Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 543
Filtrar
1.
Methods Mol Biol ; 2854: 9-18, 2025.
Artigo em Inglês | MEDLINE | ID: mdl-39192113

RESUMO

Antiviral innate immunity is the first line of defence against viruses. The interferon (IFN) signaling pathway, the DNA damage response (DDR), apoptosis, endoplasmic reticulum (ER) stress, and autophagy are involved in antiviral innate immunity. Viruses abrogate the antiviral immune response of cells to replication in various ways. Viral genes/proteins play a key role in evading antiviral innate immunity. Here, we will discuss the interference of viruses with antiviral innate immunity and the strategy for identifying viral gene/protein immune evasion.


Assuntos
Imunidade Inata , Humanos , Proteínas Virais/imunologia , Proteínas Virais/genética , Vírus/imunologia , Vírus/genética , Evasão da Resposta Imune , Viroses/imunologia , Viroses/virologia , Animais , Genes Virais , Autofagia/imunologia , Interações Hospedeiro-Patógeno/imunologia , Transdução de Sinais/imunologia
2.
Int Immunopharmacol ; 142(Pt A): 113091, 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39255680

RESUMO

The cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS)-dependent pathway is a key DNA-sensing pathway that recognizes cytosolic DNA and plays a crucial role in initiating innate immune responses against pathogenic microbes and cancer. Various molecules have been identified as regulators of the cGAS-dependent pathway that controls innate immune responses. However, despite the important roles of Stimulator-of-interferon genes (STING) in the cGAS-dependent pathway, the regulation of its activation has not been elucidated. Here, we show that the E3 ubiquitin ligase, RING finger protein 39 (RNF39), interacts with STING in macrophages and HERK293T cells. Moreover, RNF39 accelerates DNA-sensing pathways by promoting lysine (K)63-linked ubiquitination of STING, and then facilitating the formation of STING-TBK1 complex. Concordantly, Rnf39 deficiency inhibits innate immune responses triggered by DNA viral infection and accelerates viral replication. Furthermore, herpes simplex virus-1 (HSV-1) infection induces RNF39 expression in an IFN-I-dependent manner. Thus, we outline a novel mechanism for controlling STING activation and a feedback mechanism for controlling antiviral immune responses. RNF39 could be a priming intervention target for the prevention and treatment of viral diseases, especially DNA viral infections.

4.
Cell Insight ; 3(5): 100185, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39144256

RESUMO

Monkeypox virus (MPXV) is a DNA virus belonging to the Orthopoxvirus genus within the Poxviridae family which can cause a zoonotic infection. The unexpected non-endemic outbreak of mpox in 2022 is considered as a new global threat. It is imperative to take proactive measures, including enhancing our understanding of MPXV's biology and pathogenesis, and developing novel antiviral strategies. The host immune responses play critical roles in defensing against MPXV infection while the virus has also evolved multiple strategies for immune escape. This review summarizes the biological features, antiviral immunity, immune evasion mechanisms, pathogenicity, and prevention strategies for MPXV.

5.
Virol J ; 21(1): 197, 2024 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-39182136

RESUMO

Serine/threonine kinase receptor-associated protein (STRAP) serves as a scaffold protein and is engaged in a variety of cellular activities, although its importance in antiviral innate immunity is unknown. We discovered that STRAP works as an interferon (IFN)-inducible positive regulator, facilitating type I IFN signaling during pseudorabies virus infection. Mechanistically, STRAP interacts with TBK1 to activate type I IFN signaling. Both the CT and WD40 7 - 6 domains contribute to the function of STRAP. Furthermore, TBK1 competes with PRV-UL50 for binding to STRAP, and STRAP impedes the degradation of TBK1 mediated by PRV-UL50, thereby increasing the interaction between STRAP and TBK1. Overall, these findings reveal a previously unrecognized role for STRAP in innate antiviral immune responses during PRV infection. STRAP could be a potential therapeutic target for viral infectious diseases.


Assuntos
Herpesvirus Suídeo 1 , Imunidade Inata , Interferon Tipo I , Proteínas Serina-Treonina Quinases , Animais , Linhagem Celular , Herpesvirus Suídeo 1/imunologia , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Pseudorraiva/imunologia , Pseudorraiva/virologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , Regulação para Cima
6.
Cell ; 2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39197447

RESUMO

Animal and bacterial cells sense and defend against viral infections using evolutionarily conserved antiviral signaling pathways. Here, we show that viruses overcome host signaling using mechanisms of immune evasion that are directly shared across the eukaryotic and prokaryotic kingdoms of life. Structures of animal poxvirus proteins that inhibit host cGAS-STING signaling demonstrate architectural and catalytic active-site homology shared with bacteriophage Acb1 proteins, which inactivate CBASS anti-phage defense. In bacteria, phage Acb1 proteins are viral enzymes that degrade host cyclic nucleotide immune signals. Structural comparisons of poxvirus protein-2'3'-cGAMP and phage Acb1-3'3'-cGAMP complexes reveal a universal mechanism of host nucleotide immune signal degradation and explain kingdom-specific additions that enable viral adaptation. Chimeric bacteriophages confirm that animal poxvirus proteins are sufficient to evade immune signaling in bacteria. Our findings identify a mechanism of immune evasion conserved between animal and bacterial viruses and define shared rules that explain host-virus interactions across multiple kingdoms of life.

7.
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
8.
Proc Natl Acad Sci U S A ; 121(34): e2403235121, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39145933

RESUMO

The ZIKA virus (ZIKV) evades the host immune response by degrading STAT2 through its NS5 protein, thereby inhibiting type I interferon (IFN)-mediated antiviral immunity. However, the molecular mechanism underlying this process has remained elusive. In this study, we performed a genome-wide CRISPR/Cas9 screen, revealing that ZSWIM8 as the substrate receptor of Cullin3-RING E3 ligase is required for NS5-mediated STAT2 degradation. Genetic depletion of ZSWIM8 and CUL3 substantially impeded NS5-mediated STAT2 degradation. Biochemical analysis illuminated that NS5 enhances the interaction between STAT2 and the ZSWIM8-CUL3 E3 ligase complex, thereby facilitating STAT2 ubiquitination. Moreover, ZSWIM8 knockout endowed A549 and Huh7 cells with partial resistance to ZIKV infection and protected cells from the cytopathic effects induced by ZIKV, which was attributed to the restoration of STAT2 levels and the activation of IFN signaling. Subsequent studies in a physiologically relevant model, utilizing human neural progenitor cells, demonstrated that ZSWIM8 depletion reduced ZIKV infection, resulting from enhanced IFN signaling attributed to the sustained levels of STAT2. Our findings shed light on the role of ZIKV NS5, serving as the scaffold protein, reprograms the ZSWIM8-CUL3 E3 ligase complex to orchestrate STAT2 proteasome-dependent degradation, thereby facilitating evasion of IFN antiviral signaling. Our study provides unique insights into ZIKV-host interactions and holds promise for the development of antivirals and prophylactic vaccines.


Assuntos
Proteínas Culina , Interferon Tipo I , Proteólise , Fator de Transcrição STAT2 , Transdução de Sinais , Ubiquitina-Proteína Ligases , Ubiquitinação , Proteínas não Estruturais Virais , Infecção por Zika virus , Zika virus , Humanos , Fator de Transcrição STAT2/metabolismo , Zika virus/imunologia , Zika virus/fisiologia , Zika virus/metabolismo , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Interferon Tipo I/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Infecção por Zika virus/metabolismo , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia , Proteínas Culina/metabolismo , Células A549 , Células HEK293 , Sistemas CRISPR-Cas
9.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167457, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39134287

RESUMO

DNA virus infection is a significant cause of morbidity and mortality in patients with multiple myeloma (MM). Monocyte dysfunction in MM patients plays a central role in infectious complications, but the precise molecular mechanism underlying the reduced resistance of monocytes to viruses in MM patients remains to be elucidated. Here, we found that MM cells were able to transfer microRNAs (miRNAs) to host monocytes/macrophages via MM cell-derived exosomes, resulting in the inhibition of innate antiviral immune responses. The screening of miRNAs enriched in exosomes derived from the bone marrow (BM) of MM patients revealed five miRNAs that negatively regulate the cGAS-STING antiviral immune response. Notably, silencing these miRNAs with antagomiRs in MM-bearing C57BL/KaLwRijHsd mice markedly reduced viral replication. These findings identify a novel mechanism whereby MM cells possess the capacity to inhibit the innate immune response of the host, thereby rendering patients susceptible to viral infection. Consequently, targeting the aberrant expression patterns of characteristic miRNAs in MM patients is a promising avenue for therapeutic intervention. Considering the miRNA score and relevant clinical factors, we formulated a practical and efficient model for the optimal assessment of susceptibility to DNA viral infection in patients with MM.


Assuntos
Exossomos , Imunidade Inata , Proteínas de Membrana , Camundongos Endogâmicos C57BL , MicroRNAs , Mieloma Múltiplo , Nucleotidiltransferases , MicroRNAs/genética , MicroRNAs/imunologia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Animais , Humanos , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/imunologia , Exossomos/imunologia , Exossomos/genética , Exossomos/metabolismo , Camundongos , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Infecções por Vírus de DNA/imunologia , Linhagem Celular Tumoral , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Feminino , Replicação Viral
10.
mBio ; 15(9): e0146924, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39158293

RESUMO

RNA interference (RNAi) drives powerful antiviral immunity in plants and animals so that many viruses must express viral suppressor of RNAi (VSR) to establish virulent infection. However, little is known about the immune responses conferring resistance against viruses that have evolved the counter-defensive strategy to suppress antiviral RNAi. In this study, we discover that Drosophila cells infected with Drosophila C virus (DCV), a natural viral pathogen of Drosophila known to harbor a potent VSR, exhibit heightened expression of circular RNA circZfh1. circZfh1 confers virus resistance in the presence of viral suppression of antiviral RNAi. Furthermore, we validate that circZfh1 encodes a 274-amino acid protein, CRAV, essential for its antiviral activity. Notably, CRAV differs from its parental Zfh1 gene in a different reading frame, with the C-terminal 69 amino acids unique to CRAV. Our analysis also reveals the presence of CRAV in species within the melanogaster subgroup, with the C-terminal unique fragment undergoing accelerated evolution. Expression of CRAV upregulates the expression of the cytokine Upd3, which binds to its receptor, stimulating the JAK-STAT pathway and enhancing the immune response to DCV infection. Notably, CRISPR/Cas9 knockout of circZfh1 significantly enhances DCV replication in vitro and in vivo, with circZfh1-knockout adult flies displaying heightened disease susceptibility to DCV. In summary, our findings unveil a Drosophila protein-coding circular RNA that activates an innate immune signaling pathway crucial for virus resistance following the suppression of antiviral RNAi by viruses, thereby elucidating a novel counter-defensive strategy.IMPORTANCEEukaryotic hosts possess a complex, multilayered immune system that guards against pathogen invasion. In fruit flies, RNA interference (RNAi) drives robust antiviral immunity, prompting many viruses to express viral suppressors of RNAi (VSRs) to establish virulent infections. However, little is known about immune responses that confer resistance against viruses with potent VSRs. In this study, we discovered that Drosophila cells infected with Drosophila C virus (DCV), a natural viral pathogen possessing a potent VSR, upregulated the expression of circular RNA circZfh1. circZfh1 exhibits DCV-specific antiviral activity, encoding a 274-amino acid protein, CRAV, crucial for its antiviral effects. As a different reading frame from its parental Zfh1 gene, the C-terminal 69 amino acids are unique to CRAV, undergoing faster evolution. CRAV activates the JAK-STAT pathway, enhancing the immune response to DCV infection. Therefore, our work uncovers a new strategy for suppressing viral counter-defense through protein-coding circular RNA in fruit flies.


Assuntos
Dicistroviridae , Proteínas de Drosophila , Drosophila melanogaster , Janus Quinases , RNA Circular , Fatores de Transcrição STAT , Animais , RNA Circular/genética , RNA Circular/imunologia , Janus Quinases/metabolismo , Janus Quinases/genética , Janus Quinases/imunologia , Drosophila melanogaster/imunologia , Drosophila melanogaster/genética , Drosophila melanogaster/virologia , Dicistroviridae/genética , Dicistroviridae/imunologia , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Fatores de Transcrição STAT/imunologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/imunologia , Imunidade Inata/genética , Transdução de Sinais , Interferência de RNA , Drosophila/genética , Drosophila/imunologia , Drosophila/virologia , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/genética
11.
Dev Comp Immunol ; 161: 105254, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39214323

RESUMO

Grass carp (Ctenopharyngodon idella), crucial to global inland aquaculture with a production of 5.8 million tones in 2020, faces significant challenges from hemorrhagic disease caused by grass carp reovirus (GCRV). Rapid mutations compromise current vaccines, underscoring the need for a deeper understanding of antiviral mechanisms to enhance molecular marker-assisted selection. This study investigates the role of Tripartite Motif (TRIM) family in the innate immune response of grass carp, focusing on TRIM103 from Ctenopharyngodon Idella (CiTRIM103), a member of the TRIM-B30.2 family, which includes proteins with the B30.2 domain at the N-terminus, known for antiviral properties in teleosts. CiTRIM103 bind to the outer coat proteins VP5 and VP7 of GCRV. This binding is theorized to strengthen the function of the RIG-I-like Receptor (RLR) signaling pathway, crucial for antiviral responses. Demonstrations using overexpression and RNA interference (RNAi) techniques have shown that CiTRIM103 effectively inhibits GCRV replication. Moreover, molecular docking and pulldown assays suggest potential binding interactions of CiTRIM103's B30.2 domain with GCRV outer coat proteins VP5 and VP7. These interactions impede viral replication, enhance RLR receptor expression, and activate key transcription factors to induce type I interferons (IFNs). These findings elucidate the antiviral mechanisms of CiTRIM103, provide a foundation for future Molecular genetic breeding in grass carp.


Assuntos
Proteínas do Capsídeo , Carpas , Doenças dos Peixes , Proteínas de Peixes , Imunidade Inata , Infecções por Reoviridae , Reoviridae , Transdução de Sinais , Proteínas com Motivo Tripartido , Animais , Reoviridae/fisiologia , Reoviridae/imunologia , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/imunologia , Carpas/imunologia , Infecções por Reoviridae/imunologia , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Proteínas de Peixes/imunologia , Transdução de Sinais/imunologia , Proteínas com Motivo Tripartido/metabolismo , Proteínas com Motivo Tripartido/genética , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Replicação Viral , Ligação Proteica , Simulação de Acoplamento Molecular , Proteína DEAD-box 58/metabolismo , Proteína DEAD-box 58/genética
12.
J Med Virol ; 96(7): e29772, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38949201

RESUMO

The distinct composition and immune response characteristics of bats' innate and adaptive immune systems, which enable them to serve as host of numerous serious zoonotic viruses without falling ill, differ substantially from those of other mammals, it have garnered significant attention. In this article, we offer a systematic review of the names, attributes, and functions of innate and adaptive immune cells & molecules across different bat species. This includes descriptions of the differences shown by research between 71 bat species in 10 families, as well as comparisons between bats and other mammals. Studies of the immune cells & molecules of different bat species are necessary to understand the unique antiviral immunity of bats. By providing comprehensive information on these unique immune responses, it is hoped that new insights will be provided for the study of co-evolutionary dynamics between viruses and the bat immune system, as well as human antiviral immunity.


Assuntos
Imunidade Adaptativa , Quirópteros , Imunidade Inata , Quirópteros/virologia , Quirópteros/imunologia , Animais , Humanos , Vírus/imunologia , Vírus/classificação , Viroses/imunologia , Viroses/virologia
13.
Integr Zool ; 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39016029

RESUMO

Ticks can transmit many pathogens, including arboviruses, to their vertebrate hosts. Arboviruses must overcome or evade defense mechanisms during their passage from the tick gut to the hemolymph, salivary glands, and the feeding site in the host skin. This review summarizes current knowledge of defense mechanisms in specific tick tissues and at the feeding site in the host skin. We discuss the possible roles of these defense mechanisms in viral infection and transmission. The responses of tick salivary proteins to arbovirus infection are also discussed. This review provides information that may help accelerate research on virus-tick interactions.

14.
Annu Rev Plant Biol ; 75(1): 655-677, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39038248

RESUMO

Viruses, causal agents of devastating diseases in plants, are obligate intracellular pathogens composed of a nucleic acid genome and a limited number of viral proteins. The diversity of plant viruses, their diminutive molecular nature, and their symplastic localization pose challenges to understanding the interplay between these pathogens and their hosts in the currently accepted framework of plant innate immunity. It is clear, nevertheless, that plants can recognize the presence of a virus and activate antiviral immune responses, although our knowledge of the breadth of invasion signals and the underpinning sensing events is far from complete. Below, I discuss some of the demonstrated or hypothesized mechanisms enabling viral recognition in plants, the step preceding the onset of antiviral immunity, as well as the strategies viruses have evolved to evade or suppress their detection.


Assuntos
Doenças das Plantas , Imunidade Vegetal , Vírus de Plantas , Plantas , Vírus de Plantas/fisiologia , Vírus de Plantas/patogenicidade , Vírus de Plantas/imunologia , Vírus de Plantas/genética , Doenças das Plantas/virologia , Doenças das Plantas/imunologia , Plantas/virologia , Plantas/imunologia , Interações Hospedeiro-Patógeno/imunologia , Evasão da Resposta Imune
15.
Fish Shellfish Immunol ; 153: 109765, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39004296

RESUMO

TRIM family proteins are widely found in multicellular organisms and are involved in a wide range of life activities, and also act as crucial regulators in the antiviral natural immune response. This study aimed to reveal the molecular mechanism of rainbow trout TRIM protein in the anti-IHNV process. The results demonstrated that 99.1 % homology between the rainbow trout and the chinook salmon (Oncorhynchus tshawytscha) TRIM32. When rainbow trout were infected with IHNV, the TRIM32 was highly expressed in the gill, spleen, kidney and blood. Meanwhile, rainbow trout TRIM32 has E3 ubiquitin ligase activity and undergoes K29-linked polyubiquitination modifications dependent on the RING structural domain was determined by immunoprecipitation. TRIM32 could interact with the NV protein of IHNV and degrade NV protein through the ubiquitin-proteasome pathway, and was also able to activate NF-κB transcription, thereby inhibiting the replication of IHNV. Moreover, the results of the animal studies showed that the survival rate of rainbow trout overexpressing TRIM32 was 70.2 % which was significantly higher than that of the control group, and stimulating the body to produce high levels of IgM when the host was infected with the virus. In addition, TRIM32 can activate the NF-κB signalling pathway and participate in the antiviral natural immune response. The results of this study will help us to understand the molecular mechanism of TRIM protein resistance in rainbow trout, and provide new ideas for disease resistance breeding, vaccine development and immune formulation development in rainbow trout.


Assuntos
Doenças dos Peixes , Proteínas de Peixes , Imunidade Inata , Oncorhynchus mykiss , Infecções por Rhabdoviridae , Proteínas com Motivo Tripartido , Animais , Oncorhynchus mykiss/imunologia , Doenças dos Peixes/imunologia , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Infecções por Rhabdoviridae/imunologia , Infecções por Rhabdoviridae/veterinária , Imunidade Inata/genética , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/imunologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia , Regulação da Expressão Gênica/imunologia , Perfilação da Expressão Gênica/veterinária , Vírus da Necrose Hematopoética Infecciosa/imunologia , Vírus da Necrose Hematopoética Infecciosa/fisiologia , Alinhamento de Sequência/veterinária , Filogenia
16.
Methods Mol Biol ; 2813: 309-320, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38888786

RESUMO

Nanoparticle carriers enable the multivalent delivery of nucleic acids to cells and protect them from degradation. In this chapter, we present a comprehensive overview of four methodologies: electrophoretic mobility shift assay (EMSA), alamarBlue/CFDA-AM cell viability dyes, fluorescence microscopy, and antiviral assays, which collectively are tools to explore interactions between nucleic acids and nanoparticles, and their biological efficacy. These assays provide insights into binding potential, cytotoxicity, and antiviral efficacy of nucleic acid-based nanoparticle treatments furthering the development of effective antiviral therapeutics.


Assuntos
Antivirais , Nanopartículas , Ácidos Nucleicos , Nanopartículas/química , Antivirais/farmacologia , Humanos , Ácidos Nucleicos/química , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Cátions/química , Sobrevivência Celular/efeitos dos fármacos , Microscopia de Fluorescência , Portadores de Fármacos/química , Animais
17.
Vaccines (Basel) ; 12(6)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38932335

RESUMO

Porcine reproductive and respiratory syndrome (PRRS) remains a formidable challenge for the global pig industry. Caused by PRRS virus (PRRSV), this disease primarily affects porcine reproductive and respiratory systems, undermining effective host interferon and other immune responses, resulting in vaccine ineffectiveness. In the absence of specific antiviral treatments for PRRSV, vaccines play a crucial role in managing the disease. The current market features a range of vaccine technologies, including live, inactivated, subunit, DNA, and vector vaccines, but only modified live virus (MLV) and killed virus (KV) vaccines are commercially available for PRRS control. Live vaccines are promoted for their enhanced protective effectiveness, although their ability to provide cross-protection is modest. On the other hand, inactivated vaccines are emphasized for their safety profile but are limited in their protective efficacy. This review updates the current knowledge on PRRS vaccines' interactions with the host interferon system, and other immunological aspects, to assess their current status and evaluate advents in PRRSV vaccine development. It presents the strengths and weaknesses of both live attenuated and inactivated vaccines in the prevention and management of PRRS, aiming to inspire the development of innovative strategies and technologies for the next generation of PRRS vaccines.

18.
Microorganisms ; 12(6)2024 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-38930591

RESUMO

Lacticaseibacillus rhamnosus CRL1505 possesses immunomodulatory activities in the gastrointestinal and respiratory tracts when administered orally. Its adhesion to the intestinal mucosa does not condition its beneficial effects. The intranasal administration of L. rhamnosus CRL1505 is more effective than the oral route at modulating immunity in the respiratory tract. Nonetheless, it has not yet been established whether the adherence of the CRL1505 strain to the respiratory mucosa is needed to provide the immune benefits to the host. In this study, we evaluated the role of adhesion to the respiratory mucosa of the mucus-binding factor (mbf) knock-out L. rhamnosus CRL1505 mutant (Δmbf CRL1505) in the context of a Toll-like receptor 3 (TLR3)-triggered innate immunity response. In vitro adhesion studies in porcine bronchial epitheliocytes (PBE cells) indicated that L. rhamnosus Δmbf CRL1505 adhered weakly compared to the wild-type strain. However, in vivo studies in mice demonstrated that the Δmbf CRL1505 also reduced lung damage and modulated cytokine production in the respiratory tract after the activation of TLR3 to a similar extent as the wild-type strain. In addition, the mutant and the wild-type strains modulated the production of cytokines and antiviral factors by alveolar macrophages in the same way. These results suggest that the Mbf protein is partially involved in the ability of L. rhamnosus CRL1505 to adhere to the respiratory epithelium, but the protein is not necessary for the CRL1505 strain to exert its immunomodulatory beneficial effects. These findings are a step forward in the understanding of molecular interactions that mediate the beneficial effects of nasally administered probiotics.

19.
Gene ; 927: 148649, 2024 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-38852697

RESUMO

During the birch pollen season an enhanced incidence of virus infections is noticed, raising the question whether pollen can affect anti-viral responses independent of allergic reactions. We previously showed that birch pollen-treatment of monocyte-derived dendritic cells (moDC) enhances human cytomegalovirus (HCMV) infection. Here we addressed how in moDC the relatively weak pollen response can affect the comparably strong response to HCMV. To this end, moDC were stimulated with aqueous birch pollen extract (APE), HCMV, and APE with HCMV, and transcriptomic signatures were determined after 6 and 24 h of incubation. Infection was monitored upon exposure of moDC to GFP expressing HCMV by flow cytometric analysis of GFP expressing cells. Principle component analysis of RNA sequencing data revealed close clustering of mock and APE treated moDC, whereas HCMV as well as APE with HCMV treated moDC clustered separately after 6 and 24 h of incubation, respectively. Communally induced genes were detected in APE, HCMV and APE with HCMV treated moDC. In APE with HCMV treated moDC, the comparably weak APE induced signatures were maintained after HCMV exposure. In particular, NF-κB/RELA and PI3K/AKT/MAPK signaling were altered upon APE with HCMV exposure. Earlier, we discovered that NF-κB inhibition alleviated APE induced enhancement of HCMV infection. Here we additionally found that impairment of PI3K signaling reduced HCMV infection in HCMV and APE with HCMV treated moDC. APE treated moDC that were exposed to HCMV show a unique host gene signature, which to a large extent is regulated by NF-κB activation and PI3K/AKT/MAPK signaling.


Assuntos
Betula , Citomegalovirus , Células Dendríticas , Pólen , Células Dendríticas/virologia , Células Dendríticas/metabolismo , Células Dendríticas/imunologia , Pólen/genética , Pólen/imunologia , Citomegalovirus/genética , Citomegalovirus/fisiologia , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/imunologia , Transcriptoma , Transdução de Sinais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição RelA/metabolismo , Fator de Transcrição RelA/genética , Células Cultivadas
20.
Dev Comp Immunol ; 158: 105208, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38834141

RESUMO

Interferon regulatory factors (IRFs) are transcription factors involved in immune responses, such as pathogen response regulation, immune cell growth, and differentiation. IRFs are necessary for the synthesis of type I interferons through a signaling cascade when pathogen recognition receptors identify viral DNA or RNA. We discovered that irf3 is expressed in the early embryonic stages and in all immune organs of adult zebrafish. We demonstrated the antiviral immune mechanism of Irf3 against viral hemorrhagic septicemia virus (VHSV) using CRISPR/Cas9-mediated knockout zebrafish (irf3-KO). In this study, we used a truncated Irf3 protein, encoded by irf3 with a 10 bp deletion, for further investigation. Upon VHSV injection, irf3-KO zebrafish showed dose-dependent high and early mortality compared with zebrafish with the wild-type Irf3 protein (WT), confirming the antiviral activity of Irf3. Based on the results of expression analysis of downstream genes upon VHSV challenge, we inferred that Irf3 deficiency substantially affects the expression of ifnphi1 and ifnphi2. However, after 5 days post infection (dpi), ifnphi3 expression was not significantly altered in irf3-KO compared to that in WT, and irf7 transcription showed a considerable increase in irf3-KO after 5 dpi, indicating irf7's control over ifnphi3 expression. The significantly reduced expression of isg15, viperin, mxa, and mxb at 3 dpi also supported the effect of Irf3 deficiency on the antiviral activity in the early stage of infection. The higher mortality in irf3-KO zebrafish than in WT might be due to an increased inflammation and tissue damage that occurs in irf3-KO because of delayed immune response. Our results suggest that Irf3 plays a role in antiviral immunity of zebrafish by modulating critical immune signaling molecules and regulating antiviral immune genes.


Assuntos
Sistemas CRISPR-Cas , Técnicas de Inativação de Genes , Septicemia Hemorrágica Viral , Fator Regulador 3 de Interferon , Novirhabdovirus , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Peixe-Zebra/genética , Peixe-Zebra/imunologia , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Novirhabdovirus/fisiologia , Novirhabdovirus/imunologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Septicemia Hemorrágica Viral/imunologia , Septicemia Hemorrágica Viral/genética , Septicemia Hemorrágica Viral/virologia , Animais Geneticamente Modificados , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Doenças dos Peixes/genética , Imunidade Inata/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Infecções por Rhabdoviridae/imunologia , Infecções por Rhabdoviridae/virologia , Modelos Animais de Doenças , Interferons
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA