Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27.287
Filtrar
1.
Methods Mol Biol ; 2854: 19-28, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39192114

RESUMEN

The classic dual luciferase reporter assay has been widely used to rapidly and accurately determine the transcriptional activity of a given promoter induced by certain signal pathways in the cells. In particular, the sensitive characteristics of luciferase highlight its significance in many experiments, such as weak promoter analysis, transfection studies using small amounts of DNA, and detection in cell lines with low transfection efficiency. This chapter presents detailed information and experimental procedures for measuring interferon (IFN)-induced Interferon-Stimulated Response Element (ISRE) promoter activity using the dual luciferase reporter assay.


Asunto(s)
Genes Reporteros , Interferones , Luciferasas , Regiones Promotoras Genéticas , Elementos de Respuesta , Transducción de Señal , Humanos , Interferones/metabolismo , Interferones/genética , Luciferasas/metabolismo , Luciferasas/genética , Transfección , Animales
2.
Methods Mol Biol ; 2854: 127-141, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39192125

RESUMEN

Luciferase reporter systems are commonly used in scientific research to investigate a variety of biological processes, including antiviral innate immunity. These systems employ the use of luciferase enzymes derived from organisms such as fireflies or renilla reniformis, which emit light upon reaction with a substrate. In the context of antiviral innate immunity, the luciferase reporter systems offer a noninvasive and highly sensitive approach for real-time monitoring of immune responses in vitro and in vivo, enabling researchers to delve into the intricate interactions and signaling pathways involved in host-virus dynamic interactions. Here, we describe the methods of the promoter-luciferase reporter and enhancer-luciferase reporter, which provide insights into the transcriptional and post-transcriptional regulation of antiviral innate immunity. Additionally, we outline the split-luciferase complementary reporter method, which was designed to explore protein-protein interactions associated with antiviral immunity. These methodologies offer invaluable knowledge regarding the molecular mechanisms underlying antiviral immune pathways and have the potential to support the development of effective antiviral therapies.


Asunto(s)
Genes Reporteros , Inmunidad Innata , Luciferasas , Humanos , Luciferasas/metabolismo , Luciferasas/genética , Animales , Interferones/metabolismo , Interferones/inmunología , Regiones Promotoras Genéticas , Antivirales/farmacología , Células HEK293 , Interacciones Huésped-Patógeno/inmunología , Interacciones Huésped-Patógeno/genética
3.
Nat Commun ; 15(1): 8394, 2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39333139

RESUMEN

Host factors that define the cellular tropism of SARS-CoV-2 beyond the cognate ACE2 receptor are poorly defined. Here we report that SARS-CoV-2 replication is restricted at a post-entry step in a number of ACE2-positive airway-derived cell lines due to tonic activation of the cGAS-STING pathway mediated by mitochondrial DNA leakage and naturally occurring cGAS and STING variants. Genetic and pharmacological inhibition of the cGAS-STING and type I/III IFN pathways as well as ACE2 overexpression overcome these blocks. SARS-CoV-2 replication in STING knockout cell lines and primary airway cultures induces ISG expression but only in uninfected bystander cells, demonstrating efficient antagonism of the type I/III IFN-pathway in productively infected cells. Pharmacological inhibition of STING in primary airway cells enhances SARS-CoV-2 replication and reduces virus-induced innate immune activation. Together, our study highlights that tonic activation of the cGAS-STING and IFN pathways can impact SARS-CoV-2 cellular tropism in a manner dependent on ACE2 expression levels.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Proteínas de la Membrana , Nucleotidiltransferasas , SARS-CoV-2 , Transducción de Señal , Replicación Viral , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Humanos , SARS-CoV-2/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , COVID-19/inmunología , COVID-19/virología , COVID-19/metabolismo , Línea Celular , Interferones/metabolismo , Inmunidad Innata , Animales , Interferón Tipo I/metabolismo
4.
Biomed Pharmacother ; 179: 117426, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39243429

RESUMEN

Interferons are a family of cytokines that are famously known for their involvement in innate and adaptive immunity. Type I interferons (IFNs) exert pleiotropic effects on various immune cells and contribute to tumor-intrinsic and extrinsic mechanisms. Their pleiotropic effects and ubiquitous expression on nucleated cells have made them attractive candidates for cytokine engineering to deliver to largely immunosuppressive tumors. Type III interferons were believed to play overlapping roles with type I IFNs because they share a similar signaling pathway and induce similar transcriptional programs. However, type III IFNs are unique in their cell specific receptor expression and their antitumor activity is specific to a narrow range of cell types. Thus, type III IFN based therapies may show reduced toxic side effects compared with type I IFN based treatment. In this review, we focus on the development of IFN-based therapeutics used to treat different tumors. We highlight how the development in cytokine engineering has allowed for efficient delivery of type I and type III IFNs to tumor sites and look ahead to the obstacles that are still associated with IFN-based therapies before they can be fully and safely integrated into clinical settings.


Asunto(s)
Inmunoterapia , Interferones , Neoplasias , Humanos , Neoplasias/terapia , Neoplasias/inmunología , Inmunoterapia/métodos , Animales , Interferones/inmunología , Ingeniería de Proteínas/métodos , Interferón Tipo I/inmunología
5.
Sci Adv ; 10(39): eado4618, 2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39331709

RESUMEN

Patients with small-cell lung cancer (SCLC) have poor prognosis and typically experience only transient benefits from combined immune checkpoint blockade (ICB) and chemotherapy. Here, we show that inhibition of ataxia telangiectasia and rad3 related (ATR), the primary replication stress response activator, induces DNA damage-mediated micronuclei formation in SCLC models. ATR inhibition in SCLC activates the stimulator of interferon genes (STING)-mediated interferon signaling, recruits T cells, and augments the antitumor immune response of programmed death-ligand 1 (PD-L1) blockade in mouse models. We demonstrate that combined ATR and PD-L1 inhibition causes improved antitumor response than PD-L1 alone as the second-line treatment in SCLC. This study shows that targeting ATR up-regulates major histocompatibility class I expression in preclinical models and SCLC clinical samples collected from a first-in-class clinical trial of ATR inhibitor, berzosertib, with topotecan in patients with relapsed SCLC. Targeting ATR represents a transformative vulnerability of SCLC and is a complementary strategy to induce STING-interferon signaling-mediated immunogenicity in SCLC.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada , Neoplasias Pulmonares , Proteínas de la Membrana , Nucleotidiltransferasas , Transducción de Señal , Carcinoma Pulmonar de Células Pequeñas , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Animales , Humanos , Nucleotidiltransferasas/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/inmunología , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología , Ratones , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Transducción de Señal/efectos de los fármacos , Línea Celular Tumoral , Interferones/metabolismo , Antígeno B7-H1/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Topotecan/farmacología , Pirazinas/farmacología , Pirazinas/uso terapéutico , Isoxazoles
6.
Cell Rep ; 43(9): 114719, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39255062

RESUMEN

Neuroinflammation and dysregulated energy metabolism are linked to motor neuron degeneration in amyotrophic lateral sclerosis (ALS). The egl-9 family hypoxia-inducible factor (EGLN) enzymes, also known as prolyl hydroxylase domain (PHD) enzymes, are metabolic sensors regulating cellular inflammation and metabolism. Using an oligonucleotide-based and a genetic approach, we showed that the downregulation of Egln2 protected motor neurons and mitigated the ALS phenotype in two zebrafish models and a mouse model of ALS. Single-nucleus RNA sequencing of the murine spinal cord revealed that the loss of EGLN2 induced an astrocyte-specific downregulation of interferon-stimulated genes, mediated via the stimulator of interferon genes (STING) protein. In addition, we found that the genetic deletion of EGLN2 restored this interferon response in patient induced pluripotent stem cell (iPSC)-derived astrocytes, confirming the link between EGLN2 and astrocytic interferon signaling. In conclusion, we identified EGLN2 as a motor neuron protective target normalizing the astrocytic interferon-dependent inflammatory axis in vivo, as well as in patient-derived cells.


Asunto(s)
Esclerosis Amiotrófica Lateral , Astrocitos , Neuronas Motoras , Pez Cebra , Animales , Astrocitos/metabolismo , Neuronas Motoras/metabolismo , Pez Cebra/metabolismo , Ratones , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Humanos , Interferones/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Modelos Animales de Enfermedad , Células Madre Pluripotentes Inducidas/metabolismo
7.
Viruses ; 16(9)2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39339961

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) remains a significant concern for patients with chronic hepatitis C (HCV), even after achieving a sustained virological response (SVR) with direct-acting antivirals (DAAs) or interferon (IFN)-based therapies. This study compared the risk of HCC in patients with HCV who achieved SVR through the DAA versus IFN regimens. METHODS: A retrospective analysis was conducted on 4806 HCV patients, without coinfection nor prior HCC history, treated at the Chang Gung Memorial Hospital, Taiwan (DAA: 2825, IFN: 1981). Kaplan-Meier and Cox regression analyses with propensity score matching (PSM) were used to adjust for baseline differences. RESULTS: DAA-treated patients exhibited a higher incidence of HCC than IFN-treated patients before and after PSM (after PSM: annual: 1% vs. 0.5%; 6-year: 6% vs. 3%, p = 0.01). Both DAA and IFN patients had a decreased HCC incidence during follow-up (>3 vs. <3 years from the end of treatment: DAA: 1.43% vs. 1.00% per year; IFN: 0.47% vs. 0.36% per year, both p < 0.05). HCC incidence was higher in the first three years post-SVR in DAA-treated ACLD patients and then decreased (3.26% vs. 1.39% per year, p < 0.01). In contrast, HCC incidence remained constant in the non-ACLD and IFN-treated groups. Multivariate Cox regression identified age ≥ 60, male sex, BMI, AFP ≥ 6 ng/mL, FIB-4, and ACLD status as independent risk factors for HCC, but antiviral regimens were not an independent factor for HCC. CONCLUSION: DAA treatment significantly affects HCC risk primarily within three years post-treatment, especially in younger HCV patients with ACLD. HCC incidence was reduced after three years in ACLD patients treated by DAA, but continued surveillance was still necessary. However, patients under 60 without advanced liver disease may require less intensive follow-up.


Asunto(s)
Antivirales , Carcinoma Hepatocelular , Hepatitis C Crónica , Interferones , Neoplasias Hepáticas , Respuesta Virológica Sostenida , Humanos , Carcinoma Hepatocelular/epidemiología , Carcinoma Hepatocelular/virología , Carcinoma Hepatocelular/etiología , Masculino , Femenino , Antivirales/uso terapéutico , Persona de Mediana Edad , Neoplasias Hepáticas/epidemiología , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/virología , Incidencia , Estudios Retrospectivos , Factores de Riesgo , Hepatitis C Crónica/tratamiento farmacológico , Hepatitis C Crónica/complicaciones , Interferones/uso terapéutico , Taiwán/epidemiología , Anciano , Adulto , Hepacivirus/efectos de los fármacos
8.
Cytokine ; 183: 156751, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39244831

RESUMEN

Orthopoxviruses, a group of zoonotic viral infections, have emerged as a significant health emergency and global concern, particularly exemplified by the re-emergence of monkeypox (Mpox). Effectively addressing these viral infections necessitates a comprehensive understanding of the intricate interplay between the viruses and the host's immune response. In this review, we aim to elucidate the multifaceted aspects of innate immunity in the context of orthopoxviruses, with a specific focus on monkeypox virus (MPXV). We provide an in-depth analysis of the roles of key innate immune cells, including natural killer (NK) cells, dendritic cells (DCs), and granulocytes, in the host defense against MPXV. Furthermore, we explore the interferon (IFN) response, highlighting the involvement of toll-like receptors (TLRs) and cytosolic DNA/RNA sensors in detecting and responding to the viral presence. This review also examines the complement system's contribution to the immune response and provides a detailed analysis of the immune evasion strategies employed by MPXV to evade host defenses. Additionally, we discuss current prevention and treatment strategies for Mpox, including pre-exposure (PrEP) and post-exposure (PoEP) prophylaxis, supportive treatments, antivirals, and vaccinia immune globulin (VIG).


Asunto(s)
Células Dendríticas , Evasión Inmune , Inmunidad Innata , Monkeypox virus , Mpox , Inmunidad Innata/inmunología , Humanos , Animales , Células Dendríticas/inmunología , Evasión Inmune/inmunología , Mpox/inmunología , Monkeypox virus/inmunología , Células Asesinas Naturales/inmunología , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , Interferones/inmunología , Interferones/metabolismo , Granulocitos/inmunología
9.
Nat Commun ; 15(1): 8224, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39300098

RESUMEN

Severe febrile illnesses in children encompass life-threatening organ dysfunction caused by diverse pathogens and other severe inflammatory syndromes. A comparative approach to these illnesses may identify shared and distinct features of host immune dysfunction amenable to immunomodulation. Here, using immunophenotyping with mass cytometry and cell stimulation experiments, we illustrate trajectories of immune dysfunction in 74 children with multi-system inflammatory syndrome in children (MIS-C) associated with SARS-CoV-2, 30 with bacterial infection, 16 with viral infection, 8 with Kawasaki disease, and 42 controls. We explore these findings in a secondary cohort of 500 children with these illnesses and 134 controls. We show that neutrophil activation and apoptosis are prominent in multi-system inflammatory syndrome, and that this is partially shared with bacterial infection. We show that memory T cells from patients with multi-system inflammatory syndrome and bacterial infection are exhausted. In contrast, we show viral infection to be characterized by a distinct signature of decreased interferon signaling and lower interferon receptor gene expression. Improved understanding of immune dysfunction may improve approaches to immunomodulator therapy in severe febrile illnesses in children.


Asunto(s)
COVID-19 , Neutrófilos , Síndrome de Respuesta Inflamatoria Sistémica , Humanos , Niño , Neutrófilos/inmunología , COVID-19/inmunología , COVID-19/virología , COVID-19/complicaciones , Masculino , Femenino , Preescolar , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Fiebre/inmunología , SARS-CoV-2/inmunología , Lactante , Interferones/metabolismo , Infecciones Bacterianas/inmunología , Linfocitos T/inmunología , Síndrome Mucocutáneo Linfonodular/inmunología , Adolescente , Apoptosis , Activación Neutrófila
11.
Commun Biol ; 7(1): 1162, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39289468

RESUMEN

Toll-like receptor 7 (Tlr7) deficiency-accelerated severe COVID-19 is associated with reduced production of interferons (IFNs). However, the underlying mechanisms remain elusive. To address these questions, we utilize Tlr7 and Irf7 deficiency mice, single-cell RNA analysis together with bone marrow transplantation approaches. We demonstrate that at the early phase of infection, SARS-CoV-2 causes the upregulation of Tlr7, Irf7, and IFN pathways in the lungs of the infected mice. The deficiency of Tlr7 and Irf7 globally and/or in immune cells in mice increases the severity of COVID-19 via impaired IFN activation in both immune and/or non-immune cells, leading to increased lung viral loads. These effects are associated with reduced IFN alpha and gamma levels in the circulation. The deficiency of Tlr7 tends to cause the reduced production and nuclear translocation of interferon regulatory factor 7 (IRF7) in the lungs of the infected mice, indicative of reduced IRF7 activation. Despite higher amounts of lung viral antigen, Tlr7 or Irf7 deficiency resulted in substantially reduced production of antibodies against SARS-CoV-2, thereby delaying the viral clearance. These results highlight the importance of the activation of TLR7 and IRF7 leading to IFN production on the development of innate and adaptive immunity against COVID-19.


Asunto(s)
COVID-19 , Factor 7 Regulador del Interferón , Pulmón , Ratones Noqueados , SARS-CoV-2 , Receptor Toll-Like 7 , Animales , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , COVID-19/inmunología , COVID-19/virología , COVID-19/metabolismo , Ratones , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Pulmón/inmunología , Pulmón/virología , Pulmón/metabolismo , Interferones/metabolismo , Ratones Endogámicos C57BL , Índice de Severidad de la Enfermedad , Carga Viral , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Modelos Animales de Enfermedad
12.
Front Immunol ; 15: 1441908, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39224597

RESUMEN

Introduction: The antiviral activity of recombinant bovine interferon lambda 3 (bovIFN-λ3) against bovine viral diarrhea virus (BVDV) has been demonstrated in vitro in Madin-Darby bovine kidney cells (MDBK) and in vivo in cattle. However, anti-BVDV activity of bovIFN-λ3 has not been studied in bovine respiratory tract epithelial cells, supposedly a primary target of BVDV infection when entering the host by the oronasal route. Methods: Here we investigated the anti-BVDV activity of bovIFN-λ3 in bovine turbinate-derived primary epithelial cells (BTu) using BVDV infection and immunoperoxidase staining, TCID50, RT-qPCR, DNA and transcriptome sequencing, and transfection with plasmids containing the two subunits, IL-28Rα and IL-10Rß that constitute the bovIFN-λ3 receptor. Results: Our immunoperoxidase staining, RT-qPCR, and TCID50 results show that while BVDV was successfully cleared in MDBK cells treated with bovIFN-λ3 and bovIFN-α, only the latter, bovIFN-α, cleared BVDV in BTu cells. Preincubation of MDBK cells with bovIFN-λ3 before BVDV infection was needed to induce optimal antiviral state. Both cell types displayed intact type I and III IFN signaling pathways and expressed similar levels of IL-10Rß subunit of the type III IFN receptor. Sequencing of PCR amplicon of the IL-28Rα subunit revealed intact transmembrane domain and lack of single nucleotide polymorphisms (SNPs) in BTu cells. However, RT-qPCR and transcriptomic analyses showed a lower expression of IL-28Rα transcripts in BTu cells as compared to MDBK cells. Interestingly, transfection of BTu cells with a plasmid encoding IL-28Rα subunit, but not IL-10Rß subunit, established the bovIFN-λ3 sensitivity showing similar anti-BVDV activity to the response in MDBK cells. Conclusion: Our results demonstrate that the sensitivity of cells to bovIFN-λ3 depends not only on the quality but also of the quantity of the IL-28Rα subunit of the heterodimeric receptor. A reduction in IL-28Rα transcript expression was detected in BTu as compared to MDBK cells, despite the absence of spliced variants or SNPs. The establishment of bovIFN-λ3 induced anti-BVDV activity in BTu cells transfected with an IL-28Rα plasmid suggests that the level of expression of this receptor subunit is crucial for the specific antiviral activity of type III IFN in these cells.


Asunto(s)
Interferón lambda , Interferones , Cornetes Nasales , Animales , Bovinos , Interferones/metabolismo , Interferones/inmunología , Cornetes Nasales/virología , Cornetes Nasales/inmunología , Cornetes Nasales/metabolismo , Antivirales/farmacología , Virus de la Diarrea Viral Bovina/inmunología , Virus de la Diarrea Viral Bovina/fisiología , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Células Epiteliales/virología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Interleucinas/genética , Interleucinas/farmacología , Interleucinas/inmunología , Interleucinas/metabolismo , Línea Celular , Diarrea Mucosa Bovina Viral/inmunología , Diarrea Mucosa Bovina Viral/virología , Proteínas Recombinantes/farmacología , Subunidad beta del Receptor de Interleucina-10/genética , Subunidad beta del Receptor de Interleucina-10/metabolismo , Receptores de Citocinas
13.
Front Immunol ; 15: 1402951, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39286258

RESUMEN

Background: OASL (Oligoadenylate Synthetase-Like), an interferon-induced protein in the OAS family, plays a significant role in anti-viral response. Studies have demonstrated its association with prognosis of certain tumors. However, the mechanism through which OASL affects tumors is unclear. A systemic pan-cancer study of OASL needs to be illustrated. Methods: Analysis of OASL expression across 33 tumors was conducted utilizing TCGA, GTEx and CPTAC databases. COX and Log-Rank regressions were employed to calculate the prognosis. We validated the impact of OASL on apoptosis, migration, and invasion in pancreatic cancer cell lines. Moreover, we employed seven algorithms in bulk data to investigate the association of OASL expression and immune cell infiltration within tumor immune microenvironment (TIME) and ultimately validated at single-cell transcriptome level. Results: We discovered elevated expression of OASL and its genetic heterogeneity in certain tumors, which link closely to prognosis. Validation experiments were conducted in PAAD and confirmed these findings. Additionally, OASL regulates immune checkpoint ligand such as programmed death ligand 1 (PD-L1), through IFN-γ/STAT1 and IL-6/JAK/STAT3 pathways in tumor cells. Meanwhile, OASL affects macrophages infiltration in TIME. By these mechanisms OASL could cause dysfunction of cytotoxic T lymphocytes (CTLs) in tumors. Discussion: Multi-omics analysis reveals OASL as a prognostic and immunological biomarker in pan-cancer.


Asunto(s)
Biomarcadores de Tumor , Microambiente Tumoral , Humanos , Microambiente Tumoral/inmunología , Biomarcadores de Tumor/genética , Pronóstico , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Neoplasias/inmunología , Neoplasias/genética , 2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/metabolismo , Interferones/metabolismo , Interferones/genética , Perfilación de la Expresión Génica , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Multiómica
14.
Parasitol Res ; 123(9): 319, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39251458

RESUMEN

Initial studies using bioinformatics analysis revealed DNA sequence similarities between Trypanosoma cruzi GenBank® M21331, coding for Antigen 36 (Ag 36), and tripartite motif (TRIM) genes. TRIM40 showed 9.7% identity to GenBank M21331, and four additional TRIM genes had identities greater than 5.0%. TRIM37 showed a continuous stretch of identity of 12 nucleotides, that is, at least 25% longer than any of the other TRIMs. When we extended our analysis on the relationships of GenBank M21331 to further innate immune genes, using the Needleman-Wunsch (NW) algorithm for alignment, identities to human IFN-α, IFN-ß, and IFN-γ genes of 13.6%, 12.6%, and 17.9%, respectively, were found. To determine the minimum number of genes coding for proteins closely related to Ag 36, a BLAST-p search was conducted with it versus the T. cruzi genome. The BLAST-p search revealed that T. cruzi GenBank M21331 had 14 gene sequences homologous to microtubule-associated protein (MAP) genes with 100% amino acid sequence identity. To verify the similarities in non-human genes, a study comparing TRIM21 region sequences among mammalian species to the comparable human TRIM21 region showed that related sequences were also present in 11 mammalian species. The MAP genes homologous to Ag 36 form a family of at least 14 genes which mimic human immune genes in the IFN and TRIM families. This mimicry is of gene sequences and not their protein products or epitopes. These results appear to be the first description of molecular mimicry of immune genes in humans by a protozoan parasite.


Asunto(s)
Trypanosoma cruzi , Trypanosoma cruzi/genética , Trypanosoma cruzi/inmunología , Humanos , Animales , Proteínas Protozoarias/genética , Interferones/genética , Biología Computacional , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Proteínas de Motivos Tripartitos/genética
15.
Leukemia ; 38(10): 2210-2224, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39117798

RESUMEN

Despite selective HDAC3 inhibition showing promise in a subset of lymphomas with CREBBP mutations, wild-type tumors generally exhibit resistance. Here, using unbiased genome-wide CRISPR screening, we identify GNAS knockout (KO) as a sensitizer of resistant lymphoma cells to HDAC3 inhibition. Mechanistically, GNAS KO-induced sensitization is independent of the canonical G-protein activities but unexpectedly mediated by viral mimicry-related interferon (IFN) responses, characterized by TBK1 and IRF3 activation, double-stranded RNA formation, and transposable element (TE) expression. GNAS KO additionally synergizes with HDAC3 inhibition to enhance CD8+ T cell-induced cytotoxicity. Moreover, we observe in human lymphoma patients that low GNAS expression is associated with high baseline TE expression and upregulated IFN signaling and shares common disrupted biological activities with GNAS KO in histone modification, mRNA processing, and transcriptional regulation. Collectively, our findings establish an unprecedented link between HDAC3 inhibition and viral mimicry in lymphoma. We suggest low GNAS expression as a potential biomarker that reflects viral mimicry priming for enhanced response to HDAC3 inhibition in the clinical treatment of lymphoma, especially the CREBBP wild-type cases.


Asunto(s)
Cromograninas , Subunidades alfa de la Proteína de Unión al GTP Gs , Histona Desacetilasas , Linfoma , Humanos , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Linfoma/genética , Linfoma/patología , Linfoma/virología , Linfoma/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Cromograninas/genética , Ratones , Interferones/metabolismo , Animales , Inhibidores de Histona Desacetilasas/farmacología , Línea Celular Tumoral , Técnicas de Inactivación de Genes , Transducción de Señal
16.
Ophthalmic Res ; 67(1): 470-477, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39097965

RESUMEN

INTRODUCTION: Systemic implications create a critical need for identification of dry eye patients with Sjögren syndrome (SS). Herein, we aimed to determine expressions of type I-III interferons (IFNs) in dry eye patients with or without underlying SS and their differential diagnosis. METHODS: A prospective, observational, case-control study was performed on 140 dry eye patients among which 78 patients were diagnosed with SS. Clinical evaluations included ELISA detections of serum type I IFN (IFN-α and IFN-ß, type II IFN (IFN-γ), and type III IFN (IFN-λ1/IL-29, IFN-λ2/IL-28, and IFN-λ3/IL-28B), as well as reporter cell assay for serum type I IFN activity. RESULTS: The serum levels of IFN-α and IFN-ß were notably higher in dry eye patients with SS than those without underlying SS (p < 0.0001). The functional assay for serum type I IFN activity showed the mean summed scores in dry eye patients with SS were remarkably increased compared to those without underlying SS (p < 0.0001). The serum levels of IFN-γ and IFN-λ1/IL-29 seemed higher in dry eye patients with SS than those without underlying SS (p < 0.0001). The serum levels of type I IFN (IFN-α combined with IFN-ß), type II IFN (IFN-γ level), and type III IFN (IFN-λ1/IL-29) used as a test to predict underlying SS among dry eye patients produced an area under the curve of 0.86, 0.73, and 0.94, respectively. CONCLUSION: Serum levels of type I-III IFNs, especially IFN-α, IFN-ß, and IFN-λ1/IL-29, may serve as a useful biomarker for identification of SS dry eye from non-SS dry eye.


Asunto(s)
Síndromes de Ojo Seco , Ensayo de Inmunoadsorción Enzimática , Síndrome de Sjögren , Humanos , Síndrome de Sjögren/sangre , Síndrome de Sjögren/diagnóstico , Síndrome de Sjögren/metabolismo , Femenino , Estudios Prospectivos , Masculino , Persona de Mediana Edad , Síndromes de Ojo Seco/diagnóstico , Síndromes de Ojo Seco/sangre , Síndromes de Ojo Seco/metabolismo , Estudios de Casos y Controles , Biomarcadores/sangre , Interferones/sangre , Adulto , Interferón Tipo I/sangre , Anciano , Curva ROC
17.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(8): 704-709, 2024 Aug.
Artículo en Chino | MEDLINE | ID: mdl-39215668

RESUMEN

Objective To explore the effects of Myxovirus resistance protein A (MxA) on the Janus kinase/Signal transducer and activator of transcription (JAK/STAT) pathway in HepG2 cells. Methods HepG2 cells were transfected with the pcDNA3.1-Flag-MxA construct, and subsequent localization and expression of the MxA protein were detected through immunofluorescence cytochemistry. The presence of MxA protein was further confirmed by using Western blot analysis. Following transfection with MxA small interfering RNA (si-MxA) and subsequent treatment with alpha interferon (IFN-α), real-time fluorescent quantitative PCR was employed to measure the mRNA levels of myxovirus resistance protein A (MxA), protein kinase R (PKR), and oligoadenylate synthase (OAS). Western blot analysis was used to detect the protein expression of MxA, PKR, OAS, signal transducer and activator of transcription 1 (STAT1), phosphorylated STAT1 (pSTAT1), STAT2, phosphorylated STAT2 (p-STAT2) and interferon regulatory factor 9 (IRF9). Additionally, pcDNA3.1-Flag-MxA and pISRE-TA-luc were co-transfected into HepG2 and HepG2.2.15 cells, respectively, to assess the activity of the interferon-stimulated response element (ISRE) by using a luciferase activity assay. Results MxA protein was expressed in both the cytoplasm and nucleus of HepG2 cells, with higher expression levels in the cytoplasm than in the nucleus. Knocking down MxA expression in HepG2 cells did not affect the expression of STAT1, p-STAT1, STAT2, p-STAT2, and IRF9 proteins induced by IFN-α, but significantly reduced the expression of antiviral proteins PKR and OAS. Overexpression of MxA in HepG2 cells enhanced ISRE activity and increased the expression of PKR and OAS proteins, but this effect was inhibited in HepG2.2.15 cells. Conclusion MxA induces the expression of antiviral proteins by enhancing the activity of the JAK/STAT signaling pathway ISRE.


Asunto(s)
2',5'-Oligoadenilato Sintetasa , Proteínas de Resistencia a Mixovirus , Factor de Transcripción STAT1 , eIF-2 Quinasa , Humanos , Células Hep G2 , Proteínas de Resistencia a Mixovirus/genética , Proteínas de Resistencia a Mixovirus/metabolismo , 2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/metabolismo , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Interferón-alfa/farmacología , Interferón-alfa/genética , Interferón-alfa/metabolismo , Elementos de Respuesta/genética , Transducción de Señal , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/metabolismo , Interferones/genética , Interferones/metabolismo , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Regulación de la Expresión Génica
18.
Antiviral Res ; 230: 105989, 2024 10.
Artículo en Inglés | MEDLINE | ID: mdl-39154753

RESUMEN

Recently it was discovered that extracellular 2'-3'cGAMP can activate the STING pathway in a cGAS-independent fashion by being transported across the cell membrane via the folate transporter, SLC19A1, the first identified extracellular antiporter of this critical signaling molecule in cancer cells. We hypothesized that this non-canonical activation of STING pathway would function to establish an antiviral state similar to that seen with the paracrine antiviral activities of interferon. Herein, we report that treatment of the monocytic cell line, THP-1 cells and SH-SY5Y neuronal cell line with exogenous 2'-3'cGAMP induces interferon production and establishes an antiviral state that limits herpes simplex virus-1 (HSV-1), a ubiquitous virus with high seropositivity in the human population. Using either pharmaceutical inhibition or genetic knockout of SLC19A1 blocks the 2'-3'cGAMP-induced inhibition of viral replication. Our data indicate SLC19A1 functions as a newly identified antiviral mediator for extracellular 2'-3'cGAMP. This work presents novel and important findings about an antiviral mechanism which information could aid in the development of better antiviral drugs in the future.


Asunto(s)
Herpesvirus Humano 1 , Proteína Portadora de Folato Reducido , Replicación Viral , Humanos , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 1/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Proteína Portadora de Folato Reducido/metabolismo , Proteína Portadora de Folato Reducido/genética , Antivirales/farmacología , Antivirales/metabolismo , Línea Celular , Herpes Simple/virología , Herpes Simple/metabolismo , Células THP-1 , Transporte Biológico , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Interferones/metabolismo , Transducción de Señal
19.
Virus Res ; 349: 199451, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39168375

RESUMEN

Recent studies indicate that treatment of chronic hepatitis D virus (HDV) with either pegylated interferon (IFN)λ or pegylated IFNα monotherapy leads to a dramatic decline in HDV RNA. Herein, we investigated the innate antiviral efficacy of IFNλ and IFNα in humanized mice that lack an adaptive immune response. Humanized mice were either co-infected with hepatitis B virus (HBV) and HDV simultaneously, or HDV infection was performed subsequent to HBV infection (i.e., superinfected). After steady viral replication was achieved, mice received either IFNλ (n = 6) or IFNα (n = 7) for 12 (or 13) weeks. Pretreatment median levels of serum HBV DNA (8.8 [IQR:0.2] log IU/ml), HDV RNA (9.8 [0.5] log IU/ml), HBsAg (4.0 [0.4] log IU/ml) and human albumin, hAlb (6.9 [0.1] log ng/mL) were similar between mice treated with IFNα or IFNλ and between those coinfected versus superinfected. Compared to mice treated with IFNλ, mice treated with IFNα had a significantly greater decline in HBV, HDV, and HBsAg levels. In conclusion, IFNα induces stronger inhibition of HBV and HDV than IFNλ in humanized mice that lack an adaptive immune response. Further studies are needed to assess the respective role of the combined innate-and adaptive-immune systems in the treatment of HBV and HDV with IFNα and IFNλ.


Asunto(s)
Antivirales , Modelos Animales de Enfermedad , Virus de la Hepatitis B , Virus de la Hepatitis Delta , Interferón-alfa , Animales , Ratones , Interferón-alfa/uso terapéutico , Interferón-alfa/farmacología , Antivirales/uso terapéutico , Antivirales/farmacología , Virus de la Hepatitis Delta/efectos de los fármacos , Humanos , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/inmunología , Replicación Viral/efectos de los fármacos , Hepatitis D Crónica/tratamiento farmacológico , Hepatitis D Crónica/virología , Coinfección/tratamiento farmacológico , Coinfección/virología , Interferones , Hepatitis B/tratamiento farmacológico , Hepatitis B/virología , Hepatitis B/inmunología , Hepatitis D/tratamiento farmacológico , Hepatitis D/virología , Hepatitis D/inmunología , ARN Viral/sangre , ADN Viral/sangre , Hepatitis B Crónica/tratamiento farmacológico , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/virología , Carga Viral/efectos de los fármacos
20.
Dev Comp Immunol ; 161: 105252, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39173725

RESUMEN

Signal Recognition Particle 54 kDa (SRP54) is a subunit of the signal recognition particle (SRP), a cytoplasmic ribonucleoprotein complex guiding the transportation of newly synthesized proteins from polyribosomes to endoplasmic reticulum. In mammals, it has been reported to regulate the RLR signaling pathway negatively by impairing the association between MAVS and MDA5/RIG-I. However, the role of SRP54 in teleost antiviral innate immune response remains obscure. In this study, the SRP54 homolog of black carp (bcSRP54) has been cloned, and its function in antiviral innate immunity has been elucidated. The CDS of bcSRP54 gene consists of 1515 nucleotides and encodes 504 amino acids. Immunofluorescence (IF) showed that bcSRP54 was mainly distributed in the cytoplasm. Overexpressed bcSRP54 significantly reduced bcMDA5-mediated transcription of interferon (IFN) promoter in reporter assay. Co-expression of bcSRP54 and bcMDA5 significantly suppressed bcMDA5-mediated IFN signaling and antiviral activity, while bcSRP54 knockdown increased the antiviral ability of host cells. In addition, the results of the immunofluorescence staining demonstrated the subcellular overlapping between bcSRP54 and bcMDA5, and the co-immunoprecipitation (co-IP) experiment identified their association. Furthermore, the over-expression of bcSRP54 did not influence the protein expression and ubiquitination modification level of bcMDA5, however, hindered the binding of bcMDA5 to bcMAVS. In summary, our results conclude that bcSRP54 targets bcMDA5 and inhibits the interaction between bcMDA5 and bcMAVS, thereby negatively regulating antiviral innate immunity, which provides insight into how teleost SRP54 regulates IFN signaling.


Asunto(s)
Carpas , Proteínas de Peces , Inmunidad Innata , Helicasa Inducida por Interferón IFIH1 , Transducción de Señal , Animales , Carpas/inmunología , Carpas/genética , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Proteínas de Peces/inmunología , Transducción de Señal/inmunología , Helicasa Inducida por Interferón IFIH1/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Interferones/metabolismo , Interferones/inmunología , Interferones/genética , Partícula de Reconocimiento de Señal/metabolismo , Partícula de Reconocimiento de Señal/inmunología , Partícula de Reconocimiento de Señal/genética , Humanos , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Ubiquitinación , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/veterinaria , Rhabdoviridae
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA