Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.531
Filtrar
1.
Nature ; 626(7997): 194-206, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38096902

RESUMEN

The LINE-1 (L1) retrotransposon is an ancient genetic parasite that has written around one-third of the human genome through a 'copy and paste' mechanism catalysed by its multifunctional enzyme, open reading frame 2 protein (ORF2p)1. ORF2p reverse transcriptase (RT) and endonuclease activities have been implicated in the pathophysiology of cancer2,3, autoimmunity4,5 and ageing6,7, making ORF2p a potential therapeutic target. However, a lack of structural and mechanistic knowledge has hampered efforts to rationally exploit it. We report structures of the human ORF2p 'core' (residues 238-1061, including the RT domain) by X-ray crystallography and cryo-electron microscopy in several conformational states. Our analyses identified two previously undescribed folded domains, extensive contacts to RNA templates and associated adaptations that contribute to unique aspects of the L1 replication cycle. Computed integrative structural models of full-length ORF2p show a dynamic closed-ring conformation that appears to open during retrotransposition. We characterize ORF2p RT inhibition and reveal its underlying structural basis. Imaging and biochemistry show that non-canonical cytosolic ORF2p RT activity can produce RNA:DNA hybrids, activating innate immune signalling through cGAS/STING and resulting in interferon production6-8. In contrast to retroviral RTs, L1 RT is efficiently primed by short RNAs and hairpins, which probably explains cytosolic priming. Other biochemical activities including processivity, DNA-directed polymerization, non-templated base addition and template switching together allow us to propose a revised L1 insertion model. Finally, our evolutionary analysis demonstrates structural conservation between ORF2p and other RNA- and DNA-dependent polymerases. We therefore provide key mechanistic insights into L1 polymerization and insertion, shed light on the evolutionary history of L1 and enable rational drug development targeting L1.


Asunto(s)
Endonucleasas , Elementos de Nucleótido Esparcido Largo , ADN Polimerasa Dirigida por ARN , Transcripción Reversa , Humanos , Microscopía por Crioelectrón , Endonucleasas/química , Endonucleasas/genética , Endonucleasas/metabolismo , Elementos de Nucleótido Esparcido Largo/genética , ARN/genética , ADN Polimerasa Dirigida por ARN/química , ADN Polimerasa Dirigida por ARN/genética , ADN Polimerasa Dirigida por ARN/metabolismo , Cristalografía por Rayos X , ADN/biosíntesis , ADN/genética , Inmunidad Innata , Interferones/biosíntesis
2.
Viruses ; 14(2)2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-35215858

RESUMEN

The genus Pestivirus, family Flaviviridae, includes four historically accepted species, i.e., bovine viral diarrhea virus (BVDV)-1 and -2, classical swine fever virus (CSFV), and border disease virus (BDV). A large number of new pestivirus species were identified in recent years. A common feature of most members is the presence of two unique proteins, Npro and Erns, that pestiviruses evolved to regulate the host's innate immune response. In addition to its function as a structural envelope glycoprotein, Erns is also released in the extracellular space, where it is endocytosed by neighboring cells. As an endoribonuclease, Erns is able to cleave viral ss- and dsRNAs, thus preventing the stimulation of the host's interferon (IFN) response. Here, we characterize the basic features of soluble Erns of a large variety of classified and unassigned pestiviruses that have not yet been described. Its ability to form homodimers, its RNase activity, and the ability to inhibit dsRNA-induced IFN synthesis were investigated. Overall, we found large differences between the various Erns proteins that cannot be predicted solely based on their primary amino acid sequences, and that might be the consequence of different virus-host co-evolution histories. This provides valuable information to delineate the structure-function relationship of pestiviral endoribonucleases.


Asunto(s)
Endorribonucleasas/metabolismo , Evasión Inmune , Inmunidad Innata , Pestivirus/inmunología , Pestivirus/patogenicidad , Proteínas del Envoltorio Viral/metabolismo , Animales , Línea Celular , Endocitosis , Endorribonucleasas/química , Endorribonucleasas/genética , Interferones/antagonistas & inhibidores , Interferones/biosíntesis , Mutación , Proteínas de Resistencia a Mixovirus/genética , Proteínas de Resistencia a Mixovirus/metabolismo , Pestivirus/metabolismo , ARN Bicatenario/metabolismo , ARN Viral/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
3.
Front Immunol ; 12: 753683, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899705

RESUMEN

Human parechovirus type 3 (PeV-A3) infection has been recognized as an emerging etiologic factor causing severe nerve disease or sepsis in infants and young children. But the neuropathogenic mechanisms of PeV-A3 remain unknown. To understand the pathogenesis of PeV-A3 infection in the neuronal system, PeV-A3-mediated cytopathic effects were analyzed in human glioblastoma cells and neuroblastoma cells. PeV-A3 induced interferons and inflammatory cytokine expression in these neuronal cells. The pronounced cytopathic effects accompanied with activation of death signaling pathways of apoptosis, autophagy, and pyroptosis were detected. A new experimental disease model of parechovirus encephalitis was established. In the disease model, intracranial inoculation with PeV-A3 in C57BL/6 neonatal mice showed body weight loss, hindlimb paralysis, and approximately 20% mortality. PeV-A3 infection in the hippocampus and cortex regions of the neonatal mouse brain was revealed. Mechanistic assay supported the in vitro results, indicating detection of PeV-A3 replication, inflammatory cytokine expression, and death signaling transduction in mouse brain tissues. These in vitro and in vivo studies revealed that the activation of death signaling and inflammation responses is involved in PeV-A3-mediated neurological disorders. The present results might account for some of the PeV-A3-associated clinical manifestations.


Asunto(s)
Efecto Citopatogénico Viral , Modelos Animales de Enfermedad , Encefalitis Viral/metabolismo , Parechovirus/patogenicidad , Infecciones por Picornaviridae/metabolismo , Animales , Animales Recién Nacidos , Apoptosis , Autofagia , Línea Celular Tumoral , Corteza Cerebral/virología , Chlorocebus aethiops , Citocinas/biosíntesis , Citocinas/genética , Encefalitis Viral/patología , Encefalitis Viral/virología , Glioblastoma/patología , Hipocampo/virología , Humanos , Inflamación , Interferón Tipo I/biosíntesis , Interferón Tipo I/genética , Interferón Tipo I/farmacología , Interferones/biosíntesis , Interferones/genética , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Neuroblastoma/patología , Parechovirus/efectos de los fármacos , Parechovirus/fisiología , Infecciones por Picornaviridae/patología , Infecciones por Picornaviridae/virología , Piroptosis , Células Vero , Replicación Viral/efectos de los fármacos , Interferón lambda
4.
Front Immunol ; 12: 718380, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34630393

RESUMEN

Estradiol (E2) and progesterone (P) have potent effects on immune function in the human uterine endometrium which is essential for creating an environment conducive for successful reproduction. Type III/lambda (λ) interferons (IFN) are implicated in immune defense of the placenta against viral pathogens, which occurs against the backdrop of high E2 and P levels. However, the effect of E2 and P in modulating the expression and function of IFNλ1 in the non-pregnant human uterine endometrium is unknown. We generated purified in vitro cultures of human uterine epithelial cells and stromal fibroblast cells recovered from hysterectomy specimens. Poly (I:C), a viral dsRNA mimic, potently increased secretion of IFNλ1 by both epithelial cells and fibroblasts. The secretion of IFNλ1 by epithelial cells significantly increased with increasing age following poly (I:C) stimulation. Stimulation of either cell type with E2 (5x10-8M) or P (1x10-7M) had no effect on expression or secretion of IFNλ1 either alone or in the presence of poly (I:C). E2 suppressed the IFNλ1-induced upregulation of the antiviral IFN-stimulated genes (ISGs) MxA, OAS2 and ISG15 in epithelial cells, but not fibroblasts. Estrogen receptor alpha (ERα) blockade using Raloxifene indicated that E2 mediated its inhibitory effects on ISG expression via ERα. In contrast to E2, P potentiated the upregulation of ISG15 in response to IFNλ1 but had no effect on MxA and OAS2 in epithelial cells. Our results demonstrate that the effects of E2 and P on IFNλ1-induced ISGs are cell-type specific. E2-mediated suppression, and selective P-mediated stimulation, of IFNλ1-induced ISG expression in uterine epithelial cells suggest that the effects of IFNλ1 varies with menstrual cycle stage, pregnancy, and menopausal status. The suppressive effect of E2 could be a potential mechanism by which ascending pathogens from the lower reproductive tract can infect the pregnant and non-pregnant endometrium.


Asunto(s)
Envejecimiento/metabolismo , Endometrio/citología , Endometrio/metabolismo , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Interferones/biosíntesis , Interleucinas/biosíntesis , Transducción de Señal , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/genética , Línea Celular , Células Cultivadas , Citocinas/metabolismo , Estradiol/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Progesterona/metabolismo
5.
Viruses ; 13(8)2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34452468

RESUMEN

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19), a global pandemic characterized by an exaggerated immune response and respiratory illness. Age (>60 years) is a significant risk factor for developing severe COVID-19. To better understand the host response of the aged airway epithelium to SARS-CoV-2 infection, we performed an in vitro study using primary human bronchial epithelial cells from donors >67 years of age differentiated on an air-liquid interface culture. We demonstrate that SARS-CoV-2 infection leads to early induction of a proinflammatory response and a delayed interferon response. In addition, we observed changes in the genes and pathways associated with cell death and senescence throughout infection. In summary, our study provides new and important insights into the temporal kinetics of the airway epithelial innate immune response to SARS-CoV-2 in older individuals.


Asunto(s)
Bronquios/inmunología , Bronquios/virología , Inmunidad Innata , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/virología , SARS-CoV-2/inmunología , Anciano , Envejecimiento/inmunología , Bronquios/citología , Bronquios/metabolismo , COVID-19/inmunología , Muerte Celular/genética , Células Cultivadas , Senescencia Celular/genética , Citocinas/biosíntesis , Citocinas/genética , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Femenino , Humanos , Inflamación , Interferones/biosíntesis , Interferones/genética , Masculino , RNA-Seq , Mucosa Respiratoria/citología , Mucosa Respiratoria/metabolismo , SARS-CoV-2/fisiología , Transducción de Señal/genética
6.
Int J Mol Sci ; 22(15)2021 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-34360899

RESUMEN

(1) Background: caspase-12 is activated during cytomegalovirus retinitis, although its role is presently unclear. (2) Methods: caspase-12-/- (KO) or caspase-12+/+ (WT) mice were immunosup eyes were analyzed by plaque assay, TUNEL assay, immunohistochemical staining, western blotting, and real-time PCR. (3) Results: increased retinitis and a more extensive virus spread were detected in the retina of infected eyes of KO mice compared to WT mice at day 14 p.i. Compared to MCMV injected WT eyes, mRNA levels of interferons α, ß and γ were significantly reduced in the neural retina of MCMV-infected KO eyes at day 14 p.i. Although similar numbers of MCMV infected cells, similar virus titers and similar numbers of TUNEL-staining cells were detected in injected eyes of both KO and WT mice at days 7 and 10 p.i., significantly lower amounts of cleaved caspase-3 and p53 protein were detected in infected eyes of KO mice at both time points. (4) Conclusions: caspase-12 contributes to caspase-3-dependent and independent retinal bystander cell death during MCMV retinitis and may also play an important role in innate immunity against virus infection of the retina.


Asunto(s)
Apoptosis/genética , Caspasa 12/deficiencia , Retinitis por Citomegalovirus/enzimología , Inmunidad Innata/genética , Muromegalovirus/fisiología , Retina/enzimología , Neuronas Retinianas/enzimología , Animales , Caspasa 12/genética , Retinitis por Citomegalovirus/genética , Retinitis por Citomegalovirus/virología , Femenino , Etiquetado Corte-Fin in Situ/métodos , Interferones/biosíntesis , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Retina/virología , Neuronas Retinianas/virología , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/metabolismo , Replicación Viral/genética
7.
Front Immunol ; 12: 705342, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34249014

RESUMEN

The intestine can be the target of several immunologically mediated diseases, including graft-versus-host disease (GVHD) and inflammatory bowel disease (IBD). GVHD is a life-threatening complication that occurs after allogeneic hematopoietic stem cell transplantation. Involvement of the gastrointestinal tract is associated with a particularly high mortality. GVHD development starts with the recognition of allo-antigens in the recipient by the donor immune system, which elicits immune-mediated damage of otherwise healthy tissues. IBD describes a group of immunologically mediated chronic inflammatory diseases of the intestine. Several aspects, including genetic predisposition and immune dysregulation, are responsible for the development of IBD, with Crohn's disease and ulcerative colitis being the two most common variants. GVHD and IBD share multiple key features of their onset and development, including intestinal tissue damage and loss of intestinal barrier function. A further common feature in the pathophysiology of both diseases is the involvement of cytokines such as type I and II interferons (IFNs), amongst others. IFNs are a family of protein mediators produced as a part of the inflammatory response, typically to pathogens or malignant cells. Diverse, and partially paradoxical, effects have been described for IFNs in GVHD and IBD. This review summarizes current knowledge on the role of type I, II and III IFNs, including basic concepts and controversies about their functions in the context of GVHD and IBD. In addition, therapeutic options, research developments and remaining open questions are addressed.


Asunto(s)
Enfermedad Injerto contra Huésped/fisiopatología , Enfermedades Inflamatorias del Intestino/fisiopatología , Interferones/fisiología , Enfermedades Intestinales/fisiopatología , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Infecciones Bacterianas/inmunología , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/patología , Trasplante de Células Madre Hematopoyéticas , Humanos , Inflamación , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/patología , Interferones/antagonistas & inhibidores , Interferones/biosíntesis , Enfermedades Intestinales/tratamiento farmacológico , Enfermedades Intestinales/etiología , Enfermedades Intestinales/patología , Ratones , Modelos Animales , Transducción de Señal , Virosis/inmunología
8.
Cell Death Dis ; 12(8): 743, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34315861

RESUMEN

Transcription factor IRF3 is critical for the induction of antiviral type I interferon (IFN-I). The epigenetic regulation of IFN-I production in antiviral innate immunity needs to be further identified. Here, we reported that epigenetic remodeler ARID1A, a critical component of the mSWI/SNF complex, could bind IRF3 and then was recruited to the Ifn-I promoter by IRF3, thus selectively promoting IFN-I but not TNF-α, IL-6 production in macrophages upon viral infection. Myeloid cell-specific deficiency of Arid1a rendered mice more susceptible to viral infection, accompanied with less IFN-I production. Mechanistically, ARID1A facilitates chromatin accessibility of IRF3 at the Ifn-I promoters by interacting with histone methyltransferase NSD2, which methylates H3K4 and H3K36 of the promoter regions. Our findings demonstrated the new roles of ARID1A and NSD2 in innate immunity, providing insight into the crosstalks of chromatin remodeling, histone modification, and transcription factors in the epigenetic regulation of antiviral innate immunity.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferones/biosíntesis , Macrófagos/metabolismo , Factores de Transcripción/metabolismo , Animales , Antivirales/metabolismo , Cromatina/metabolismo , Células HEK293 , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Humanos , Inmunidad Innata , Interferones/genética , Lisina/metabolismo , Metilación , Ratones , Regiones Promotoras Genéticas , Unión Proteica , Células RAW 264.7 , Vesiculovirus/fisiología
9.
Front Immunol ; 12: 679242, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995423

RESUMEN

Interferons are the first lines of defense against viral pathogen invasion during the early stages of infection. Their synthesis is tightly regulated to prevent excessive immune responses and possible deleterious effects on the host organism itself. The RIG-I-like receptor signaling cascade is one of the major pathways leading to the production of interferons. This pathway amplifies danger signals and mounts an appropriate innate response but also needs to be finely regulated to allow a rapid return to immune homeostasis. Recent advances have characterized different cellular factors involved in the control of the RIG-I pathway. This has been most extensively studied in mammalian species; however, some inconsistencies remain to be resolved. The IFN system is remarkably well conserved in vertebrates and teleost fish possess all functional orthologs of mammalian RIG-I-like receptors as well as most downstream signaling molecules. Orthologs of almost all mammalian regulatory components described to date exist in teleost fish, such as the widely used zebrafish, making fish attractive and powerful models to study in detail the regulation and evolution of the RIG-I pathway.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Peces/genética , Peces/metabolismo , Transducción de Señal , Animales , Proteínas Portadoras , Proteína 58 DEAD Box/genética , Peces/inmunología , Regulación de la Expresión Génica , Homeostasis , Inmunidad Innata , Interferones/biosíntesis , Monoéster Fosfórico Hidrolasas/metabolismo , Fosfotransferasas/metabolismo , Unión Proteica , Complejos de Ubiquitina-Proteína Ligasa/metabolismo
10.
J Virol ; 95(13): e0221120, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-33883226

RESUMEN

Merkel cell polyomavirus (MCPyV) infects most of the human population asymptomatically, but in rare cases it leads to a highly aggressive skin cancer called Merkel cell carcinoma (MCC). MCC incidence is much higher in aging and immunocompromised populations. The epidemiology of MCC suggests that dysbiosis between the host immune response and the MCPyV infectious cycle could contribute to the development of MCPyV-associated MCC. Insufficient restriction of MCPyV by normal cellular processes, for example, could promote the incidental oncogenic MCPyV integration events and/or entry into the original cell of MCC. Progress toward understanding MCPyV biology has been hindered by its narrow cellular tropism. Our discovery that primary human dermal fibroblasts (HDFs) support MCPyV infection has made it possible to closely model cellular responses to different stages of the infectious cycle. The present study reveals that the onset of MCPyV replication and early gene expression induces an inflammatory cytokine and interferon-stimulated gene (ISG) response. The cGAS-STING pathway, in coordination with NF-κB, mediates induction of this innate immune gene expression program. Further, silencing of cGAS or NF-κB pathway factors led to elevated MCPyV replication. We also discovered that the PYHIN protein IFI16 localizes to MCPyV replication centers but does not contribute to the induction of ISGs. Instead, IFI16 upregulates inflammatory cytokines in response to MCPyV infection by an alternative mechanism. The work described herein establishes a foundation for exploring how changes to the skin microenvironment induced by aging or immunodeficiency might alter the fate of MCPyV and its host cell to encourage carcinogenesis. IMPORTANCE MCC has a high rate of mortality and an increasing incidence. Immune-checkpoint therapies have improved the prognosis of patients with metastatic MCC. Still, a significant proportion of the patients fail to respond to immune-checkpoint therapies or have a medical need for iatrogenic immune-suppression. A greater understanding of MCPyV biology could inform targeted therapies for MCPyV-associated MCC. Moreover, cellular events preceding MCC oncogenesis remain largely unknown. The present study aims to explore how MCPyV interfaces with innate immunity during its infectious cycle. We describe how MCPyV replication and/or transcription elicit an innate immune response via cGAS-STING, NF-κB, and IFI16. We also explore the effects of this response on MCPyV replication. Our findings illustrate how healthy cellular conditions may allow low-level infection that evades immune destruction until highly active replication is restricted by host responses. Conversely, pathological conditions could result in unbridled MCPyV replication that licenses MCC tumorigenesis.


Asunto(s)
Citocinas/inmunología , Fibroblastos/inmunología , Inmunidad Innata/inmunología , Poliomavirus de Células de Merkel/inmunología , Piel/inmunología , Sistemas CRISPR-Cas/genética , Carcinoma de Células de Merkel/patología , Células Cultivadas , Citocinas/biosíntesis , Fibroblastos/virología , Células HEK293 , Humanos , Inmunidad Innata/genética , Interferones/biosíntesis , Interferones/inmunología , Proteínas de la Membrana/genética , Poliomavirus de Células de Merkel/crecimiento & desarrollo , FN-kappa B/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleotidiltransferasas/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Infecciones por Polyomavirus/inmunología , Piel/citología , Infecciones Tumorales por Virus/inmunología
11.
Nephron ; 145(1): 85-90, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33059348

RESUMEN

Collapsing glomerulopathy has multiple associations, including viral infections, medications like bisphosphonates and interferon, autoimmune diseases, and genetic predisposition. We report a case of collapsing focal segmental glomerulosclerosis associated with persistently high levels of interferon gamma produced by T-cell receptor αß (+), CD4- CD8- (double negative) T lymphocytes that progressed despite treatment and improvement of other cytokine levels. Double negative T cells are elevated and activated in autoimmune lymphoproliferative syndrome (ALPS). Production of elevated interferon gamma levels from double negative T cells in ALPS despite treatment provides insight to the pathophysiology of collapsing glomerulopathy, guiding future research for collapsing glomerulopathy.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Interferones/biosíntesis , Adulto , Femenino , Glomeruloesclerosis Focal y Segmentaria/inmunología , Humanos
12.
J Clin Immunol ; 41(1): 109-124, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33078349

RESUMEN

BACKGROUND: STK4 deficiency due to homozygous mutations in the STK4 gene encoding the STK4/MST1 kinase was first described in 2012. STK4/MST1 kinase regulates cell proliferation, survival, differentiation, and immune responses through canonical and non-canonical Hippo signaling pathways. OBJECTIVE: We describe an 11-year-old girl with a clinical presentation consisting of severe recurrent herpes zoster, chronic warts, and recurrent pneumonias, as well as a somatic phenotype with hypothyroidism and low stature. Whole exome sequencing revealed STK4 deficiency due to homozygosity for a novel frameshift variant in STK4, c.523dupA, p.(L174fsTer45), resulting in a premature stop codon within the kinase domain. METHODS: We performed a thorough investigation of the genetics and innate and adaptive immunological abnormalities in STK4 deficiency. RESULTS: We show significantly impaired type I, II, and III interferon (IFN) responses and partly reduced proinflammatory cytokine responses to ligands of Toll-like receptor (TLR)3, TLR9, and the cytosolic RNA and DNA sensors as well as to microorganisms. Impaired IFN responses could be attributed to reduced phosphorylation of TBK1 and IRF3. Moreover, virus infection induced enhanced cell death by apoptosis. Importantly, autophagy pathways were slightly disturbed, with enhanced LC3B-Ito LCB3-II conversion at the single cell level but normal overall formation of LCB3 punctae. Finally, the patient displayed some indicators of impaired adaptive immunity in the form of insufficient vaccination responses, T cell lymphopenia, and reduced Treg fractions, although with largely normal T cell proliferation and normal IFNg production. CONCLUSION: Here, we demonstrate disturbances in various immune cell populations and pathways involved in innate immune responses, cell death, autophagy, and adaptive immunity in a patient homozygous for a novel STK4 frameshift mutation.


Asunto(s)
Inmunidad Innata/genética , Factor 3 Regulador del Interferón/metabolismo , Interferones/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Inmunidad Adaptativa , Alelos , Autofagia , Diferenciación Celular , Proliferación Celular , Supervivencia Celular/genética , Citocinas/biosíntesis , Femenino , Genotipo , Vía de Señalización Hippo , Humanos , Huésped Inmunocomprometido , Inmunofenotipificación , Infecciones/etiología , Infecciones/metabolismo , Activación de Linfocitos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Mutación , Neutrófilos/inmunología , Neutrófilos/metabolismo , Linaje , Fenotipo , Linfocitos T/inmunología , Linfocitos T/metabolismo
13.
Signal Transduct Target Ther ; 5(1): 299, 2020 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-33372174

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has quickly spread worldwide and has affected more than 10 million individuals. A typical feature of COVID-19 is the suppression of type I and III interferon (IFN)-mediated antiviral immunity. However, the molecular mechanism by which SARS-CoV-2 evades antiviral immunity remains elusive. Here, we reported that the SARS-CoV-2 membrane (M) protein inhibits the production of type I and III IFNs induced by the cytosolic dsRNA-sensing pathway mediated by RIG-I/MDA-5-MAVS signaling. In addition, the SARS-CoV-2 M protein suppresses type I and III IFN induction stimulated by SeV infection or poly (I:C) transfection. Mechanistically, the SARS-CoV-2 M protein interacts with RIG-I, MAVS, and TBK1, thus preventing the formation of the multiprotein complex containing RIG-I, MAVS, TRAF3, and TBK1 and subsequently impeding the phosphorylation, nuclear translocation, and activation of IRF3. Consequently, ectopic expression of the SARS-CoV-2 M protein facilitates the replication of vesicular stomatitis virus. Taken together, these results indicate that the SARS-CoV-2 M protein antagonizes type I and III IFN production by targeting RIG-I/MDA-5 signaling, which subsequently attenuates antiviral immunity and enhances viral replication. This study provides insight into the interpretation of SARS-CoV-2-induced antiviral immune suppression and illuminates the pathogenic mechanism of COVID-19.


Asunto(s)
COVID-19/metabolismo , Proteína 58 DEAD Box/metabolismo , Interferón Tipo I/biosíntesis , Helicasa Inducida por Interferón IFIH1/metabolismo , Interferones/biosíntesis , SARS-CoV-2/metabolismo , Transducción de Señal , Proteínas de la Matriz Viral/metabolismo , Animales , COVID-19/genética , Chlorocebus aethiops , Proteína 58 DEAD Box/genética , Células HEK293 , Células HeLa , Humanos , Interferón Tipo I/genética , Helicasa Inducida por Interferón IFIH1/genética , Interferones/genética , Receptores Inmunológicos , SARS-CoV-2/genética , Células Vero , Proteínas de la Matriz Viral/genética , Interferón lambda
14.
Med Hypotheses ; 143: 110153, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32763662

RESUMEN

Reports from various countries suggest that tobacco smoking might protect from SARS-CoV-2 infection, since the prevalence of smoking in COVID-19 hospitalized patients is lower than in the respective general population. Apart from nicotine or other chemicals contained in tobacco smoke, we propose that a single-stranded RNA virus that infects tobacco leaves, tobacco mosaic virus (TMV), might be implicated in this effect. TMV, though non-pathogenic, is found in smokers' airways, and stimulates adaptive and innate immunity, with release of specific antibodies and interferons. The latter may have preventive and/or therapeutic effects against COVID-19. If confirmed by epidemiological and interventional studies, this might lead to the use of TMV as an immunological adjuvant against SARS-CoV-2 infection and COVID-19 disease.


Asunto(s)
Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Modelos Inmunológicos , Pandemias , Neumonía Viral/inmunología , Fumadores , Virus del Mosaico del Tabaco/inmunología , Productos de Tabaco/virología , Fumar Tabaco , Animales , Anticuerpos Antivirales/biosíntesis , COVID-19 , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/virología , Resistencia a la Enfermedad , Humanos , Interferones/biosíntesis , Ratones , Neumonía Viral/epidemiología , Neumonía Viral/virología , Sistema Respiratorio/inmunología , Sistema Respiratorio/virología , SARS-CoV-2 , Virus del Mosaico del Tabaco/aislamiento & purificación , Fumar Tabaco/epidemiología , Receptores Toll-Like/inmunología
15.
Arch Virol ; 165(10): 2279-2289, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32719955

RESUMEN

In the early stage of virus infection, the pattern recognition receptor (PRR) signaling pathway of the host cell is activated to induce interferon production, activating interferon-stimulated genes (ISGs) that encode antiviral proteins that exert antiviral effects. Viperin is one of the innate antiviral proteins that exert broad-spectrum antiviral effects by various mechanisms. Porcine epidemic diarrhea virus (PEDV) is a coronavirus that causes huge losses to the pig industry. Research on early antiviral responses in the gastrointestinal tract is essential for developing strategies to prevent the spread of PEDV. In this study, we investigated the mechanisms of viperin in PEDV-infected IPEJ-C2 cells. Increased expression of interferon and viperin and decreased replication of PEDV with a clear reduction in the viral load were observed in PEDV-infected IPEC-J2 cells. Amino acids 1-50 of porcine viperin contain an endoplasmic reticulum signal sequence that allows viperin to be anchored to the endoplasmic reticulum and are necessary for its function in inhibiting PEDV proliferation. The interaction of the viperin S-adenosylmethionine domain with the N protein of PEDV was confirmed via confocal laser scanning microscopy and co-immunoprecipitation. This interaction might interfere with viral replication or assembly to reduce virus proliferation. Our results highlight a potential mechanism whereby viperin is able to inhibit PEDV replication and play an antiviral role in innate immunity.


Asunto(s)
Antivirales/metabolismo , Interacciones Microbiota-Huesped/fisiología , Proteínas de la Nucleocápside/fisiología , Virus de la Diarrea Epidémica Porcina/fisiología , Animales , Línea Celular , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Inmunidad Innata , Interferones/biosíntesis , Ratones , Ratones Endogámicos BALB C , Proteínas de la Nucleocápside/antagonistas & inhibidores , Proteínas de la Nucleocápside/química , Virus de la Diarrea Epidémica Porcina/inmunología , Virus de la Diarrea Epidémica Porcina/patogenicidad , Dominios y Motivos de Interacción de Proteínas , Proteínas/química , Proteínas/genética , Proteínas/fisiología , Interferencia de ARN , Porcinos , Replicación Viral
16.
Acta Trop ; 211: 105619, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32634389

RESUMEN

Chikungunya virus (CHIKV) is a zoonotic arthropod-borne virus that has caused several outbreaks in tropical and subtropical areas worldwide during the last 50 years. The virus is known to target different human cell types throughout the course of infection including epithelial and endothelial cells, fibroblasts, primary monocytes and monocyte-derived macrophages (MDMs). The two latter are phagocytic cell populations of the innate immune system which are involved in some aspects of CHIKV pathogenesis. However, monocytes and macrophages also potentially contribute to the control of viral replication through the expression of different pattern recognition receptors sensing viral pathogens and subsequently, inducing an type I interferone (IFN-I)-dependent antiviral immune response. The aim of this study was to determine the modulation of the expression of Toll-like receptors (TLRs), cytokine secretion capabilities and antiviral factor production in monocytes and MDMs following infection with CHIKV. Moreover, we sought to determine the replication kinetics of CHIKV in these two cell populations. We found that the maximum peak of CHIKV replication was observed between 18- and 24-hours post-infection (hpi), while after that the is strongly reduced. Furthermore, CHIKV infection induced the pro-inflammatory cytokine production starting from the first 6 hpi in both monocytes and MDMs, with similar kinetics but different protein levels. In contrast, the kinetics of transcriptional expression of some TLRs were different between both cell types. In addition, IFN-I, 2',5'-oligoadenylate synthetase 1 (OAS1), and double-stranded RNA-activated protein kinase R (PKR) mRNA levels were detected in response to CHIKV infection of monocytes and MDMs, resulting the highest expression levels at 48 hpi. In conclusion, our data provides evidence that CHIKV infection activates the TLR pathways in primary monocytes and MDMs, which play a crucial role in CHIKV pathogenesis and/or host defense, differentially. However, additional studies are required to determine the functional role of TLRs in monocytes and MDMs.


Asunto(s)
Fiebre Chikungunya/inmunología , Citocinas/biosíntesis , Macrófagos/metabolismo , Monocitos/metabolismo , Animales , Fiebre Chikungunya/virología , Células Endoteliales/metabolismo , Fibroblastos , Humanos , Interferones/biosíntesis , Replicación Viral
17.
Antiviral Res ; 180: 104860, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32565134

RESUMEN

Middle East Respiratory Syndrome Coronavirus (MERS-CoV) causes severe respiratory in human with high mortality and it has been a challenge to determine optimum treatment for MERS-CoV-induced respiratory infection. Here, we observed the distribution of MERS-CoV receptors using human respiratory mucosa and also evaluated the contribution of interferon-lambdas (IFN-λs) in response to MERS-CoV infection using in vitro normal human nasal epithelial (NHNE) and bronchial epithelial (NHBE) cells. We found that the gene and protein expression of DPPIV, MERS-CoV receptor, were more dominantly located in nasal and bronchial epithelium although human nasal mucosa exhibited relatively lower DPPIV expression than lung parenchymal tissues. The quantitative mRNA level of the MERS-CoV envelope (upE) gene was significantly induced in MERS-CoV-infected cultured NHNE and NHBE cells until 3 days after infection. The induction of IFNs was identified in NHNE and NHBE cells after MERS-CoV infection and IFN-λs were predominantly increased in MERS-CoV-infected respiratory epithelial cells. Inoculation of IFN-λs to NHNE and NHBE cells suppressed MERS-CoV replication and in particular, IFN-λ4 showed a strong therapeutic effect in reducing MERS-CoV infection with higher induction of IFN-stimulated genes. Thus, IFN-λ has a decisive function in the respiratory epithelium that greatly limits MERS-CoV replication, and may be a key cytokine for better therapeutic outcomes against MERS-CoV infection in respiratory tract.


Asunto(s)
Antivirales/uso terapéutico , Interferones/uso terapéutico , Interleucinas/uso terapéutico , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Mucosa Respiratoria/virología , Replicación Viral/efectos de los fármacos , Bronquios/virología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Citocinas/metabolismo , Células Epiteliales/virología , Regulación Viral de la Expresión Génica , Humanos , Inmunidad Innata/inmunología , Interferones/biosíntesis , Interleucinas/biosíntesis , Mucosa Laríngea/virología , Pulmón/virología , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Reacción en Cadena de la Polimerasa , Mucosa Respiratoria/efectos de los fármacos
18.
J Virol ; 94(16)2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32461321

RESUMEN

The 5' cap methylation of viral RNA plays important roles in RNA stability, efficient translation, and immune evasion. Thus, RNA cap methylation is an attractive target for antiviral discovery and development of new live attenuated vaccines. For coronaviruses, RNA cap structure is first methylated at the guanine-N-7 (G-N-7) position by nonstructural protein 14 (nsp14), which facilitates and precedes the subsequent ribose 2'-O methylation by the nsp16-nsp10 complex. Using porcine epidemic diarrhea virus (PEDV), an Alphacoronavirus, as a model, we showed that G-N-7 methyltransferase (G-N-7 MTase) of PEDV nsp14 methylated RNA substrates in a sequence-unspecific manner. PEDV nsp14 can efficiently methylate RNA substrates with various lengths in both neutral and alkaline pH environments and can methylate cap analogs (GpppA and GpppG) and single-nucleotide GTP but not ATP, CTP, or UTP. Mutations to the S-adenosyl-l-methionine (SAM) binding motif in the nsp14 abolished the G-N-7 MTase activity and were lethal to PEDV. However, recombinant rPEDV-D350A with a single mutation (D350A) in nsp14, which retained 29.0% of G-N-7 MTase activity, was viable. Recombinant rPEDV-D350A formed a significantly smaller plaque and had significant defects in viral protein synthesis and viral replication in Vero CCL-81 cells and intestinal porcine epithelial cells (IPEC-DQ). Notably, rPEDV-D350A induced significantly higher expression of both type I and III interferons in IPEC-DQ cells than the parental rPEDV. Collectively, our results demonstrate that G-N-7 MTase activity of PEDV modulates viral replication, gene expression, and innate immune responses.IMPORTANCE Coronaviruses (CoVs) include a wide range of important human and animal pathogens. Examples of human CoVs include severe acute respiratory syndrome coronavirus (SARS-CoV-1), Middle East respiratory syndrome coronavirus (MERS-CoV), and the most recently emerged SARS-CoV-2. Examples of pig CoVs include porcine epidemic diarrhea virus (PEDV), porcine deltacoronavirus (PDCoV), and swine enteric alphacoronavirus (SeACoV). There are no vaccines or antiviral drugs for most of these viruses. All known CoVs encode a bifunctional nsp14 protein which possesses ExoN and guanine-N-7 methyltransferase (G-N-7 MTase) activities, responsible for replication fidelity and RNA cap G-N-7 methylation, respectively. Here, we biochemically characterized G-N-7 MTase of PEDV nsp14 and found that G-N-7 MTase-deficient PEDV was defective in replication and induced greater responses of type I and III interferons. These findings highlight that CoV G-N-7 MTase may be a novel target for rational design of live attenuated vaccines and antiviral drugs.


Asunto(s)
Exorribonucleasas/metabolismo , Interferón Tipo I/biosíntesis , Interferones/biosíntesis , Virus de la Diarrea Epidémica Porcina/fisiología , Caperuzas de ARN/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Sitios de Unión , Línea Celular , Chlorocebus aethiops , Exorribonucleasas/genética , Expresión Génica , Guanina/metabolismo , Inmunidad Innata , Metilación , Mutación , Virus de la Diarrea Epidémica Porcina/enzimología , Virus de la Diarrea Epidémica Porcina/genética , Virus de la Diarrea Epidémica Porcina/patogenicidad , ARN Viral/metabolismo , S-Adenosilmetionina/metabolismo , Porcinos , Células Vero , Proteínas no Estructurales Virales/genética , Replicación Viral , Interferón lambda
19.
J Virol ; 94(13)2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32295922

RESUMEN

Type I and type III interferons (IFNs) are the frontline of antiviral defense mechanisms that trigger hundreds of downstream antiviral genes. In this study, we observed that MERS-CoV nucleocapsid (N) protein suppresses type I and type III IFN gene expression. The N protein suppresses Sendai virus-induced IFN-ß and IFN-λ1 by reducing their promoter activity and mRNA levels, as well as downstream IFN-stimulated genes (ISGs). Retinoic acid-inducible gene I (RIG-I) is known to recognize viral RNA and induce IFN expression through tripartite motif-containing protein 25 (TRIM25)-mediated ubiquitination of RIG-I caspase activation and recruitment domains (CARDs). We discovered that MERS-CoV N protein suppresses RIG-I-CARD-induced, but not MDA5-CARD-induced, IFN-ß and IFN-λ1 promoter activity. By interacting with TRIM25, N protein impedes RIG-I ubiquitination and activation and inhibits the phosphorylation of transcription factors IFN-regulatory factor 3 (IRF3) and NF-κB that are known to be important for IFN gene activation. By employing a recombinant Sindbis virus-EGFP replication system, we showed that viral N protein downregulated the production of not only IFN mRNA but also bioactive IFN proteins. Taken together, MERS-CoV N protein functions as an IFN antagonist. It suppresses RIG-I-induced type I and type III IFN production by interfering with TRIM25-mediated RIG-I ubiquitination. Our study sheds light on the pathogenic mechanism of how MERS-CoV causes disease.IMPORTANCE MERS-CoV causes death of about 35% of patients. Published studies showed that some coronaviruses are capable of suppressing interferon (IFN) expression in the early phase of infection and MERS-CoV proteins can modulate host immune response. In this study, we demonstrated that MERS-CoV nucleocapsid (N) protein suppresses the production of both type I and type III IFNs via sequestering TRIM25, an E3 ubiquitin ligase that is essential for activating the RIG-I signaling pathway. Ectopic expression of TRIM25 rescues the suppressive effect of the N protein. In addition, the C-terminal domain of the viral N protein plays a pivotal role in the suppression of IFN-ß promoter activity. Our findings reveal how MERS-CoV evades innate immunity and provide insights into the interplay between host immune response and viral pathogenicity.


Asunto(s)
Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Proteína 58 DEAD Box/metabolismo , Interferón Tipo I/biosíntesis , Interferones/biosíntesis , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Proteínas de la Nucleocápside/metabolismo , Transducción de Señal , Proteínas Adaptadoras de Señalización CARD/metabolismo , Línea Celular , Infecciones por Coronavirus/genética , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/genética , Interferones/genética , Regiones Promotoras Genéticas , Unión Proteica , Receptores Inmunológicos , Factores de Transcripción , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Interferón lambda
20.
Sci Immunol ; 5(46)2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32303573

RESUMEN

Dendritic cells play a key role in the orchestration of antitumor immune responses. The cDC1 (conventional dendritic cell 1) subset has been shown to be essential for antitumor responses and response to immunotherapy, but its precise role in humans is largely unexplored. Using a multidisciplinary approach, we demonstrate that human cDC1 play an important role in the antitumor immune response through their capacity to produce type III interferon (IFN-λ). By analyzing a large cohort of breast primary tumors and public transcriptomic datasets, we observed specific production of IFN-λ1 by cDC1. In addition, both IFN-λ1 and its receptor were associated with favorable patient outcomes. We show that IFN-III promotes a TH1 microenvironment through increased production of IL-12p70, IFN-γ, and cytotoxic lymphocyte-recruiting chemokines. Last, we showed that engagement of TLR3 is a therapeutic strategy to induce IFN-III production by tumor-associated cDC1. These data provide insight into potential IFN- or cDC1-targeting antitumor therapies.


Asunto(s)
Neoplasias de la Mama/inmunología , Células Dendríticas/inmunología , Interferones/biosíntesis , Neoplasias de la Mama/diagnóstico , Femenino , Humanos , Inmunidad Innata/inmunología , Interferones/inmunología , Interferón lambda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA