Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.049
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.
J Virol ; 97(12): e0130423, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-37982645

RESUMEN

IMPORTANCE: Interferon-stimulated genes (ISGs) are induced in response to interferon expression due to viral infections. Role of these ISGs can be variable in different cells or organs. Our study highlights such cell-specific role of an ISG, Ddx3, which regulates the translation of mRNAs essential for interferon induction (PACT) and interferon signaling (STAT1) in a cell-specific manner. Our study also highlights the role of PACT in RNA virus-induced RLR signaling. Our study depicts how Ddx3 regulates innate immune signaling pathways in an indirect manner. Such cell-specific behavior of ISGs helps us to better understand viral pathogenesis and highlights the complexities of viral tropism and innate immune responses.


Asunto(s)
Inmunidad Innata , Interferones , Virus ARN , Inmunidad Innata/inmunología , Interferones/biosíntesis , Interferones/inmunología , Virus ARN/inmunología , Virus ARN/patogenicidad , Transducción de Señal , Humanos , Animales , Ratones
3.
J Virol ; 97(11): e0143423, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37882518

RESUMEN

IMPORTANCE: Mitochondrial antiviral signaling protein (MAVS) and stimulator of interferon (IFN) genes (STING) are key adaptor proteins required for innate immune responses to RNA and DNA virus infection. Here, we show that zebrafish transmembrane protein 47 (TMEM47) plays a critical role in regulating MAVS- and STING-triggered IFN production in a negative feedback manner. TMEM47 interacted with MAVS and STING for autophagic degradation, and ATG5 was essential for this process. These findings suggest the inhibitory function of TMEM47 on MAVS- and STING-mediated signaling responses during RNA and DNA virus infection.


Asunto(s)
Infecciones por Virus ADN , Inmunidad Innata , Interferones , Infecciones por Virus ARN , Proteínas de Pez Cebra , Pez Cebra , Animales , Infecciones por Virus ADN/inmunología , Infecciones por Virus ADN/virología , Interferones/antagonistas & inhibidores , Interferones/biosíntesis , Transducción de Señal , Pez Cebra/inmunología , Pez Cebra/metabolismo , Pez Cebra/virología , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/virología , Retroalimentación Fisiológica , Proteínas de Pez Cebra/inmunología , Proteínas de Pez Cebra/metabolismo
4.
J Immunol ; 210(4): 442-458, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36602826

RESUMEN

African swine fever is one of the most serious viral diseases that affects domestic and wild pigs. The causative agent, African swine fever virus (ASFV), has evolved sophisticated immune evasion mechanisms that target both innate and adaptive immune responses. However, the underlying molecular mechanisms have not been fully understood. Here, we report that ASFV E184L protein inhibits host innate immune response via targeting the stimulator of IFN genes (STING)-mediated signaling pathway in both human embryonic kidney HEK-293T cells and porcine pulmonary alveolar macrophages. E184L interacts with STING, impairing dimerization and oligomerization of STING but not affecting its puncta formation at the perinuclear region. Furthermore, E184L disrupts STING-TBK1-IRF3 complex formation, leading to inhibition of STING phosphorylation, and IRF3 dimerization and nuclear translocation. The 1-20 aa region in E184L is essential for E184L-STING interaction and blocking IL-1ß and type I IFN production. Deletion of E184L in ASFV considerably impairs antagonistic function of the virus in suppression of the STING-mediated antiviral response, an effect that is reversible by introduction of E184L. Importantly, the virulence of mutant ASFV lacking E184L is reduced in pigs compared with its parental virus due to induction of higher IFN production in vivo. Our findings indicate that ASFV E184L is an important antagonist of IFN signaling to evade host innate immune antiviral responses, which improves our understanding of immune evasion mechanisms of ASFV.


Asunto(s)
Virus de la Fiebre Porcina Africana , Fiebre Porcina Africana , Animales , Humanos , Antivirales/metabolismo , Inmunidad Innata , Porcinos , Proteínas Virales , Replicación Viral , Proteínas de la Membrana/metabolismo , Interferones/biosíntesis
5.
J Immunol ; 209(2): 326-336, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35777851

RESUMEN

Nervous necrosis virus (NNV), a highly pathogenic RNA virus, is a major pathogen in the global aquaculture industry. To efficiently infect fish, NNV must evade or subvert the host IFN for their replication; however, the precise mechanisms remain to be elucidated. In this study, we reported that capsid protein (CP) of red-spotted grouper NNV (RGNNV) suppressed the IFN antiviral response to promote RGNNV replication in Lateolabrax japonicus brain cells, which depended on the ARM, S, and P domains of CP. CP showed an indirect or direct association with the key components of retinoic acid-inducible gene-I-like receptors signaling, L. japonicus TNFR-associated factor 3 (LjTRAF3) and IFN regulatory factor (LjIRF3), respectively, and degraded LjTRAF3 and LjIRF3 through the ubiquitin-proteasome pathway in HEK293T cells. Furthermore, we found that CP potentiated LjTRAF3 K48 ubiquitination degradation in a L. japonicus ring finger protein 114-dependent manner. LjIRF3 interacted with CP through the S domain of CP and the transcriptional activation domain or regulatory domain of LjIRF3. CP promoted LjIRF3 K48 ubiquitination degradation, leading to the reduced phosphorylation level and nuclear translocation of LjIRF3. Taken together, we demonstrated that CP inhibited type I IFN response by a dual strategy to potentiate the ubiquitination degradation of LjTRAF3 and LjIRF3. This study reveals a novel mechanism of RGNNV evading host immune response via its CP protein that will provide insights into the complex pathogenesis of NNV.


Asunto(s)
Enfermedades de los Peces , Nodaviridae , Infecciones por Virus ARN , Animales , Proteínas de la Cápside , Proteínas de Peces/metabolismo , Peces/metabolismo , Células HEK293 , Humanos , Factores Reguladores del Interferón/metabolismo , Interferones/biosíntesis , Necrosis , Nodaviridae/fisiología , Tretinoina
6.
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
7.
Front Immunol ; 12: 666134, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867938

RESUMEN

Sjögren's syndrome (SjS) is a frequent systemic autoimmune disease responsible for a major decrease in patients' quality of life, potentially leading to life-threatening conditions while facing an unmet therapeutic need. Hence, we assessed the immunogenicity, efficacy, and tolerance of IFN-Kinoid (IFN-K), an anti-IFNα vaccination strategy, in a well-known mouse model of systemic autoimmunity with SjS-like features: MRL/MpJ-Faslpr/lpr (MRL/lpr) mice. Two cohorts (with ISA51 or SWE01 as adjuvants) of 26 female MRL/lpr were divided in parallel groups, "controls" (not treated, PBS and Keyhole Limpet Hemocyanin [KLH] groups) or "IFN-K" and followed up for 122 days. Eight-week-old mice received intra-muscular injections (days 0, 7, 28, 56 and 84) of PBS, KLH or IFN-K, emulsified in the appropriate adjuvant, and blood samples were serially collected. At sacrifice, surviving mice were euthanized and their organs were harvested for histopathological analysis (focus score in salivary/lacrimal glands) and IFN signature evaluation. SjS-like features were monitored. IFN-K induced a disease-modifying polyclonal anti-IFNα antibody response in all treated mice with high IFNα neutralization capacities, type 1 IFN signature's reduction and disease features' (ocular and oral sicca syndrome, neuropathy, focus score, glandular production of BAFF) improvement, as reflected by the decrease in Murine Sjögren's Syndrome Disease Activity Index (MuSSDAI) modelled on EULAR Sjögren's Syndrome Disease Activity Index (ESSDAI). No adverse effects were observed. We herein report on the strong efficacy of an innovative anti-IFNα vaccination strategy in a mouse model of SjS, paving the way for further clinical development (a phase IIb trial has just been completed in systemic lupus erythematosus with promising results).


Asunto(s)
Interferón-alfa/antagonistas & inhibidores , Síndrome de Sjögren/inmunología , Síndrome de Sjögren/terapia , Animales , Anticuerpos Neutralizantes/sangre , Autoanticuerpos/sangre , Autoinmunidad , Linfocitos B/inmunología , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Hemocianinas/administración & dosificación , Hemocianinas/inmunología , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/inmunología , Inmunoterapia Activa , Interferón-alfa/administración & dosificación , Interferón-alfa/inmunología , Interferones/biosíntesis , Interferones/genética , Aparato Lagrimal/inmunología , Aparato Lagrimal/patología , Ratones , Ratones Endogámicos MRL lpr , Glándulas Salivales/inmunología , Glándulas Salivales/patología , Síndrome de Sjögren/genética
8.
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
9.
Front Immunol ; 12: 750279, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858406

RESUMEN

SARS-CoV-2 coronavirus infection induces heterogeneous symptoms, ranging from asymptomatic to lethal forms. Severe forms usually occur in the elderly and/or individuals with comorbidities. Children generally remain asymptomatic to primary infection, suggesting that they may have an effective local innate immune response. IFN-I and -III have non-redundant protective roles against SARS-CoV-2, although sometimes damaging the host. The expression and role of anti-viral peptides during SARS-CoV-2 infection have thus far been little studied. We aimed to identify the innate immune molecules present at the SARS-CoV-2 entry point. We analyzed the mRNA levels of type I (IFN-α and -ß) and type III (IFN-λ1-3) interferons and selected antiviral peptides (i.e., ß-defensins 1-3, α-defensins [HNP1-3, HD5] pentraxin-3, surfactant protein D, the cathelicidin LL-37 and interleukin-26) in nasopharyngeal swabs from 226 individuals of various ages, either infected with SARS-CoV-2 (symptomatic or asymptomatic) or negative for the virus. We observed that infection induced selective upregulation of IFN-λ1 expression in pediatric subjects (≤15 years), whereas IFN-α, IFN-ß, IFN-λ2/λ3, and ß-defensin 1-3 expression was unaffected. Conversely, infection triggered upregulation of IFN-α, IFN-ß, IFN-λ2/λ3, and ß-defensin 1-3 mRNA expression in adults (15-65 years) and the elderly (≥ 65 years), but without modulation of IFN-λ1. The expression of these innate molecules was not associated with gender or symptoms. Expression of the interferon-stimulated genes IFITM1 and IFITM3 was upregulated in SARS-CoV-2-positive subjects and reached similar levels in the three age groups. Finally, age-related differences in nasopharyngeal innate immunity were also observed in SARS-CoV-2-negative subjects. This study shows that the expression patterns of IFN-I/-III and certain anti-viral molecules in the nasopharyngeal mucosa of SARS-CoV-2-infected subjects differ with age and suggests that susceptibility to SARS-CoV-2 may be related to intrinsic differences in the nature of mucosal anti-viral innate immunity.


Asunto(s)
Factores de Restricción Antivirales/análisis , Interferón Tipo I/biosíntesis , Interferón gamma/biosíntesis , Mucosa Nasal/inmunología , SARS-CoV-2/inmunología , beta-Defensinas/biosíntesis , Adolescente , Adulto , Factores de Edad , Anciano , COVID-19/inmunología , Células Cultivadas , Femenino , Humanos , Inmunidad Innata/inmunología , Interferón Tipo I/inmunología , Interferón gamma/inmunología , Interferones/biosíntesis , Interferones/inmunología , Interleucinas/biosíntesis , Interleucinas/inmunología , Masculino , Persona de Mediana Edad , Nasofaringe/inmunología , Adulto Joven , beta-Defensinas/inmunología , Interferón lambda
10.
Bull Exp Biol Med ; 172(1): 53-56, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34791556

RESUMEN

The antiviral activity of recombinant human IFN-lambda type 1 (IFNλ-1) against culture strain of SARS-CoV-2 virus was determined by infecting a highly sensitive VeroE6 coronavirus cell culture after preincubation test (the cell monolayer was incubated with 4-fold dilutions of IFNλ-1 in a concentration range of 0.16-42,500 ng/ml in a culture medium for 12 h at 37°C) and without preincubation (simultaneous addition of different concentrations of IFNλ-1 and SARS-CoV-2 infection in a dose of 102 TCID50). The created recombinant human IFNλ-1 demonstrated obvious antiviral activity against SARS-CoV-2 virus in vitro. In the tests with and without preincubation, IFNλ-1 exhibited significant activity, although somewhat lower in variant with simultaneous addition of IFNλ-1 and virus to the cell culture. It should be noted that the antiviral effect of IFNλ-1 was observed in a wide range of concentrations.


Asunto(s)
Antivirales/farmacología , Interferones/farmacología , Proteínas Recombinantes/farmacología , SARS-CoV-2/efectos de los fármacos , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Antivirales/aislamiento & purificación , COVID-19/virología , Chlorocebus aethiops , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Humanos , Interferones/biosíntesis , Interferones/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , SARS-CoV-2/genética , SARS-CoV-2/crecimiento & desarrollo , Células Vero , Carga Viral/genética , Tratamiento Farmacológico de COVID-19
11.
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
12.
J Gen Virol ; 102(10)2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34698626

RESUMEN

Human noroviruses (HuNoVs) are increasingly becoming the main cause of transmissible gastroenteritis worldwide, with hundreds of thousands of deaths recorded annually. Yet, decades after their discovery, there is still no effective treatment or vaccine. Efforts aimed at developing vaccines or treatment will benefit from a greater understanding of norovirus-host interactions, including the host response to infection. In this review, we provide a concise overview of the evidence establishing the significance of type I and type III interferon (IFN) responses in the restriction of noroviruses. We also critically examine our current understanding of the molecular mechanisms of IFN induction in norovirus-infected cells, and outline the diverse strategies deployed by noroviruses to supress and/or avoid host IFN responses. It is our hope that this review will facilitate further discussion and increase interest in this area.


Asunto(s)
Infecciones por Caliciviridae/inmunología , Infecciones por Caliciviridae/virología , Interferones/fisiología , Norovirus/inmunología , Norovirus/patogenicidad , Animales , Línea Celular , Humanos , Evasión Inmune , Inmunidad Innata , Interferones/biosíntesis , Proteínas Virales/metabolismo , Replicación Viral
13.
Int J Mol Sci ; 22(17)2021 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-34502167

RESUMEN

Hepatitis E virus (HEV) usually causes self-limiting acute hepatitis, but the disease can become chronic in immunocompromised individuals. HEV infection in pregnant women is reported to cause up to 30% mortality, especially in the third trimester. Additionally, extrahepatic manifestations like neuronal and renal diseases and pancreatitis are also reported during the course of HEV infection. The mechanism of HEV pathogenesis remains poorly understood. Innate immunity is the first line of defense triggered within minutes to hours after the first pathogenic insult. Growing evidence based on reverse genetics systems, in vitro cell culture models, and representative studies in animal models including non-human primates, has implicated the role of the host's innate immune response during HEV infection. HEV persists in presence of interferons (IFNs) plausibly by evading cellular antiviral defense. This review summarizes our current understanding of recognizing HEV-associated molecular patterns by host cell Pattern Recognition Receptors (PRRs) in eliciting innate immune response during HEV infection as well as mechanisms of virus-mediated immune evasion.


Asunto(s)
Virus de la Hepatitis E/fisiología , Hepatitis E/metabolismo , Hepatitis E/virología , Interacciones Huésped-Patógeno , Receptores de Reconocimiento de Patrones/metabolismo , Animales , Biomarcadores , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Regulación Viral de la Expresión Génica , Hepatitis E/genética , Hepatitis E/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interferones/biosíntesis , Unión Proteica , Receptores de Reconocimiento de Patrones/genética
14.
Front Immunol ; 12: 718744, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34531865

RESUMEN

COVID-19 associated multisystem inflammatory syndrome (MIS) is a rare condition mostly affecting children but also adults (MIS-A). Although severe systemic inflammation and multiorgan dysfunction are hallmarks of the syndrome, the underlying pathogenesis is unclear. We aimed to provide novel immunological and genetic descriptions of MIS-A patients. Cytokine responses (IL-6, IL-1ß, TNFα, CXCL10, type I, II and III interferons) following SARS-CoV-2 infection of peripheral blood mononuclear cells in vitro were analyzed as well as antibodies against IFNα and IFNω (by ELISA) in patients and healthy controls. We also performed whole exome sequencing (WES) of patient DNA. A total of five patients (ages 19, 23, 33, 38, 50 years) were included. The patients shared characteristic features, although organ involvement and the time course of disease varied slightly. SARS-CoV-2 in vitro infection of patient PBMCs revealed impaired type I and III interferon responses and reduced CXCL10 expression, whereas production of proinflammatory cytokines were less affected, compared to healthy controls. Presence of interferon autoantibodies was not detected. Whole exome sequencing analysis of patient DNA revealed 12 rare potentially disease-causing variants in genes related to autophagy, classical Kawasaki disease, restriction factors and immune responses. In conclusion, we observed an impaired production of type I and III interferons in response to SARS-CoV-2 infection and detected several rare potentially disease-causing gene variants potentially contributing to MIS-A.


Asunto(s)
COVID-19/patología , Citocinas/sangre , Interferón-alfa/biosíntesis , Interferones/biosíntesis , Síndrome de Respuesta Inflamatoria Sistémica/patología , Adulto , Autoanticuerpos/sangre , Quimiocina CXCL10/biosíntesis , Comorbilidad , Exoma/genética , Femenino , Humanos , Interferón-alfa/inmunología , Interferones/inmunología , Leucocitos Mononucleares/inmunología , Masculino , Persona de Mediana Edad , SARS-CoV-2/inmunología , Secuenciación del Exoma , Adulto Joven , Interferón lambda
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...