Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.541
Filtrar
1.
Sci Rep ; 14(1): 5731, 2024 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-38459088

RESUMEN

Triple-negative breast cancer (TNBC) is one of the most aggressive types of cancer. Despite decades of intense investigation, treatment options remain limited, and rapid recurrence with distant metastases remains a significant challenge. Cancer cell-intrinsic production of cytokines such as type I interferons (IFN-I) is a known potent modulator of response to therapy in many cancers, including TNBC, and can influence therapeutic outcome. Here, we report that, in TNBC systems, the aryl hydrocarbon receptor (AhR) suppresses IFN-I expression via inhibition of STImulator of Interferon Genes (STING), a key mediator of interferon production. Intratumoral STING activity is essential in mediating the efficacy of PARP inhibitors (PARPi) which are used in the treatment of cancers harboring BRCA1 deficiency. We find that, in TNBC cells, PARPi treatment activates AhR in a BRCA1 deficiency-dependent manner, thus suggesting the presence of a negative feedback loop aimed at modulating PARPi efficacy. Importantly, our results indicate that the combined inhibition of PARP and AhR is superior in elevating IFN-I expression as compared to PARPi-alone. Thus, AhR inhibition may allow for enhanced IFN-I production upon PARPi in BRCA1-deficient breast cancers, most of which are of TNBC origin, and may represent a therapeutically viable strategy to enhance PARPi efficacy.


Asunto(s)
Interferón Tipo I , Neoplasias de la Mama Triple Negativas , Humanos , Proteína BRCA2/genética , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Receptores de Hidrocarburo de Aril/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
2.
J Virol ; 98(2): e0168223, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38289117

RESUMEN

Porcine deltacoronavirus (PDCoV) has caused enormous economic losses to the global pig industry. However, the immune escape mechanism of PDCoV remains to be fully clarified. Transcriptomic analysis revealed a high abundance of interferon (IFN)-induced protein with tetratricopeptide repeats 3 (IFIT3) transcripts after PDCoV infection, which initially implied a correlation between IFIT3 and PDCoV. Further studies showed that PDCoV nsp5 could antagonize the host type I interferon signaling pathway by cleaving IFIT3. We demonstrated that PDCoV nsp5 cleaved porcine IFIT3 (pIFIT3) at Gln-406. Similar cleavage of endogenous IFIT3 has also been observed in PDCoV-infected cells. The pIFIT3-Q406A mutant was resistant to nsp5-mediated cleavage and exhibited a greater ability to inhibit PDCoV infection than wild-type pIFIT3. Furthermore, we found that cleavage of IFIT3 is a common characteristic of nsp5 proteins of human coronaviruses, albeit not alphacoronavirus. This finding suggests that the cleavage of IFIT3 is an important mechanism by which PDCoV nsp5 antagonizes IFN signaling. Our study provides new insights into the mechanisms by which PDCoV antagonizes the host innate immune response.IMPORTANCEPorcine deltacoronavirus (PDCoV) is a potential emerging zoonotic pathogen, and studies on the prevalence and pathogenesis of PDCoV are ongoing. The main protease (nsp5) of PDCoV provides an excellent target for antivirals due to its essential and conserved function in the viral replication cycle. Previous studies have revealed that nsp5 of PDCoV antagonizes type I interferon (IFN) production by targeting the interferon-stimulated genes. Here, we provide the first demonstration that nsp5 of PDCoV antagonizes IFN signaling by cleaving IFIT3, which affects the IFN response after PDCoV infection. Our findings reveal that PDCoV nsp5 is an important interferon antagonist and enhance the understanding of immune evasion by deltacoronaviruses.


Asunto(s)
Proteasas 3C de Coronavirus , Infecciones por Coronavirus , Deltacoronavirus , Interferón Tipo I , Péptidos y Proteínas de Señalización Intracelular , Enfermedades de los Porcinos , Porcinos , Animales , Humanos , Proteasas 3C de Coronavirus/metabolismo , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Deltacoronavirus/enzimología , Deltacoronavirus/metabolismo , Deltacoronavirus/patogenicidad , Inmunidad Innata , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteolisis , Transducción de Señal/inmunología , Porcinos/inmunología , Porcinos/virología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/metabolismo , Enfermedades de los Porcinos/virología , Factores de Transcripción/metabolismo , Zoonosis Virales/inmunología , Zoonosis Virales/virología , Replicación Viral
3.
J Virol ; 97(10): e0092623, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37754758

RESUMEN

IMPORTANCE: Type I interferon (IFN-I), produced by the innate immune system, plays an essential role in host antiviral responses. Proper regulation of IFN-I production is required for the host to balance immune responses and prevent superfluous inflammation. IFN regulatory factor 3 (IRF3) and subsequent sensors are activated by RNA virus infection to induce IFN-I production. Therefore, proper regulation of IRF3 serves as an important way to control innate immunity and viral replication. Here, we first identified Prohibitin1 (PHB1) as a negative regulator of host IFN-I innate immune responses. Mechanistically, PHB1 inhibited the nucleus import of IRF3 by impairing its binding with importin subunit alpha-1 and importin subunit alpha-5. Our study demonstrates the mechanism by which PHB1 facilitates the replication of multiple RNA viruses and provides insights into the negative regulation of host immune responses.


Asunto(s)
Proteína 58 DEAD Box , Prohibitinas , Virus ARN , Receptores Inmunológicos , Transducción de Señal , Replicación Viral , Proteína 58 DEAD Box/antagonistas & inhibidores , Proteína 58 DEAD Box/metabolismo , Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Carioferinas/metabolismo , Prohibitinas/metabolismo , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/metabolismo , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Virus ARN/crecimiento & desarrollo , Virus ARN/inmunología , Virus ARN/metabolismo
4.
J Immunol ; 209(10): 2012-2021, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36426941

RESUMEN

Germline-encoded pattern recognition receptors (PRRs) recognize molecules frequently found in pathogens (pathogen-associated molecular patterns [PAMPs]) during viral infection. This process induces production of IFNs, leading to expression of IFN-stimulated genes to establish a cellular antiviral state against viral infection. However, aberrant activation of the IFN system may cause immunopathological damage and systemic autoimmune diseases such as systemic lupus erythematosus. Stringent control of IFN signaling activation is critical for maintaining homoeostasis of the immune system; yet, the mechanisms responsible for its precise regulation remain to be elucidated. In this study, we identified that ring finger protein 215 (RNF215), a zinc finger protein, was upregulated by viral infection in human macrophages. In addition, we demonstrated that RNF215 inhibited the production of type I IFNs at least in part via interacting with p65, a subunit of NF-κB, and repressed the accumulation of NF-κB in the promoter region of IFNB1. Moreover, we found that the expression of RNF215 negatively correlated with type I IFNs in patients with systemic lupus erythematosus, indicating that RNF215 plays an important role in the pathogenesis of autoimmune diseases. Collectively, our data identified RNF215 as a key negative regulator of type I IFNs and suggested RNF215 as a potential target for intervention in diseases with aberrant IFN production.


Asunto(s)
Enfermedades Autoinmunes , Lupus Eritematoso Sistémico , Humanos , Interferón Tipo I/biosíntesis , FN-kappa B , Moléculas de Patrón Molecular Asociado a Patógenos , Transducción de Señal
5.
Cell Mol Life Sci ; 79(3): 191, 2022 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-35292881

RESUMEN

Immune checkpoint blockade (ICB) therapies have achieved remarkable clinical responses in patients with many different types of cancer; however, most patients who receive ICB monotherapy fail to achieve long-term responses, and some tumors become immunotherapy-resistant and even hyperprogressive. Type I interferons (IFNs) have been demonstrated to inhibit tumor growth directly and indirectly by acting upon tumor and immune cells, respectively. Furthermore, accumulating evidence indicates that endo- and exogenously enhancing type I IFNs have a synergistic effect on anti-tumor immunity. Therefore, clinical trials studying new treatment strategies that combine type I IFN inducers with ICB are currently in progress. Here, we review the cellular sources of type I IFNs and their roles in the immune regulation of the tumor microenvironment. In addition, we highlight immunotherapies based on type I IFNs and combination therapy between type I IFN inducers and ICBs.


Asunto(s)
Inmunoterapia/métodos , Interferón Tipo I/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Animales , Fibroblastos Asociados al Cáncer/inmunología , Terapia Combinada , Células Dendríticas/inmunología , Células Endoteliales/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Interferón Tipo I/biosíntesis , Células Asesinas Naturales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Macrófagos/inmunología , Ratones , Modelos Inmunológicos , Células Supresoras de Origen Mieloide/inmunología , Neutrófilos/inmunología , Viroterapia Oncolítica , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Receptores Toll-Like/agonistas , Microambiente Tumoral/inmunología
6.
Am J Respir Cell Mol Biol ; 66(6): 671-681, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35358404

RESUMEN

Bacterial pneumonia induces the rapid recruitment and activation of neutrophils and macrophages into the lung, and these cells contribute to bacterial clearance and other defense functions. TBK1 (TANK-binding kinase 1) performs many functions, including activation of the type I IFN pathway and regulation of autophagy and mitophagy, but its contribution to antibacterial defenses in the lung is unclear. We previously showed that lung neutrophils upregulate mRNAs for TBK1 and its accessory proteins during Streptococcus pneumoniae pneumonia, despite low or absent expression of type I IFN in these cells. We hypothesized that TBK1 performs key antibacterial functions in pneumonia apart from type I IFN expression. Using TBK1 null mice, we show that TBK1 contributes to antibacterial defenses and promotes bacterial clearance and survival. TBK1 null mice express lower concentrations of many cytokines in the infected lung. Conditional deletion of TBK1 with LysMCre results in TBK1 deletion from macrophages but not neutrophils. LysMCre TBK1 mice have no defect in cytokine expression, implicating a nonmacrophage cell type as a key TBK1-dependent cell. TBK1 null neutrophils have no defect in recruitment to the infected lung but show impaired activation of p65/NF-κB and STAT1 and lower expression of reactive oxygen species, IFNγ, and IL12p40. TLR1/2 and 4 agonists each induce phosphorylation of TBK1 in neutrophils. Surprisingly, neutrophil TBK1 activation in vivo does not require the adaptor STING. Thus, TBK1 is a critical component of STING-independent antibacterial responses in the lung, and TBK1 is necessary for multiple neutrophil functions.


Asunto(s)
Interferón Tipo I , Neumonía Neumocócica , Proteínas Serina-Treonina Quinasas , Streptococcus pneumoniae , Animales , Citocinas/inmunología , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/microbiología , Proteínas Serina-Treonina Quinasas/inmunología , Transducción de Señal , Streptococcus pneumoniae/inmunología
7.
J Immunol ; 208(3): 753-761, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34996837

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), has seriously threatened global public health. Severe COVID-19 has been reported to be associated with an impaired IFN response. However, the mechanisms of how SARS-CoV-2 antagonizes the host IFN response are poorly understood. In this study, we report that SARS-CoV-2 helicase NSP13 inhibits type I IFN production by directly targeting TANK-binding kinase 1 (TBK1) for degradation. Interestingly, inhibition of autophagy by genetic knockout of Beclin1 or pharmacological inhibition can rescue NSP13-mediated TBK1 degradation in HEK-293T cells. Subsequent studies revealed that NSP13 recruits TBK1 to p62, and the absence of p62 can also inhibit TBK1 degradation in HEK-293T and HeLa cells. Finally, TBK1 and p62 degradation and p62 aggregation were observed during SARS-CoV-2 infection in HeLa-ACE2 and Calu3 cells. Overall, our study shows that NSP13 inhibits type I IFN production by recruiting TBK1 to p62 for autophagic degradation, enabling it to evade the host innate immune response, which provides new insights into the transmission and pathogenesis of SARS-CoV-2 infection.


Asunto(s)
Autofagia , COVID-19/inmunología , ARN Polimerasa Dependiente de ARN de Coronavirus/fisiología , Interferón Tipo I/biosíntesis , Metiltransferasas/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Helicasas/fisiología , SARS-CoV-2/fisiología , Proteína Sequestosoma-1/metabolismo , Proteínas no Estructurales Virales/fisiología , Beclina-1/antagonistas & inhibidores , Línea Celular , Regulación hacia Abajo , Humanos , Evasión Inmune , Inmunidad Innata , Inmunoprecipitación , Interferón Tipo I/genética , Complejos Multiproteicos , Agregado de Proteínas , Mapeo de Interacción de Proteínas
8.
Int Immunol ; 34(1): 21-33, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34648636

RESUMEN

Plasmodium parasites that infect humans are highly polymorphic, and induce various infections ranging from an asymptomatic state to life-threatening diseases. However, how the differences between the parasites affect host immune responses during blood-stage infection remains largely unknown. We investigated the CD4+ T-cell immune responses in mice infected with P. berghei ANKA (PbA) or P. chabaudi chabaudi AS (Pcc) using PbT-II cells, which recognize a common epitope of these parasites. In the acute phase of infection, CD4+ T-cell responses in PbA-infected mice showed a lower involvement of Th1 cells and a lower proportion of Ly6Clo effector CD4+ T cells than those in Pcc-infected mice. Transcriptome analysis of PbT-II cells indicated that type I interferon (IFN)-regulated genes were expressed at higher levels in both Th1- and Tfh-type PbT-II cells from PbA-infected mice than those from Pcc-infected mice. Moreover, IFN-α levels were considerably higher in PbA-infected mice than in Pcc-infected mice. Inhibition of type I IFN signaling increased PbT-II and partially reversed the Th1 over Tfh bias of the PbT-II cells in both PbA- and Pcc-infected mice. In the memory phase, PbT-II cells in PbA-primed mice maintained higher numbers and exhibited a better recall response to the antigen. However, recall responses were not significantly different between the infection groups after re-challenge with PbA, suggesting the effect of the inflammatory environment by the infection. These observations suggest that the differences in Plasmodium-specific CD4+ T-cell responses between PbA- and Pcc-infected mice were associated with the difference in type I IFN production during the early phase of the infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Interferón Tipo I/biosíntesis , Malaria/inmunología , Plasmodium berghei/inmunología , Plasmodium chabaudi/inmunología , Animales , Células Cultivadas , Ratones , Ratones Transgénicos
9.
Mol Immunol ; 142: 11-21, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34959069

RESUMEN

Senecavirus A (SVA) is an oncolytic virus, which can propagate in human tumor cells and has been used as an oncolytic virotherapy candidate in humans. Besides, SVA circulates in pigs and causes vesicles and coalescing erosions on the snouts and coronary bands in infected pigs and results in neonatal morbidity. SVA has evolved the ability to suppress host innate immune response to benefit viral replication. SVA 3Cpro and 2C protein inhibit the production of host type I interferon (IFN) by degradation of several components of innate immune pathway. In this study, for the first time, we determined that SVA 2B antagonized host innate immune response in both human and porcine cells. SVA 2B protein degraded mitochondrial antiviral-signaling protein (MAVS), a key host molecule in the innate immune pathway, and a colocalization and interaction between 2B and MAVS was observed in the context of viral infection. Further study showed that the 1-48 and 100-128 regions of 2B were essential for inhibition of type I IFN expression. In addition, we determined that 2B degraded MAVS depending on caspase-9 and caspase-3. In conclusion, our results revealed a novel strategy for SVA 2B protein to antagonize host innate immune response, which will help for clarification of the pathogenesis of SVA and provide an insight for oncolytic virotherapy of SVA.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inmunidad Innata/inmunología , Interferón Tipo I/biosíntesis , Picornaviridae/metabolismo , Proteínas no Estructurales Virales/inmunología , Animales , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Línea Celular , Células HEK293 , Interacciones Huésped-Patógeno/inmunología , Humanos , Interferón Tipo I/inmunología , Viroterapia Oncolítica/métodos , Infecciones por Picornaviridae/patología , Infecciones por Picornaviridae/virología , Porcinos , Proteínas no Estructurales Virales/genética , Proteínas Viroporinas/genética , Proteínas Viroporinas/inmunología , Liberación del Virus/fisiología , Replicación Viral/fisiología
10.
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
11.
Front Immunol ; 12: 756262, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858409

RESUMEN

A male sex bias has emerged in the COVID-19 pandemic, fitting to the sex-biased pattern in other viral infections. Males are 2.84 times more often admitted to the ICU and mortality is 1.39 times higher as a result of COVID-19. Various factors play a role in this, and novel studies suggest that the gene-dose of Toll-Like Receptor (TLR) 7 could contribute to the sex-skewed severity. TLR7 is one of the crucial pattern recognition receptors for SARS-CoV-2 ssRNA and the gene-dose effect is caused by X chromosome inactivation (XCI) escape. Female immune cells with TLR7 XCI escape have biallelic TLR7 expression and produce more type 1 interferon (IFN) upon TLR7 stimulation. In COVID-19, TLR7 in plasmacytoid dendritic cells is one of the pattern recognition receptors responsible for IFN production and a delayed IFN response has been associated with immunopathogenesis and mortality. Here, we provide a hypothesis that females may be protected to some extend against severe COVID-19, due to the biallelic TLR7 expression, allowing them to mount a stronger and more protective IFN response early after infection. Studies exploring COVID-19 treatment via the TLR7-mediated IFN pathway should consider this sex difference. Various factors such as age, sex hormones and escape modulation remain to be investigated concerning the TLR7 gene-dose effect.


Asunto(s)
COVID-19/mortalidad , Dosificación de Gen/genética , Interferón Tipo I/biosíntesis , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo , COVID-19/patología , Cromosomas Humanos X/genética , Cuidados Críticos/estadística & datos numéricos , Células Dendríticas/inmunología , Femenino , Humanos , Interferón Tipo I/inmunología , Masculino , ARN Viral/genética , Receptores de Reconocimiento de Patrones/genética , Receptores de Reconocimiento de Patrones/metabolismo , Factores de Riesgo , SARS-CoV-2/inmunología , Factores Sexuales , Transducción de Señal/inmunología , Inactivación del Cromosoma X/genética , Tratamiento Farmacológico de COVID-19
12.
Front Immunol ; 12: 744880, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34956178

RESUMEN

Dengue Virus (DENV) infection can cause severe illness such as highly fatality dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Innate immune activation by Nod-like receptors (NLRs) is a critical part of host defense against viral infection. Here, we revealed a key mechanism of NLRP12-mediated regulation in DENV infection. Firstly, NLRP12 expression was inhibited in human macrophage following DENV or other flaviviruses (JEV, YFV, ZIKV) infection. Positive regulatory domain 1 (PRDM1) was induced by DENV or poly(I:C) and suppressed NLRP12 expression, which was dependent on TBK-1/IRF3 and NF-κB signaling pathways. Moreover, NLRP12 inhibited DENV and other flaviviruses (JEV, YFV, ZIKV) replication, which relied on the well-conserved nucleotide binding structures of its NACHT domain. Furthermore, NLRP12 could interact with heat shock protein 90 (HSP90) dependent on its Walker A and Walker B sites. In addition, NLRP12 enhanced the production of type I IFNs (IFN-α/ß) and interferon-stimulated genes (ISGs), including IFITM3, TRAIL and Viperin. Inhibition of HSP90 with 17-DMAG impaired the upregulation of type I IFNs and ISGs induced by NLRP12. Taken together, we demonstrated a novel mechanism that NLRP12 exerted anti-viral properties in DENV and other flaviviruses (JEV, YFV, ZIKV) infection, which brings up a potential target for the treatment of DENV infection.


Asunto(s)
Virus del Dengue/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Adolescente , Adulto , Anciano , Femenino , Humanos , Interferón Tipo I/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Persona de Mediana Edad , Replicación Viral/inmunología , Adulto Joven
13.
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
14.
Front Immunol ; 12: 758190, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867997

RESUMEN

Plasmacytoid dendritic cells (pDCs) are the key producers of type I interferons (IFNs), thus playing a central role in initiating antiviral immune response. Besides robust type I IFN production, pDCs also act as antigen presenting cells post immunogenic stimulation. Transcription factor Irf8 is indispensable for the development of both pDC and cDC1 subset. However, the mechanism underlying the differential regulation by IRF8 in cDC1- and pDC-specific genomic architecture of developmental pathways still remains to be fully elucidated. Previous studies indicated that the Irf8R294C mutation specifically abrogates development of cDC1 without affecting that of pDC. In the present study using RNA-seq based approach, we have found that though the point mutation Irf8R294C did not affect pDC development, it led to defective type I IFN production, thus resulting in inefficient antiviral response. This observation unraveled the distinctive roles of IRF8 in these two subpopulations-regulating the development of cDC1 whereas modulating the functionality of pDCs without affecting development. We have reported here that Irf8R294C mutation also caused defect in production of ISGs as well as defective upregulation of costimulatory molecules in pDCs in response to NDV infection (or CpG stimulation). Through in vivo studies, we demonstrated that abrogation of type I IFN production was concomitant with reduced upregulation of costimulatory molecules in pDCs and increased NDV burden in IRF8R294C mice in comparison with wild type, indicating inefficient viral clearance. Further, we have also shown that Irf8R294C mutation abolished the activation of type I IFN promoter by IRF8, justifying the low level of type I IFN production. Taken together, our study signifies that the single point mutation in Irf8, Irf8R294C severely compromised type I IFN-mediated immune response by murine pDCs, thereby causing impairment in antiviral immunity.


Asunto(s)
Células Dendríticas/inmunología , Factores Reguladores del Interferón/genética , Interferón Tipo I/inmunología , Mutación Missense , Enfermedad de Newcastle/inmunología , Mutación Puntual , Animales , Neoplasias Óseas/patología , Línea Celular Tumoral , Islas de CpG/inmunología , Células Dendríticas/metabolismo , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inmunidad Innata , Factores Reguladores del Interferón/inmunología , Interferón Tipo I/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Virus de la Enfermedad de Newcastle , Osteosarcoma/patología , Transcriptoma
15.
PLoS Comput Biol ; 17(11): e1009587, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34818337

RESUMEN

Patients with coronavirus disease 2019 (COVID-19) often exhibit diverse disease progressions associated with various infectious ability, symptoms, and clinical treatments. To systematically and thoroughly understand the heterogeneous progression of COVID-19, we developed a multi-scale computational model to quantitatively understand the heterogeneous progression of COVID-19 patients infected with severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2). The model consists of intracellular viral dynamics, multicellular infection process, and immune responses, and was formulated using a combination of differential equations and stochastic modeling. By integrating multi-source clinical data with model analysis, we quantified individual heterogeneity using two indexes, i.e., the ratio of infected cells and incubation period. Specifically, our simulations revealed that increasing the host antiviral state or virus induced type I interferon (IFN) production rate can prolong the incubation period and postpone the transition from asymptomatic to symptomatic outcomes. We further identified the threshold dynamics of T cell exhaustion in the transition between mild-moderate and severe symptoms, and that patients with severe symptoms exhibited a lack of naïve T cells at a late stage. In addition, we quantified the efficacy of treating COVID-19 patients and investigated the effects of various therapeutic strategies. Simulations results suggested that single antiviral therapy is sufficient for moderate patients, while combination therapies and prevention of T cell exhaustion are needed for severe patients. These results highlight the critical roles of IFN and T cell responses in regulating the stage transition during COVID-19 progression. Our study reveals a quantitative relationship underpinning the heterogeneity of transition stage during COVID-19 progression and can provide a potential guidance for personalized therapy in COVID-19 patients.


Asunto(s)
COVID-19/etiología , SARS-CoV-2 , Antivirales/uso terapéutico , COVID-19/inmunología , COVID-19/terapia , Biología Computacional , Simulación por Computador , Progresión de la Enfermedad , Interacciones Microbiota-Huesped/inmunología , Humanos , Interferón Tipo I/biosíntesis , Activación de Linfocitos , Modelos Inmunológicos , Modelos Estadísticos , Pandemias/estadística & datos numéricos , Pronóstico , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Índice de Severidad de la Enfermedad , Linfocitos T/inmunología , Resultado del Tratamiento
16.
Sci Rep ; 11(1): 19794, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34611284

RESUMEN

Adoptive T-cell transfer (ACT) offers a curative therapeutic option for subsets of melanoma and hematological cancer patients. To increase response rates and broaden the applicability of ACT, it is necessary to improve the post-infusion performance of the transferred T cells. The design of improved treatment strategies includes transfer of cells with a less differentiated phenotype. Such T cell subsets have high proliferative potential but require stimulatory signals in vivo to differentiate into tumor-reactive effector T cells. Thus, combination strategies are needed to support the therapeutic implementation of less differentiated T cells. Here we show that systemic delivery of tumor-associated antigens (TAAs) facilitates in vivo priming and expansion of previously non-activated T cells and enhance the cytotoxicity of activated T cells. To achieve this in vivo priming, we use flexible delivery vehicles of TAAs and a TLR7/8 agonist. Contrasting subcutaneous delivery systems, these vehicles accumulate TAAs in the spleen, thereby achieving close proximity to both cross-presenting dendritic cells and transferred T cells, resulting in robust T-cell expansion and anti-tumor reactivity. This TAA delivery platform offers a strategy to safely potentiate the post-infusion performance of T cells using low doses of antigen and TLR7/8 agonist, and thereby enhance the effect of ACT.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Inmunoterapia Adoptiva , Neoplasias/inmunología , Neoplasias/terapia , Animales , Presentación de Antígeno , Antígenos de Neoplasias/administración & dosificación , Biomarcadores , Terapia Combinada , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Epítopos/administración & dosificación , Epítopos/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunomodulación , Inmunoterapia Adoptiva/métodos , Interferón Tipo I/biosíntesis , Liposomas , Activación de Linfocitos/inmunología , Neoplasias/diagnóstico , Neoplasias/mortalidad , Especificidad del Receptor de Antígeno de Linfocitos T , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Resultado del Tratamiento , Escape del Tumor/inmunología
17.
Viruses ; 13(10)2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34696490

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a member of the Coronaviridae family, which is responsible for the COVID-19 pandemic followed by unprecedented global societal and economic disruptive impact. The innate immune system is the body's first line of defense against invading pathogens and is induced by a variety of cellular receptors that sense viral components. However, various strategies are exploited by SARS-CoV-2 to disrupt the antiviral innate immune responses. Innate immune dysfunction is characterized by the weak generation of type I interferons (IFNs) and the hypersecretion of pro-inflammatory cytokines, leading to mortality and organ injury in patients with COVID-19. This review summarizes the existing understanding of the mutual effects between SARS-CoV-2 and the type I IFN (IFN-α/ß) responses, emphasizing the relationship between host innate immune signaling and viral proteases with an insight on tackling potential therapeutic targets.


Asunto(s)
COVID-19/inmunología , Evasión Inmune/inmunología , Inmunidad Innata/inmunología , Interferón Tipo I/inmunología , SARS-CoV-2/inmunología , Antivirales/uso terapéutico , COVID-19/patología , Citocinas/metabolismo , Combinación de Medicamentos , Humanos , Interferón Tipo I/biosíntesis , Lopinavir/uso terapéutico , Ribavirina/uso terapéutico , Ritonavir/uso terapéutico , Transducción de Señal/inmunología , Tratamiento Farmacológico de COVID-19
18.
Viruses ; 13(9)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34578420

RESUMEN

Type I Interferons (IFN-I) are a family of potent antiviral cytokines that act through the direct restriction of viral replication and by enhancing antiviral immunity. However, these powerful cytokines are a caged lion, as excessive and sustained IFN-I production can drive immunopathology during infection, and aberrant IFN-I production is a feature of several types of autoimmunity. As specialized producers of IFN-I plasmacytoid (p), dendritic cells (DCs) can secrete superb quantities and a wide breadth of IFN-I isoforms immediately after infection or stimulation, and are the focus of this review. Notably, a few days after viral infection pDCs tune down their capacity for IFN-I production, producing less cytokines in response to both the ongoing infection and unrelated secondary stimulations. This process, hereby referred to as "pDC exhaustion", favors viral persistence and associates with reduced innate responses and increased susceptibility to secondary opportunistic infections. On the other hand, pDC exhaustion may be a compromise to avoid IFN-I driven immunopathology. In this review we reflect on the mechanisms that initially induce IFN-I and subsequently silence their production by pDCs during a viral infection. While these processes have been long studied across numerous viral infection models, the 2019 coronavirus disease (COVID-19) pandemic has brought their discussion back to the fore, and so we also discuss emerging results related to pDC-IFN-I production in the context of COVID-19.


Asunto(s)
COVID-19/metabolismo , COVID-19/virología , Interacciones Huésped-Patógeno , Interferón Tipo I/biosíntesis , SARS-CoV-2/fisiología , Biomarcadores , COVID-19/inmunología , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/inmunología , Inmunomodulación , Receptores Toll-Like/metabolismo
19.
Infect Immun ; 89(11): e0040721, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34370509

RESUMEN

During chronic infection with Helicobacter pylori, Schlafen 4-expressing myeloid-derived suppressor cells (SLFN4+ MDSCs) create a microenvironment favoring intestinal metaplasia and neoplastic transformation. SLFN4 can be induced by alpha interferon (IFN-α), which is mainly secreted from plasmacytoid dendritic cells (pDCs). This study tested the hypothesis that Helicobacter pylori infection promotes SLFN4+ MDSC differentiation by inducing pDCs to secrete IFN-α. C57BL/6 mice were gavaged with H. pylori, and infection lasted 2, 4, or 6 months. Mouse pDCs were isolated from bone marrow of wild-type C57BL/6J mice. The results showed that H. pylori infection increased the number of SLFN4+ MDSCs by inducing IFN-α expression in mice. Further mechanistic experiments unraveled that IFN-α induced SLFN4 transcription by binding to the Slfn4 promoter. Furthermore, H. pylori infection stimulated pDCs to secrete IFN-α by activating the TLR9-MyD88-IRF7 pathway. Collectively, Helicobacter pylori infection promotes SLFN4+ MDSC differentiation by inducing secretion of IFN-α from pDCs.


Asunto(s)
Proteínas Portadoras/genética , Células Dendríticas/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori , Interferón Tipo I/biosíntesis , Células Supresoras de Origen Mieloide/citología , Animales , Diferenciación Celular , Factor 7 Regulador del Interferón/fisiología , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/fisiología , Regiones Promotoras Genéticas , Receptor Toll-Like 9/fisiología
20.
J Virol ; 95(19): e0065221, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34346762

RESUMEN

The filovirus family includes deadly pathogens such as Ebola virus (EBOV) and Marburg virus (MARV). A substantial portion of filovirus genomes encode 5' and 3' untranslated regions (UTRs) of viral mRNAs. Select viral genomic RNA sequences corresponding to 3' UTRs are prone to editing by adenosine deaminase acting on RNA 1 (ADAR1). A reporter mRNA approach, in which different 5' or 3' UTRs were inserted into luciferase-encoding mRNAs, demonstrates that MARV 3' UTRs yield different levels of reporter gene expression, suggesting modulation of translation. The modulation occurs in cells unable to produce microRNAs (miRNAs) and can be recapitulated in a MARV minigenome assay. Deletion mutants identified negative regulatory regions at the ends of the MARV nucleoprotein (NP) and large protein (L) 3' UTRs. Apparent ADAR1 editing mutants were previously identified within the MARV NP 3' UTR. Introduction of these changes into the MARV nucleoprotein (NP) 3' UTR or deletion of the region targeted for editing enhances translation, as indicated by reporter assays and polysome analysis. In addition, the parental NP 3' UTR, but not the edited or deletion mutant NP 3' UTRs, induces a type I interferon (IFN) response upon transfection into cells. Because some EBOV isolates from the West Africa outbreak exhibited ADAR1 editing of the viral protein of 40 kDa (VP40) 3' UTR, VP40 3' UTRs with parental and edited sequences were similarly assayed. The EBOV VP40 3' UTR edits also enhanced translation, but neither the wild-type nor the edited 3' UTRs induced IFN. These findings implicate filoviral mRNA 3' UTRs as negative regulators of translation that can be inactivated by innate immune responses that induce ADAR1. IMPORTANCE UTRs comprise a large percentage of filovirus genomes and are apparent targets of editing by ADAR1, an enzyme with pro- and antiviral activities. However, the functional significance of the UTRs and ADAR1 editing has been uncertain. This study demonstrates that MARV and EBOV 3' UTRs can modulate translation, in some cases negatively. ADAR1 editing or deletion of select regions within the translation suppressing 3' UTRs relieves the negative effects of the UTRs. These data indicate that filovirus 3' UTRs contain translation regulatory elements that are modulated by activation of ADAR1, suggesting a complex interplay between filovirus gene expression and innate immunity.


Asunto(s)
Regiones no Traducidas 3' , Adenosina Desaminasa/metabolismo , Ebolavirus/genética , Marburgvirus/genética , Biosíntesis de Proteínas , Proteínas de Unión al ARN/metabolismo , Animales , Línea Celular , Ebolavirus/metabolismo , Genes Reporteros , Humanos , Interferón Tipo I/biosíntesis , Marburgvirus/metabolismo , MicroARNs/genética , Mutación , Proteínas de la Nucleocápside/genética , Proteínas de la Nucleocápside/metabolismo , Polirribosomas/metabolismo , Edición de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...