arthropod-borne viral pathogens, dengue virus (DENV) and chikungunya virus (CHIKV), for which no therapeutic options currently exist. We demonstrate that when a low, noncytotoxic dose of an optimized 5'triphosphorylated RNA (5'pppRNA) molecule was administered, RIG-I stimulation generated a robust antiviral response against these two viruses. Strikingly, 5'pppRNA treatment before or after challenge with DENV or CHIKV provided protection against infection. In primary human monocytes and monocyte-derived dendritic cells, the RIG-I agonist blocked both primary infection and antibody-dependent enhancement of DENV infection. The protective response against DENV and CHIKV induced by 5'pppRNA was dependent on an intact RIG-I/MAVS/TBK1/IRF3 axis and was largely independent of the type I IFN response. Altogether, this in vitro analysis of the antiviral efficacy of 5'pppRNA highlights the therapeutic potential of RIG-I agonists against emerging viruses such as DENV and CHIKV. IMPORTANCE: DENV and CHIKV are two reemerging mosquito-borne viruses for which no therapeutic options currently exist. Both viruses overlap geographically in tropical regions of the world, produce similar fever-like symptoms, and are difficult to diagnose. This study investigated the inhibitory effect of a RIG-I agonist on the replication of these two viruses. RIG-I stimulation using 5'pppRNA before or after DENV or CHIKV infection generated a protective antiviral response against both pathogens in immune and nonimmune cells; interestingly, the protective response against the viruses was largely independent of the classical type I interferon response. The antiviral efficacy of 5'pppRNA highlights the therapeutic potential of RIG-I agonists against emerging viruses such as DENV and CHIKV.


Asunto(s)
Infecciones por Alphavirus/inmunología , Virus Chikungunya/fisiología , ARN Helicasas DEAD-box/inmunología , Dengue/inmunología , Inmunidad Innata , Interferón Tipo I/inmunología , Infecciones por Alphavirus/genética , Infecciones por Alphavirus/virología , Animales , Línea Celular , Fiebre Chikungunya , Virus Chikungunya/genética , Virus Chikungunya/inmunología , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Dengue/genética , Dengue/virología , Virus del Dengue/genética , Virus del Dengue/inmunología , Virus del Dengue/fisiología , Humanos , Interferón Tipo I/genética , Ratones , Receptores Inmunológicos , Replicación Viral
19.
J Virol ; 88(5): 2927-40, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24371063

RESUMEN

Vesicular stomatitis virus (VSV) is an oncolytic virus that induces cancer cell death through activation of the apoptotic pathway. Intrinsic resistance to oncolysis is found in some cell lines and many primary tumors as a consequence of residual innate immunity to VSV. In resistant-tumor models, VSV oncolytic potential can be reversibly stimulated by combination with epigenetic modulators, such as the histone deacetylase inhibitor vorinostat. Based on this reversible effect of vorinostat, we reasoned that critical host genes involved in oncolysis may likewise be reversibly regulated by vorinostat. A transcriptome analysis in prostate cancer PC3 cells identified a subset of NF-κB target genes reversibly regulated by vorinostat, as well as a group of interferon (IFN)-stimulated genes (ISGs). Consistent with the induction of NF-κB target genes, vorinostat-mediated enhancement of VSV oncolysis increased hyperacetylation of NF-κB RELA/p65. Additional bioinformatics analysis revealed that NF-κB signaling also increased the expression of several autophagy-related genes. Kinetically, autophagy preceded apoptosis, and apoptosis was observed only when cells were treated with both VSV and vorinostat. VSV replication and cell killing were suppressed when NF-κB signaling was inhibited using pharmacological or genetic approaches. Inhibition of autophagy by 3-methyladenine (3-MA) enhanced expression of ISGs, and either 3-MA treatment or genetic ablation of the autophagic marker Atg5 decreased VSV replication and oncolysis. Together, these data demonstrate that vorinostat stimulates NF-κB activity in a reversible manner via modulation of RELA/p65 signaling, leading to induction of autophagy, suppression of the IFN-mediated response, and subsequent enhancement of VSV replication and apoptosis.


Asunto(s)
Autofagia , Inhibidores de Histona Desacetilasas/farmacología , FN-kappa B/metabolismo , Virus Oncolíticos/efectos de los fármacos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Acetilación , Animales , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Cromatina/metabolismo , Análisis por Conglomerados , Técnicas de Silenciamiento del Gen , Humanos , Ácidos Hidroxámicos/farmacología , Masculino , Ratones , FN-kappa B/antagonistas & inhibidores , Viroterapia Oncolítica , Virus Oncolíticos/genética , Neoplasias de la Próstata/terapia , Unión Proteica , Transporte de Proteínas/efectos de los fármacos , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Transcriptoma , Virus de la Estomatitis Vesicular Indiana/genética , Replicación Viral , Vorinostat
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA