Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Viruses ; 16(8)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39205214

RESUMEN

Arteriviruses can establish persistent infections in animals such as equids, pigs, nonhuman primates, rodents, and possums. Some Arteriviruses can even cause overt and severe diseases such as Equine Arteritis in horses and Porcine Reproductive and Respiratory Syndrome in pigs, leading to huge economic losses. Arteriviruses have evolved viral proteins to antagonize the host cell's innate immune responses by inhibiting type I interferon (IFN) signaling, assisting viral evasion and persistent infection. So far, the role of the Arterivirus glycoprotein 5 (GP5) protein in IFN signaling inhibition remains unclear. Here, we investigated the inhibitory activity of 47 Arterivirus GP5 proteins derived from various hosts. We demonstrated that all GP5 proteins showed conserved activity for antagonizing TIR-domain-containing adapter proteins inducing interferon-ß (TRIF)-mediated IFN-ß signaling through TRIF degradation. In addition, Arterivirus GP5 proteins showed a conserved inhibitory activity against IFN-ß signaling, induced by either pig or human TRIF. Furthermore, certain Arterivirus GP5 proteins could inhibit the induction of IFN-stimulated genes. These findings highlight the role of Arterivirus GP5 proteins in supporting persistent infection.


Asunto(s)
Arterivirus , Interferón Tipo I , Transducción de Señal , Animales , Arterivirus/genética , Arterivirus/metabolismo , Humanos , Porcinos , Interferón Tipo I/metabolismo , Infecciones por Arterivirus/veterinaria , Infecciones por Arterivirus/virología , Infecciones por Arterivirus/inmunología , Interferón beta/metabolismo , Interferón beta/genética , Inmunidad Innata , Proteínas Virales/metabolismo , Proteínas Virales/genética , Células HEK293
2.
Front Immunol ; 14: 1190261, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37942320

RESUMEN

Glucocorticoids potently inhibit expression of many inflammatory mediators, and have been widely used to treat both acute and chronic inflammatory diseases for more than seventy years. However, they can have several unwanted effects, amongst which immunosuppression is one of the most common. Here we used microarrays and proteomic approaches to characterise the effect of dexamethasone (a synthetic glucocorticoid) on the responses of primary mouse macrophages to a potent pro-inflammatory agonist, lipopolysaccharide (LPS). Gene ontology analysis revealed that dexamethasone strongly impaired the lipopolysaccharide-induced antimicrobial response, which is thought to be driven by an autocrine feedback loop involving the type I interferon IFNß. Indeed, dexamethasone strongly and dose-dependently inhibited the expression of IFNß by LPS-activated macrophages. Unbiased proteomic data also revealed an inhibitory effect of dexamethasone on the IFNß-dependent program of gene expression, with strong down-regulation of several interferon-induced antimicrobial factors. Surprisingly, dexamethasone also inhibited the expression of several antimicrobial genes in response to direct stimulation of macrophages with IFNß. We tested a number of hypotheses based on previous publications, but found that no single mechanism could account for more than a small fraction of the broad suppressive impact of dexamethasone on macrophage type I interferon signaling, underlining the complexity of this pathway. Preliminary experiments indicated that dexamethasone exerted similar inhibitory effects on primary human monocyte-derived or alveolar macrophages.


Asunto(s)
Antiinfecciosos , Lipopolisacáridos , Ratones , Animales , Humanos , Lipopolisacáridos/farmacología , Interferón beta/farmacología , Proteómica , Macrófagos , Glucocorticoides/farmacología , Dexametasona/farmacología , Antiinfecciosos/farmacología
3.
Cell Immunol ; 393-394: 104782, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37931572

RESUMEN

Type I interferons (IFN), especially human IFN alpha (IFNα), have been utilized for antitumor therapy for decades. Human interferon beta (IFNß) is rarely used for cancer treatment, despite advantages over IFNα in biological activities such as tumor growth inhibition and dendritic cell (DC) activation. The utilization of pegylated human IFNß (PEG-IFNß), as monotherapy or in combination with immune checkpoint inhibitors (ICIs) was evaluated in this study through in vivo efficacy studies in syngeneic mouse melanoma, non-small cell lung cancer (NSCLC), and colon adenocarcinoma (COAD) models resistant to immune checkpoint inhibitors (ICIs). In vitro comparative study of PEG-IFNß and pegylated IFNα-2b was performed in terms of tumor growth inhibition against human melanoma, NSCLC and COAD cell lines and activation of human monocyte-derived DCs (MoDCs). Our data demonstrate that the in vivo antitumor effects of PEG-IFNß are partially attributable to tumor growth-inhibitory effects and DC-activating activities, superior to pegylated IFNα-2b. Our findings suggest that utilizing PEG-IFNß as an antitumor therapy can enhance the therapeutic effect of ICIs in ICI-resistant tumors by directly inhibiting tumor growth and induction of DC maturation.


Asunto(s)
Adenocarcinoma , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias del Colon , Neoplasias Pulmonares , Melanoma , Animales , Ratones , Humanos , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Adenocarcinoma/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias del Colon/tratamiento farmacológico , Interferón-alfa/farmacología , Interferón alfa-2/uso terapéutico , Melanoma/tratamiento farmacológico , Interferón beta/metabolismo , Interferón beta/uso terapéutico , Polietilenglicoles/uso terapéutico , Células Dendríticas/metabolismo
4.
Cell Rep ; 42(8): 112852, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37481718

RESUMEN

The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is a major mediator of inflammation following stimulation with >45 bp double-stranded DNA (dsDNA). Herein, we identify a class of ∼20-40 bp small cytosolic dsDNA (scDNA) molecules that compete with long dsDNA (200-1,500 bp herring testis [HT]-DNA) for binding to cGAS, thus repressing HT-DNA-induced cGAS activation. The scDNA promotes cGAS and Beclin-1 interaction, releasing Rubicon, a negative regulator of phosphatidylinositol 3-kinase class III (PI3KC3), from the Beclin-1-PI3KC3 complex. This leads to PI3KC3 activation and induces autophagy, causing degradation of STING and long cytosolic dsDNA. Moreover, DNA damage decreases, and autophagy inducers increase scDNA levels. scDNA transfection and treatment with autophagy inducers attenuate DNA damage-induced cGAS activation. Thus, scDNA molecules serve as effective brakes for cGAS activation, preventing excessive inflammatory cytokine production following DNA damage. Our findings may have therapeutic implications for cytosolic DNA-associated inflammatory diseases.


Asunto(s)
ADN , Proteínas de la Membrana , Masculino , Humanos , Beclina-1 , Proteínas de la Membrana/metabolismo , ADN/metabolismo , Nucleotidiltransferasas/metabolismo , Fosfatidilinositol 3-Quinasa , Autofagia
5.
Front Immunol ; 14: 1161849, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37334371

RESUMEN

Interferon-beta (IFN-ß) for Multiple Sclerosis (MS) is turning 30. The COVID-19 pandemic rejuvenated the interest in interferon biology in health and disease, opening translational opportunities beyond neuroinflammation. The antiviral properties of this molecule are in accord with the hypothesis of a viral etiology of MS, for which a credible culprit has been identified in the Epstein-Barr Virus. Likely, IFNs are crucial in the acute phase of SARS-CoV-2 infection, as demonstrated by inherited and acquired impairments of the interferon response that predispose to a severe COVID-19 course. Accordingly, IFN-ß exerted protection against SARS-CoV-2 in people with MS (pwMS). In this viewpoint, we summarize the evidence on IFN-ß mechanisms of action in MS with a focus on its antiviral properties, especially against EBV. We synopsize the role of IFNs in COVID-19 and the opportunities and challenges of IFN-ß usage for this condition. Finally, we leverage the lessons learned in the pandemic to suggest a role of IFN-ß in long-COVID-19 and in special MS subpopulations.


Asunto(s)
COVID-19 , Infecciones por Virus de Epstein-Barr , Esclerosis Múltiple , Humanos , Interferón beta/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Infecciones por Virus de Epstein-Barr/complicaciones , SARS-CoV-2 , Pandemias , Síndrome Post Agudo de COVID-19 , Herpesvirus Humano 4 , Interferones/uso terapéutico , Interferones/farmacología , Antivirales/uso terapéutico , Antivirales/farmacología
6.
Med ; 3(9): 612-621.e3, 2022 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-35853458

RESUMEN

BACKGROUND: Black/African American participants are underrepresented in clinical trials for multiple sclerosis but can experience a greater burden of disease than other racial groups in the United States. A phase 1, open-label, crossover study that demonstrated bioequivalence of subcutaneous and intramuscular injection of peginterferon ß-1a in healthy volunteers enrolled similar proportions of Black and White participants, enabling a post hoc subgroup analysis comparing these groups. METHODS: Peginterferon ß-1a (125 µg) was administered by subcutaneous or intramuscular injection, followed by a washout period before a second injection using the alternative method. The primary pharmacokinetic and pharmacodynamic endpoints were maximum observed concentration (Cmax) and area under the concentration-time curve from hour 0 to infinity (AUCinf) of study drug and serum concentration of neop-terin, respectively. Safety and tolerability were included as secondary endpoints. FINDINGS: This analysis included 70 (51.5%) Black and 59 (43.3%) White participants. Peginterferon ß-1a Cmax was 29.8% higher in Black than in White participants following subcutaneous administration but was similar following intramuscular administration. Mean AUCinf was 31.0% and 11.8% greater in Black than in White participants with subcutaneous and intramuscular administration, respectively. Pharmacodynamics and safety signals were similar between groups, although Black participants reported numerically fewer adverse events. CONCLUSIONS: No clinically meaningful differences were identified between Black and White participants related to peginterferon ß-1a administration, supporting the approved dose of 125 µg/mL peginterferon ß-1a. Future clinical studies should include sufficiently diverse populations to ensure accurate assessments of treatment response. FUNDING: Funding for medical writing support was provided by Biogen (Cambridge, MA, USA).


Asunto(s)
Interferón beta , Polietilenglicoles , Estudios Cruzados , Humanos , Polietilenglicoles/efectos adversos
7.
Acta Diabetol ; 59(10): 1275-1286, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35829914

RESUMEN

AIMS: Pancreatic ß-cell apoptosis may be involved in the onset and progression of type 2 diabetes mellitus, although its mechanism remains unclear. We previously demonstrated that macrophage-derived interferon (IFN) ß induced X-linked inhibitor of apoptosis-associated factor 1 (XAF1) expression in ß-cells and accelerated ß-cell apoptosis in vitro. Here, we explored the effects of XAF1 on ß-cell function and progression of diabetes in vivo. METHODS: Pancreatic ß-cell-selective XAF1 overexpressing (Xaf1 Tg) mice were generated. Xaf1 Tg mice and their wild-type (WT) littermates were fed either a normal diet or a 40% or 60% high-fat diet (HFD). The effects of ß-cell XAF1 on ß-cell apoptosis and exacerbation of diabetes were investigated. RESULTS: Palmitic acid induced IFNß expression in macrophages, and HFD intake promoted macrophage infiltration in pancreatic islets, both of which cooperatively upregulated XAF1 expression in mouse islets. Furthermore, HFD-fed Xaf1 Tg mice demonstrated increased ß-cell apoptosis, lowered insulin expression, and impaired glucose tolerance compared with WT mice fed the same diet. These effects were more pronounced in the 60%HFD group than in the 40%HFD group. CONCLUSIONS: Pancreatic ß-cell XAF1 expression was enhanced via HFD-induced, macrophage-derived IFNß, which promoted ß-cell apoptosis and led to a reduction in insulin secretion and progression of diabetes. To our knowledge, this is the first report to demonstrate an association between pancreatic ß-cell XAF1 overexpression and exacerbation of diabetes, thus providing insight into the mechanism of ß-cell mass reduction in diabetes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Reguladoras de la Apoptosis , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Islotes Pancreáticos , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL
8.
Am J Neurodegener Dis ; 11(1): 10-16, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35600511

RESUMEN

BACKGROUND: Multiple Sclerosis (MS) is an autoimmune, inflammatory disease of the central nervous system. Magnetic resonance imaging (MRI) findings are associated with disease clinical activity and response to treatment. This study aimed to evaluate the future value of plaque number and volume in MRI as radiological criteria in determining the treatment response to INF-B in patients with MS. METHODS: This is a cross-sectional study performed in 2016-2021 in Iran on patients with the newly diagnosed (less than one year) relapsing-remitting MS. Brain MRI was taken for all patients. The number and volumes of the MS plaques were evaluated from FLAIR images by the two radiologists. Patients were treated with INF-B1a with a dosage of 12 million units equal to 44 micrograms subcutaneously, three times per week. Patients were visited monthly by neurologists to examine their clinical status. After one year, the brain MRI was conducted with the similar characteristics to the beginning of the study, and the number and volume of MS plaques were measured again. RESULTS: The study population consisted of 33 males and 90 females with a mean age of 28.37 ± 6.29 years. The mean Expanded Disability Status Scale (EDSS) of the patients was 3.16 ± 0.23 at the beginning of the study. The specificity for a 50% reduction in the number and volume of plaques as two separate criteria was the same and equal to 100%. The sensitivity of the number and volume of plaques were 65.5% and 90.6%, respectively. In addition, considering 10% as the cut-off point of the number of plaques, the sensitivity of the number of plaques as a criterion was equal to the sensitivity of the plaque volume. CONCLUSION: The results of this study showed that imaging criteria provide a more objective tool for evaluating the effectiveness of treatment. These findings indicate that the number and volume of plaques could be two reliable MRI imaging criteria for assessing therapy response. The number of plaques was less accurate than the volume of plaques.

9.
Mol Med Rep ; 25(6)2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35485288

RESUMEN

As the understanding of cancer grows, new therapies have been proposed to improve the well-known limitations of current therapies, whose efficiency relies mostly on early detection, surgery and chemotherapy. Mesenchymal stem cells (MSCs) have been introduced as a promissory and effective therapy. This fact is due to several useful features of MSCs, such as their accessibility and easy culture and expansion in vitro, and their remarkable ability for 'homing' towards tumors, allowing MSCs to exert their anticancer effects directly into tumors. Additionally, MSCs offer the practicability of being genetically engineered to carry anticancer genes, increasing their specificity and efficacy for fighting tumors. In the present study, the antitumoral efficacy and post-implant survival of mice bearing lymphomas implanted intratumorally were determined using mouse bone marrow-derived (BM)-MSCs transduced with soluble TRAIL (sTRAIL), full length TRAIL (flTRAIL), or interferon ß (IFNß), naïve BM-MSCs, or combinations of these. The percentage of surviving mice was determined once all not-implanted mice succumbed. It was found that the percentage of surviving mice implanted with the combination of MSCs-sTRAIL and MSCs-IFN-ß was 62.5%. Lymphoma model achieved 100% fatality in the non-treated group by day 41. On the other hand, the percentage of surviving mice implanted with MSCs-sTRAIL was 50% and with MSCs-INFß 25%. All the aforementioned differences were statistically significant (P<0.05). In conclusion, all implants exhibited tumor size reduction, growth delay, or apparent tumor clearance. MSCs proved to be effective anti-lymphoma agents; additionally, the combination of soluble TRAIL and IFN-ß resulted in the most effective antitumor and life enlarging treatment, showing an additive antitumoral effect compared with individual treatments.


Asunto(s)
Linfoma , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Animales , Hipertrofia , Interferón beta/genética , Linfoma/genética , Linfoma/terapia , Ratones
10.
BMC Biotechnol ; 22(1): 13, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35351081

RESUMEN

BACKGROUND: Type I interferons are widely used in research applications and as biotherapeutics. Current assays used to measure interferon concentrations, such as plaque reduction assays and ELISA, are expensive, technically challenging, and may take days to provide results. We sought to develop a robust and rapid assay to determine interferon concentrations produced from transiently transfected cell cultures. METHOD: Indirect quantification of recombinant interferon was evaluated using a novel bi-cistronic construct encoding the Foot-and-mouth disease virus 2A translational interrupter sequence to yield equimolar expression of Gaussia princeps luciferase and porcine interferon α. Direct quantification was evaluated by expression of a novel fusion protein comprised of Gaussia princeps luciferase and porcine type I interferon. Plasmids encoding constructs are transiently transfected into cell cultures and supernatant harvested for testing of luminescence, ELISA determined concentration, and anti-viral activity against vesicular stomatitis virus. RESULTS: Bi-cistronic constructs, utilized for indirect quantification, demonstrate both luciferase activity and anti-viral activity. Fusion proteins, utilized for direct quantification, retained secretion and luminescence however only the interferon α fusion protein had antiviral activity comparable to wildtype porcine interferon α. A strong linear correlation was observed between dilution and luminescence for all compounds over a dynamic range of concentrations. CONCLUSION: The correlation of antiviral and luciferase activities demonstrated the utility of this approach, both direct and indirect, to rapidly determine recombinant interferon concentrations. Concentration can be determined over a more dynamic concentration range than available ELISA based assays using this methodology.


Asunto(s)
Interferón Tipo I , Animales , Antivirales/farmacología , Interferón Tipo I/genética , Interferón-alfa/genética , Luciferasas/genética , Luciferasas/metabolismo , Luminiscencia , Porcinos
11.
J Extracell Vesicles ; 10(10): e12127, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34377374

RESUMEN

Infectious organisms and damage of cells can activate inflammasomes, which mediate tissue inflammation and adaptive immunity. These mechanisms evolved to curb the spread of microbes and to induce repair of the damaged tissue. Chronic activation of inflammasomes, however, contributes to non-resolving inflammatory responses that lead to immuno-pathologies. Inflammasome-activated cells undergo an inflammatory cell death associated with the release of potent pro-inflammatory cytokines and poorly characterized extracellular vesicles (EVs). Since inflammasome-induced EVs could signal inflammasome pathway activation in patients with chronic inflammation and modulate bystander cell activation, we performed a systems analysis of the ribonucleic acid (RNA) content and function of two EV classes. We show that EVs released from inflammasome-activated macrophages carry a specific RNA signature and contain interferon ß (IFNß). EV-associated IFNß induces an interferon signature in bystander cells and results in dampening of NLRP3 inflammasome responses. EVs could, therefore, serve as biomarkers for inflammasome activation and act to prevent systemic hyper-inflammatory states by restricting NLRP3 activation in bystander cells.


Asunto(s)
Vesículas Extracelulares/metabolismo , Inflamasomas/metabolismo , Interferón beta/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , ARN/análisis , Inmunidad Adaptativa , Animales , Biomarcadores/metabolismo , Línea Celular , Células Cultivadas , Vesículas Extracelulares/inmunología , Humanos , Inflamasomas/inmunología , Inflamación/metabolismo , Macrófagos/inmunología , Ratones , Piroptosis , ARN/metabolismo
12.
Front Microbiol ; 12: 702881, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34394047

RESUMEN

Type I interferons (IFN), a family of cytokines widely expressed in various tissues, play important roles in anti-infection immunity. Nevertheless, it is not known whether Brucella spp. could interfere with IFN-I production induced by other pathogens. This study investigated the regulatory roles of Brucella outer membrane protein (Omp)25 on the IFN-I signaling pathway and found that Omp25 inhibited the production of IFN-ß and its downstream IFN-stimulated genes induced by various DNA viruses or IFN-stimulatory DNA in human, murine, porcine, bovine, and ovine monocyte/macrophages or peripheral blood mononuclear cells. Brucella Omp25 suppressed the phosphorylation of stimulator of IFN genes (STINGs) and IFN regulatory factor 3 and nuclear translocation of phosphorylated IFN regulatory factor 3 in pseudorabies virus- or herpes simplex virus-1-infected murine, human, or porcine macrophages. Furthermore, we found that Brucella Omp25 promoted cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) degradation via the proteasome-dependent pathway, resulting in a decreased cyclic guanosine monophosphate-adenosine monophosphate production and downstream signaling activation upon DNA virus infection or IFN-stimulatory DNA stimulation. Mapping the predominant function domain of Omp25 showed that the amino acids 161 to 184 of Omp25 were required for Omp25-induced cGAS degradation, among which five amino acid residues (R176, Y179, R180, Y181, and Y184) were required for the inhibitory effect of Omp25 on IFN-ß induction. Altogether, our results demonstrated that Brucella Omp25 inhibits cGAS STING signaling pathway-induced IFN-ß via facilitating the ubiquitin-proteasome-dependent degradation of cGAS in various mammalian monocyte/macrophages.

14.
J Virol ; 95(20): e0023121, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34379499

RESUMEN

The NS1 protein of the influenza A virus plays a critical role in regulating several biological processes in cells, including the type I interferon (IFN) response. We previously profiled the cellular factors that interact with the NS1 protein of influenza A virus and found that the NS1 protein interacts with proteins involved in RNA splicing/processing, cell cycle regulation, and protein targeting processes, including 14-3-3ε. Since 14-3-3ε plays an important role in retinoic acid-inducible gene I (RIG-I) translocation to mitochondrial antiviral-signaling protein (MAVS) to activate type I IFN expression, the interaction of the NS1 and 14-3-3ε proteins may prevent the RIG-I-mediated IFN response. In this study, we confirmed that the 14-3-3ε protein interacts with the N-terminal domain of the NS1 protein and that the NS1 protein inhibits RIG-I-mediated IFN-ß promoter activation in 14-3-3ε-overexpressing cells. In addition, our results showed that knocking down 14-3-3ε can reduce IFN-ß expression elicited by influenza A virus and enhance viral replication. Furthermore, we found that threonine in the 49th amino acid position of the NS1 protein plays a role in the interaction with 14-3-3ε. Influenza A virus expressing C terminus-truncated NS1 with a T49A mutation dramatically increases IFN-ß mRNA in infected cells and causes slower replication than that of virus without the T-to-A mutation. Collectively, this study demonstrates that 14-3-3ε is involved in influenza A virus-initiated IFN-ß expression and that the interaction of the NS1 protein and 14-3-3ε may be one of the mechanisms for inhibiting type I IFN activation during influenza A virus infection. IMPORTANCE Influenza A virus is an important human pathogen causing severe respiratory disease. The virus has evolved several strategies to dysregulate the innate immune response and facilitate its replication. We demonstrate that the NS1 protein of influenza A virus interacts with the cellular chaperone protein 14-3-3ε, which plays a critical role in retinoic acid-inducible gene I (RIG-I) translocation that induces type I interferon (IFN) expression, and that NS1 protein prevents RIG-I translocation to the mitochondrial membrane. The interaction site for 14-3-3ε is the RNA-binding domain (RBD) of the NS1 protein. Therefore, this research elucidates a novel mechanism by which the NS1 RBD mediates IFN-ß suppression to facilitate influenza A viral replication. Additionally, the findings reveal the antiviral role of 14-3-3ε during influenza A virus infection.


Asunto(s)
Proteínas 14-3-3/inmunología , Gripe Humana/inmunología , Interferón beta/metabolismo , Proteínas 14-3-3/metabolismo , Línea Celular Tumoral , Proteína 58 DEAD Box/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata/inmunología , Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Interferón beta/fisiología , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional , ARN Viral/genética , Receptores Inmunológicos/metabolismo , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/genética
15.
Dev Comp Immunol ; 125: 104211, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34329648

RESUMEN

As a group of cytokines, interferons are the first line of defense in the antiviral immunity. In this study, Siberian tiger IFN-ß (PtIFN-ß) and IFN-γ (PtIFN-γ) were successfully amplified, and the two were fused (PtIFN-γ) by overlap extension polymerase chain reaction (SOE-PCR). Bioinformatics analysis disclosed that PtIFN-ß and PtIFN-γ have species-specificity and conservation in the course of evolution. After being expressed in prokaryotes, the antiviral activities and physicochemical properties of PtIFN-ß, PtIFN-γ and PtIFNß-γ were analyzed. In Feline kidney cells (F81), PtIFNß-γ showed more active antiviral activity than PtIFN-ß and PtIFN-γ, which has more stable physicochemical properties (acid and alkali resistance, high temperature resistance). In addition, PtIFN-ß, PtIFN-γ and PtIFN-γ activated the JAK-STAT pathway and induced the transcription and expression of interferon-stimulated genes (ISGs). Janus kinase (JAK) 1 inhibitor inhibited ISGs expression induced by PtIFN-ß, PtIFN-γ and PtIFN-γ. Overall, this research clarified that PtIFN-ß, PtIFN-γ and PtIFNß-γ have the ability to inhibit viral replication and send signals through the JAK-STAT pathway. These findings may facilitate further study on the role of PtIFN in the antiviral immune response, and help to develop approaches for the prophylactic and therapeutic of viral diseases based on fusion interferon.


Asunto(s)
Tigres/inmunología , Animales , Antivirales/farmacología , Gatos , China , Retroalimentación , Expresión Génica , Humanos , Inmunidad Innata , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Interferón gamma/genética , Transducción de Señal/inmunología , Virosis , Replicación Viral/inmunología
16.
Microb Pathog ; 158: 105105, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34311016

RESUMEN

COVID-19 exhibits a global health threat among the elderly and the population with underlying health conditions. During infection, the host's innate immune response acts as a frontline of defense by releasing cytokines such as type I interferon (IFN α and ß) thereby initiating antiviral activity. However, this particular interferon response is interrupted by factors such as SARS-CoV-2 non-structural proteins, aging, diabetes, and germ-line errors eventually making the host more susceptible to illness. Therefore, enhancing the host's innate immune response by administering type I IFN could be an effective treatment against COVID-19. Here, we highlight the importance of innate immune response and the role of IFN ß monotherapy against COVID-19.


Asunto(s)
COVID-19 , Interferón Tipo I , Anciano , Humanos , Inmunidad Innata , Interferón beta , SARS-CoV-2
17.
J Control Release ; 330: 1095-1105, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33188827

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant disease, where even surgical resection and aggressive chemotherapy produce dismal outcomes. Immunotherapy is a promising alternative to conventional treatments, possessing the ability to elicit T cell-mediated killing of tumor cells and prevent disease recurrence. Immunotherapeutic approaches thus far have seen limited success in PDAC due to a poorly immunogenic and exceedingly immunosuppressive tumor microenvironment, which is enriched with dysfunctional and immunosuppressed antigen-presenting cells (APCs). We developed a highly potent immunostimulatory nanoparticle (immuno-NP) to activate and expand APCs in the tumor and induce local secretion of interferon ß (IFNß), which is a pro-inflammatory cytokine that plays a major role in APC recruitment. The effectiveness of the immuno-NP stems from its dual cargo of two synergistic immune modulators consisting of an agonist of the stimulator of interferon genes (STING) pathway and an agonist of the Toll-like receptor 4 (TLR4) pathway. We show the functional synergy of the dual-agonist cargo can be tweaked by adjusting the ratio of the two agonists loaded in the immuno-NP, leading to an increase in IFNß production (11-fold) compared to any single agonist immuno-NP variant. Using the orthotopic murine Panc02 model of PDAC, we show that systemic administration allowed immuno-NPs to deposit into the perivascular regions of the tumor, which coincided with the APC-rich tumor areas leading to predominant uptake of immuno-NPs by APCs. The immuno-NPs were effectively taken up by a significant portion of dendritic cells in the tumor (>56%). This led to a significant expansion of APCs, resulting in an 11.5-fold increase of dendritic cells and infiltration of lymphocytes throughout the pancreatic tumor compared to untreated animals.


Asunto(s)
Nanopartículas , Neoplasias Pancreáticas , Animales , Células Presentadoras de Antígenos , Inmunización , Inmunoterapia , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Microambiente Tumoral
18.
Int J Mol Sci ; 23(1)2021 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-35008744

RESUMEN

Viral infections increase the risk of developing allergies in childhood, and disruption of mucosal homeostasis is presumed to be involved. However, no study has reported a role for viral infections in such disruption. In this study, we clarified the mechanism of immunoglobulin A (IgA) overproduction in viral infections. Autopsies were performed on 33 pediatric cases, IgA and interferon (IFN)ß levels were measured, and histopathological and immunohistochemical examinations were conducted. Furthermore, we cultured human cells and measured IFNß and IgA levels to examine the effect of viral infections on IgA production. Blood IgA levels in viral infections were higher than in bacterial infections. Moreover, IFNß levels in most viral cases were below the detection limit. Cell culture revealed increased IgA in gastrointestinal lymph nodes, especially in Peyer's patches, due to enhanced IFNß after viral stimulation. Conversely, respiratory regional lymph nodes showed enhanced IgA with no marked change in IFNß. Overproduction of IgA, identified as an aberration of the immune system and resulting from excessive viral infection-induced IFNß was observed in the intestinal regional lymph nodes, particularly in Peyer's patches. Further, increased IgA without elevated IFNß in the respiratory system suggested the possibility of a different mechanism from the gastrointestinal system.


Asunto(s)
Inmunidad , Intestinos/inmunología , Ganglios Linfáticos/inmunología , Virosis/inmunología , Autopsia , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Niño , Femenino , Humanos , Inmunoglobulina A/sangre , Lactante , Recién Nacido , Interferón beta/sangre , Intestinos/patología , Ganglios Linfáticos/patología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Masculino , Ganglios Linfáticos Agregados/efectos de los fármacos , Ganglios Linfáticos Agregados/inmunología , Poli I-C/farmacología , Valores de Referencia , Virosis/sangre
19.
Clin Pharmacol Drug Dev ; 10(3): 307-316, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33352008

RESUMEN

This double-blind, randomized, placebo-controlled, dose-ascending, first-in-human study (NCT02766621) assessed the safety, tolerability, and pharmacokinetics (PK) of PF-06823859, an anti-interferon ß monoclonal antibody. Healthy subjects were randomized to single ascending doses (SADs) of intravenous PF-06823859 30, 100, 300, 900, or 2000 mg or placebo; to multiple ascending doses (MADs) of subcutaneous PF-06823859 100 or 300 mg or placebo (once every 2 weeks for a total of 3 doses); or to MAD of intravenous PF-06823859 600 mg or placebo (once every 3 weeks or once every 4 weeks for a total of 2 doses). The incidence, severity, and causal relationship of adverse events (AEs) were assessed, along with immunogenicity and PK. In total, 62 subjects were randomized to treatment (SAD, n = 35; MAD, n = 27). There were 76 treatment-emergent all-causality AEs in the SAD (PF-06823859: n = 25; placebo: n = 4) and MAD (PF-06823859: n = 40; placebo: n = 7) cohorts. In the SAD cohorts, all treatment-emergent all-causality AEs were mild in severity; 4 AEs of moderate severity were identified in the MAD cohorts. No dose-limiting AEs, serious AEs, treatment-related discontinuations, dose reductions, or deaths occurred. PF-06823859 exposure increased dose-proportionally, with half-life values ranging between 23 and 35 days. The estimated subcutaneous bioavailability was 43% to 44%. Immunogenicity incidence rates were low (antidrug antibodies, 12.5%; neutralizing antibodies, 2.1%). No immunogenically related clinical responses of concern were observed. In conclusion, PF-06823859 demonstrated an acceptable safety, tolerability, and PK profile that supports clinical development for treating disorders associated with increased interferon ß levels, such as dermatomyositis or systemic lupus erythematosus.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Enfermedades Autoinmunes/tratamiento farmacológico , Inmunidad/efectos de los fármacos , Interferón beta/antagonistas & inhibidores , Administración Intravenosa , Adulto , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Neutralizantes/efectos de los fármacos , Enfermedades Autoinmunes/inmunología , Disponibilidad Biológica , Estudios de Casos y Controles , Método Doble Ciego , Tolerancia a Medicamentos , Femenino , Semivida , Voluntarios Sanos , Humanos , Inyecciones Subcutáneas , Interferón beta/sangre , Interferón beta/metabolismo , Masculino , Persona de Mediana Edad , Farmacocinética , Placebos/administración & dosificación , Seguridad
20.
Poult Sci ; 100(1): 9-18, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33357711

RESUMEN

Chicken MDA5 (chMDA5), the essential accepted pattern recognition receptors for detecting cytoplasmic viral RNA in chicken, initiates interferon ß (IFN-ß) generation. However, there is an incomplete elucidation of regulating chMDA5-mediated IFN-ß production. NEMO-related protein, optineurin, was identified as inhibitors of virus triggered IFN-ß induction in human or mice. In this study, full length of chicken optineurin (chOPTN) was cloned from chicken embryo fibroblast, and its role in inhibiting IFN-ß signaling pathway was further explored. Full-length chOPTN encodes 547 amino acids residues and contains unique LC3 interaction region and ubiquitin binding domain. Chicken optineurin mRNA and protein are widely expressed in different tissues, especially the heart, kidney, and bursal fabricius (BF). Overexpressed chOPTN not only inhibits poly I:C or homos-induced human IFN-ß promoter activation in 293T cells but also suppresses poly I:C, infectious bursal disease virus (IBDV) genome double-strand RNA (dsRNA), and chMDA5-induced chicken IFN-ß (chIFN-ß) promoter activation. In addition, we first revealed that chOPTN negatively regulates chIFN-ß production via inhibiting ubiquitination of chicken TBK1, which is dependent on the ubiquitin-binding domain of chOPTN. Moreover, chIFN-ß stimulus, poly I:C, and IBDV genome dsRNA improve chOPTN expression. Endogenous chOPTN expression is also upregulated by IBDV infection in 293T, DF-1 cells, as well as in BF. Therefore, our results suggested that chOPTN plays an inhibition role of chMDA5-mediated chIFN-ß signaling pathway in chicken cells.


Asunto(s)
Proteínas de Ciclo Celular , Pollos , Regulación de la Expresión Génica , Helicasa Inducida por Interferón IFIH1 , Animales , Infecciones por Birnaviridae/inmunología , Infecciones por Birnaviridae/veterinaria , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Embrión de Pollo , Pollos/genética , Pollos/inmunología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Virus de la Enfermedad Infecciosa de la Bolsa/fisiología , Helicasa Inducida por Interferón IFIH1/inmunología , Interferón beta/genética , ARN Bicatenario/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA