Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26.131
Filtrar
1.
Methods Mol Biol ; 2854: 19-28, 2025.
Artigo em Inglês | MEDLINE | ID: mdl-39192114

RESUMO

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.


Assuntos
Genes Reporter , Interferons , Luciferases , Regiões Promotoras Genéticas , Elementos de Resposta , Transdução de Sinais , Humanos , Interferons/metabolismo , Interferons/genética , Luciferases/metabolismo , Luciferases/genética , Transfecção , Animais
2.
Methods Mol Biol ; 2854: 127-141, 2025.
Artigo em Inglês | MEDLINE | ID: mdl-39192125

RESUMO

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.


Assuntos
Genes Reporter , Imunidade Inata , Luciferases , Humanos , Luciferases/metabolismo , Luciferases/genética , Animais , Interferons/metabolismo , Interferons/imunologia , Regiões Promotoras Genéticas , Antivirais/farmacologia , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/genética
3.
Cytokine ; 183: 156751, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39244831

RESUMO

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).


Assuntos
Células Dendríticas , Evasão da Resposta Imune , Imunidade Inata , Monkeypox virus , Mpox , Imunidade Inata/imunologia , Humanos , Animais , Células Dendríticas/imunologia , Evasão da Resposta Imune/imunologia , Mpox/imunologia , Monkeypox virus/imunologia , Células Matadoras Naturais/imunologia , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Interferons/imunologia , Interferons/metabolismo , Granulócitos/imunologia
4.
Nat Commun ; 15(1): 8224, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39300098

RESUMO

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.


Assuntos
COVID-19 , Neutrófilos , Síndrome de Resposta Inflamatória Sistêmica , Humanos , Criança , Neutrófilos/imunologia , COVID-19/imunologia , COVID-19/virologia , COVID-19/complicações , Masculino , Feminino , Pré-Escolar , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Febre/imunologia , SARS-CoV-2/imunologia , Lactente , Interferons/metabolismo , Infecções Bacterianas/imunologia , Linfócitos T/imunologia , Síndrome de Linfonodos Mucocutâneos/imunologia , Adolescente , Apoptose , Ativação de Neutrófilo
5.
Parasitol Res ; 123(9): 319, 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39251458

RESUMO

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.


Assuntos
Trypanosoma cruzi , Trypanosoma cruzi/genética , Trypanosoma cruzi/imunologia , Humanos , Animais , Proteínas de Protozoários/genética , Interferons/genética , Biologia Computacional , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Proteínas com Motivo Tripartido/genética
6.
Commun Biol ; 7(1): 1162, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39289468

RESUMO

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.


Assuntos
COVID-19 , Fator Regulador 7 de Interferon , Pulmão , Camundongos Knockout , SARS-CoV-2 , Receptor 7 Toll-Like , Animais , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/metabolismo , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , COVID-19/imunologia , COVID-19/virologia , COVID-19/metabolismo , Camundongos , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , Pulmão/imunologia , Pulmão/virologia , Pulmão/metabolismo , Interferons/metabolismo , Camundongos Endogâmicos C57BL , Índice de Gravidade de Doença , Carga Viral , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Modelos Animais de Doenças
7.
Front Immunol ; 15: 1402951, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39286258

RESUMO

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.


Assuntos
Biomarcadores Tumorais , Microambiente Tumoral , Humanos , Microambiente Tumoral/imunologia , Biomarcadores Tumorais/genética , Prognóstico , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Neoplasias/imunologia , Neoplasias/genética , 2',5'-Oligoadenilato Sintetase/genética , 2',5'-Oligoadenilato Sintetase/metabolismo , Interferons/metabolismo , Interferons/genética , Perfilação da Expressão Gênica , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Multiômica
9.
Front Immunol ; 15: 1441908, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39224597

RESUMO

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.


Assuntos
Interferon lambda , Interferons , Conchas Nasais , Animais , Bovinos , Interferons/metabolismo , Interferons/imunologia , Conchas Nasais/virologia , Conchas Nasais/imunologia , Conchas Nasais/metabolismo , Antivirais/farmacologia , Vírus da Diarreia Viral Bovina/imunologia , Vírus da Diarreia Viral Bovina/fisiologia , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Células Epiteliais/virologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Interleucinas/genética , Interleucinas/farmacologia , Interleucinas/imunologia , Interleucinas/metabolismo , Linhagem Celular , Doença das Mucosas por Vírus da Diarreia Viral Bovina/imunologia , Doença das Mucosas por Vírus da Diarreia Viral Bovina/virologia , Proteínas Recombinantes/farmacologia , Subunidade beta de Receptor de Interleucina-10/genética , Subunidade beta de Receptor de Interleucina-10/metabolismo , Receptores de Citocinas
10.
Cell Rep ; 43(8): 114581, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39102336

RESUMO

Bats harbor highly virulent viruses that can infect other mammals, including humans, posing questions about their immune tolerance mechanisms. Bat cells employ multiple strategies to limit virus replication and virus-induced immunopathology, but the coexistence of bats and fatal viruses remains poorly understood. Here, we investigate the antiviral RNA interference pathway in bat cells and discover that they have an enhanced antiviral RNAi response, producing canonical viral small interfering RNAs upon Sindbis virus infection that are missing in human cells. Disruption of Dicer function results in increased viral load for three different RNA viruses in bat cells, indicating an interferon-independent antiviral pathway. Furthermore, our findings reveal the simultaneous engagement of Dicer and pattern-recognition receptors, such as retinoic acid-inducible gene I, with double-stranded RNA, suggesting that Dicer attenuates the interferon response initiation in bat cells. These insights advance our comprehension of the distinctive strategies bats employ to coexist with viruses.


Assuntos
Quirópteros , Interferência de RNA , Ribonuclease III , Animais , Quirópteros/virologia , Quirópteros/imunologia , Humanos , Ribonuclease III/metabolismo , Ribonuclease III/genética , Sindbis virus/fisiologia , Linhagem Celular , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/genética , Replicação Viral , Interferons/metabolismo , RNA de Cadeia Dupla/metabolismo
11.
Nat Immunol ; 25(9): 1607-1622, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39138384

RESUMO

The evolution of T cell molecular signatures in the distal lung of patients with severe pneumonia is understudied. Here, we analyzed T cell subsets in longitudinal bronchoalveolar lavage fluid samples from 273 patients with severe pneumonia, including unvaccinated patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or with respiratory failure not linked to pneumonia. In patients with SARS-CoV-2 pneumonia, activation of interferon signaling pathways, low activation of the NF-κB pathway and preferential targeting of spike and nucleocapsid proteins early after intubation were associated with favorable outcomes, whereas loss of interferon signaling, activation of NF-κB-driven programs and specificity for the ORF1ab complex late in disease were associated with mortality. These results suggest that in patients with severe SARS-CoV-2 pneumonia, alveolar T cell interferon responses targeting structural SARS-CoV-2 proteins characterize individuals who recover, whereas responses against nonstructural proteins and activation of NF-κB are associated with poor outcomes.


Assuntos
COVID-19 , NF-kappa B , SARS-CoV-2 , Humanos , COVID-19/imunologia , SARS-CoV-2/imunologia , Masculino , Feminino , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Idoso , Líquido da Lavagem Broncoalveolar/imunologia , Adulto , Transdução de Sinais/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Interferons/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T/imunologia , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/patologia
12.
Antiviral Res ; 230: 105989, 2024 10.
Artigo em Inglês | MEDLINE | ID: mdl-39154753

RESUMO

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.


Assuntos
Herpesvirus Humano 1 , Proteína Carregadora de Folato Reduzido , Replicação Viral , Humanos , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 1/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Proteína Carregadora de Folato Reduzido/metabolismo , Proteína Carregadora de Folato Reduzido/genética , Antivirais/farmacologia , Antivirais/metabolismo , Linhagem Celular , Herpes Simples/virologia , Herpes Simples/metabolismo , Células THP-1 , Transporte Biológico , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Interferons/metabolismo , Transdução de Sinais
13.
Virus Res ; 349: 199451, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39168375

RESUMO

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.


Assuntos
Antivirais , Modelos Animais de Doenças , Vírus da Hepatite B , Vírus Delta da Hepatite , Interferon-alfa , Animais , Camundongos , Interferon-alfa/uso terapêutico , Interferon-alfa/farmacologia , Antivirais/uso terapêutico , Antivirais/farmacologia , Vírus Delta da Hepatite/efeitos dos fármacos , Humanos , Vírus da Hepatite B/efeitos dos fármacos , Vírus da Hepatite B/imunologia , Replicação Viral/efeitos dos fármacos , Hepatite D Crônica/tratamento farmacológico , Hepatite D Crônica/virologia , Coinfecção/tratamento farmacológico , Coinfecção/virologia , Interferons , Hepatite B/tratamento farmacológico , Hepatite B/virologia , Hepatite B/imunologia , Hepatite D/tratamento farmacológico , Hepatite D/virologia , Hepatite D/imunologia , RNA Viral/sangue , DNA Viral/sangue , Hepatite B Crônica/tratamento farmacológico , Hepatite B Crônica/imunologia , Hepatite B Crônica/virologia , Carga Viral/efeitos dos fármacos
14.
Dev Comp Immunol ; 161: 105252, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39173725

RESUMO

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.


Assuntos
Carpas , Proteínas de Peixes , Imunidade Inata , Helicase IFIH1 Induzida por Interferon , Transdução de Sinais , Animais , Carpas/imunologia , Carpas/genética , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Proteínas de Peixes/imunologia , Transdução de Sinais/imunologia , Helicase IFIH1 Induzida por Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon/genética , Interferons/metabolismo , Interferons/imunologia , Interferons/genética , Partícula de Reconhecimento de Sinal/metabolismo , Partícula de Reconhecimento de Sinal/imunologia , Partícula de Reconhecimento de Sinal/genética , Humanos , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Ubiquitinação , Infecções por Rhabdoviridae/imunologia , Infecções por Rhabdoviridae/veterinária , Rhabdoviridae
15.
Fish Shellfish Immunol ; 153: 109853, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39173983

RESUMO

Inhibitors of NF-κB (IκBs) have been implicated as major components of the Rel/NF-κB signaling pathway, playing an important negative regulatory role in host antiviral immunity such as in the activation of interferon (IFN) in vertebrates. In the present study, the immunomodulatory effect of IκB (CgIκB2) on the expression of interferon-like protein (CgIFNLP) was evaluated in Pacific oyster (Crassostrea gigas). After poly (I:C) stimulation, the mRNA expression level of CgIκB2 in haemocytes was significantly down-regulated at 3-12 h while up-regulated at 48-72 h. The mRNA expression of CgIκB2 in haemocytes was significantly up-regulated at 3 h after rCgIFNLP stimulation. In the CgIκB2-RNAi oysters, the mRNA expression of CgIFNLP, interferon regulatory factor-8 (CgIRF8) and NF-κB subunit (CgRel), the abundance of CgIFNLP and CgIRF8 protein in haemocytes, as well as the abundance of CgRel protein in nucleus were significantly increased after poly (I:C) stimulation. Immunofluorescence assay showed that nuclear translocation of CgIRF8 and CgRel protein was promoted in CgIκB2-RNAi oysters compared with that in EGFP-RNAi group. In the CgRel-RNAi oysters, the mRNA and protein expression level of CgIFNLP significantly down-regulated after poly (I:C) stimulation. The collective results indicated that CgIκB2 plays an important role in regulating CgIFNLP expression through its effects on Rel/NF-κB and IRF signaling pathways.


Assuntos
Crassostrea , Regulação da Expressão Gênica , Interferons , NF-kappa B , Poli I-C , Transdução de Sinais , Animais , Crassostrea/genética , Crassostrea/imunologia , Poli I-C/farmacologia , NF-kappa B/genética , NF-kappa B/metabolismo , Regulação da Expressão Gênica/imunologia , Interferons/genética , Interferons/imunologia , Interferons/metabolismo , Imunidade Inata/genética , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Hemócitos/imunologia , Hemócitos/metabolismo
16.
Int J Mol Sci ; 25(15)2024 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-39125716

RESUMO

In patients with endometriosis, refluxed endometrial fragments evade host immunosurveillance, developing into endometriotic lesions. However, the mechanisms underlying this evasion have not been fully elucidated. N-Myc and STAT Interactor (NMI) have been identified as key players in host immunosurveillance, including interferon (IFN)-induced cell death signaling pathways. NMI levels are markedly reduced in the stromal cells of human endometriotic lesions due to modulation by the Estrogen Receptor beta/Histone Deacetylase 8 axis. Knocking down NMI in immortalized human endometrial stromal cells (IHESCs) led to elevated RNA levels of genes involved in cell-to-cell adhesion and extracellular matrix signaling following IFNA treatment. Furthermore, NMI knockdown inhibited IFN-regulated canonical signaling pathways, such as apoptosis mediated by Interferon Stimulated Gene Factor 3 and necroptosis upon IFNA treatment. In contrast, NMI knockdown with IFNA treatment activated non-canonical IFN-regulated signaling pathways that promote proliferation, including ß-Catenin and AKT signaling. Moreover, NMI knockdown in IHESCs stimulated ectopic lesions' growth in mouse endometriosis models. Therefore, NMI is a novel endometriosis suppressor, enhancing apoptosis and inhibiting proliferation and cell adhesion of endometrial cells upon IFN exposure.


Assuntos
Apoptose , Endometriose , Transdução de Sinais , Animais , Feminino , Humanos , Camundongos , Apoptose/genética , Adesão Celular/genética , Proliferação de Células , Endometriose/metabolismo , Endometriose/patologia , Endometriose/genética , Endométrio/metabolismo , Endométrio/patologia , Interferons/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Células Estromais/metabolismo
17.
Cell Death Dis ; 15(8): 608, 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39168971

RESUMO

Recently, various cancer types have been identified to express a distinct subset of Interferon-stimulated genes (ISGs) that mediate therapy resistance. The mechanism through which cancer cells maintain prolonged Interferon stimulation effects to coordinate resistance remains unclear. Our research demonstrated that aberrant upregulation of TAGLN2 is associated with gastric cancer progression, and inhibiting its expression renders gastric cancer cells more susceptible to chemotherapy and radiation. We uncovered a novel role for TAGLN2 in the upregulation of resistance signature ISGs by enhancing YBX1-associated ssDNA aggregation and cGAS-STING pathway activation. TAGLN2 modulates YBX1 by recruiting c-Myc and SOX9 to YBX1 promoter region and directly interacting with AKT-YBX1, thereby enhancing YBX1 phosphorylation and nuclear translocation. Significantly, targeted downregulation of key proteins, inhibition of the TAGLN2-YBX1-AKT interaction (using Fisetin or MK2206) or disruption of the cGAS-STING pathway substantially reduced ssDNA accumulation, subsequent ISGs upregulation, and therapy resistance. The combination of Cisplatin with MK2206 displayed a synergistic effect in the higher TAGLN2-expressing xenograft tumors. Clinical analysis indicated that a derived nine-gene set effectively predicts therapeutic sensitivity and long-term prognosis in gastric cancer patients. These findings suggest that TAGLN2, YBX1 and induced ISGs are novel predictive markers for clinical outcomes, and targeting this axis is an attractive therapeutic sensitization strategy.


Assuntos
Dano ao DNA , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Neoplasias Gástricas , Proteína 1 de Ligação a Y-Box , Humanos , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Proteína 1 de Ligação a Y-Box/metabolismo , Proteína 1 de Ligação a Y-Box/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Linhagem Celular Tumoral , Camundongos , Resistencia a Medicamentos Antineoplásicos/genética , Compostos Heterocíclicos com 3 Anéis/farmacologia , Camundongos Nus , Interferons/metabolismo , Regulação Neoplásica da Expressão Gênica , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/genética , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Feminino , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Camundongos Endogâmicos BALB C , Masculino
18.
Cell Mol Life Sci ; 81(1): 364, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39172244

RESUMO

While severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is characterized by impaired induction of interferons (IFNs) and IFN-stimulated genes (ISGs), the IFNs and ISGs in upper airway is essential to restrict the spread of respiratory virus. Here, we identified the prominent IFN and ISG upregulation in the nasopharynx (NP) of mild and even severe coronavirus disease 2019 (COVID-19) patients (CoV2+) in Omicron era and to compare their clinical outcome depending on the level of IFNs and ISGs. Whereas the induction of IFNB was minimal, transcription of IFNA, IFNG, and IFNLs was significantly increased in the NP of CoV2 + patients. IFNs and ISGs may be more upregulated in the NP of CoV2 + patients at early phases of infection according to viral RNA levels and this is observed even in severe cases. IFN-related innate immune response might be characteristic in macrophages and monocytes at the NP and the CoV2 + patients with higher transcription of IFNs and ISGs in the NP showed a correlation with good prognosis of COVID-19. This study presents that IFNs and ISGs may be upregulated in the NP, even in severe CoV2 + patients depending on viral replication during Omicron-dominant period and the unique IFN-responsiveness in the NP links with COVID-19 clinical outcomes.


Assuntos
COVID-19 , Imunidade Inata , Interferons , Nasofaringe , SARS-CoV-2 , Humanos , COVID-19/imunologia , COVID-19/virologia , Nasofaringe/virologia , Nasofaringe/imunologia , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , Interferons/metabolismo , Interferons/genética , Interferons/imunologia , Masculino , Feminino , Pessoa de Meia-Idade , Adulto , Idoso
19.
J Med Virol ; 96(9): e29889, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39206862

RESUMO

The SARS-CoV-2 Omicron variant is characterized by its high transmissibility, which has caused a worldwide epidemiological event. Yet, it turns ominous once the disease progression degenerates into severe pneumonia and sepsis, presenting a horrendous lethality. To elucidate the alveolar immune or inflammatory landscapes of Omicron critical-ill patients, we performed single-cell RNA-sequencing (scRNA-seq) of bronchoalveolar lavage fluid (BALF) from the patients with critical pneumonia caused by Omicron infection, and analyzed the correlation between the clinical severity scores and different immune cell subpopulations. In the BALF of Omicron critical patients, the alveolar violent myeloid inflammatory environment was determined. ISG15+ neutrophils and CXCL10+ macrophages, both expressed the interferon-stimulated genes (ISGs), were negatively correlated with clinical pulmonary infection score, while septic CST7+ neutrophils and inflammatory VCAN+ macrophages were positively correlated with sequential organ failure assessment. The percentages of ISG15+ neutrophils were associated with more protective alveolar epithelial cells, and may reshape CD4+ T cells to the exhaustive phenotype, thus preventing immune injuries. The CXCL10+ macrophages may promote plasmablast/plasma cell survival and activation as well as the production of specific antibodies. As compared to the previous BALF scRNA-seq data from SARS-CoV-2 wild-type/Alpha critical patients, the subsets of neutrophils and macrophages with pro-inflammatory and immunoregulatory features presented obvious distinctions, suggesting an immune disparity in Omicron variants. Overall, this study provides a BALF single-cell atlas of Omicron critical patients, and suggests that alveolar interferon-responsive neutrophils and macrophages may extricate SARS-CoV-2 Omicron critical patients from the nasty fate of sepsis.


Assuntos
Líquido da Lavagem Broncoalveolar , COVID-19 , Macrófagos , Neutrófilos , SARS-CoV-2 , Sepse , Humanos , COVID-19/imunologia , COVID-19/virologia , Neutrófilos/imunologia , Sepse/imunologia , Sepse/virologia , SARS-CoV-2/imunologia , Masculino , Macrófagos/imunologia , Macrófagos/virologia , Feminino , Pessoa de Meia-Idade , Líquido da Lavagem Broncoalveolar/virologia , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/citologia , Idoso , Citocinas/imunologia , Interferons , Estado Terminal , Adulto
20.
J Med Virol ; 96(9): e29901, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39210614

RESUMO

The mortality and hospitalization rate by COVID-19 dropped significantly currently, but its seasonal outbreaks make antiviral treatment still vital. The mortality and hospitazation rate by COVID-19 dropped significantly currently, but its seasonal ourbreaks make antiviral treatment still vital. In our study, syrian golden hamsters were treated with molnupiravir and interferons (IFNs) after SARS-CoV-2 infection. Their weight changes, pathological changes, virus replication and inflammation levels were evaluated. In the IFNs single treatment, only IFN-α group reduced viral load (p < 0.05) and virus titer in hamster lungs. The TNF-α expression decreased significantly in both IFNs treatment at 2dpi. Histological and immunofluorescence results showed lung damage in the IFNs groups were milder at 4dpi. In the molnupiravir/IFN-α combination treatment, weight loss and virus replication in lung were significantly decreased in the mono-molnupiravir group and combination group (p < 0.05), the expression of IL-6, TNF-α, IL-1ß and MIP-1α also decreased significantly (p < 0.05), but the combination treatment was not more effective than the mono-molnupiravir treatment. Histological and immunofluorescence results showed the lung damage and inflammation in mono-molnupiravir and combination groups were milder. In summary, IFNs treatment had anti-inflammatory effect against SARS-CoV-2, only IFN-α showed a weak antiviral effect. Molnupiravir/IFN-α combination treatment was effective against SARS-CoV-2 but was not superior to mono-molnupiravir treatment. IFN-α could be considered for immunocompromised patients to stimulate and activate early immune responses.


Assuntos
Antivirais , Tratamento Farmacológico da COVID-19 , Hidroxilaminas , Pulmão , Mesocricetus , SARS-CoV-2 , Carga Viral , Replicação Viral , Animais , Antivirais/uso terapêutico , Antivirais/farmacologia , Pulmão/virologia , Pulmão/patologia , Pulmão/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Carga Viral/efeitos dos fármacos , Hidroxilaminas/uso terapêutico , Hidroxilaminas/farmacologia , Cricetinae , Modelos Animais de Doenças , COVID-19/imunologia , COVID-19/virologia , Citidina/análogos & derivados , Citidina/uso terapêutico , Citidina/farmacologia , Quimioterapia Combinada , Interferon-alfa/uso terapêutico , Interferon-alfa/farmacologia , Citocinas/metabolismo , Interferons/uso terapêutico , Masculino , Leucina/análogos & derivados , Leucina/uso terapêutico , Leucina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA