Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.109
Filtrar
1.
Arch Virol ; 169(8): 157, 2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-38969819

RESUMO

Viruses use various strategies and mechanisms to deal with cells and proteins of the immune system that form a barrier against infection. One of these mechanisms is the encoding and production of viral microRNAs (miRNAs), whose function is to regulate the gene expression of the host cell and the virus, thus creating a suitable environment for survival and spreading viral infection. miRNAs are short, single-stranded, non-coding RNA molecules that can regulate the expression of host and viral proteins, and due to their non-immunogenic nature, they are not eliminated by the cells of the immune system. More than half of the viral miRNAs are encoded and produced by Orthoherpesviridae family members. Human cytomegalovirus (HCMV) produces miRNAs that mediate various processes in infected cells to contribute to HCMV pathogenicity, including immune escape, viral latency, and cell apoptosis. Here, we discuss which cellular and viral proteins or cellular pathways and processes these mysterious molecules target to evade immunity and support viral latency in infected cells. We also discuss current evidence that their function of bypassing the host's innate and adaptive immune system is essential for the survival and multiplication of the virus and the spread of HCMV infection.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus , Evasão da Resposta Imune , MicroRNAs , Latência Viral , Citomegalovirus/genética , Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Latência Viral/genética , MicroRNAs/genética , Humanos , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/imunologia , RNA Viral/genética , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/genética , Regulação Viral da Expressão Gênica
2.
Virologie (Montrouge) ; 28(3): 187-197, 2024 Jun 01.
Artigo em Francês | MEDLINE | ID: mdl-38970340

RESUMO

Orthoflaviviruses are enveloped positive-sense RNA viruses comprising numerous human pathogens transmitted by hematophagous arthropods. This includes viruses such as dengue virus, Zika virus, and yellow fever virus. The viral nonstructural protein NS1 plays a central role in the pathogenesis and cycle of these viruses by acting in two different forms: associated with the plasma membrane (NS1m) or secreted outside the cell (NS1s). The versatility of NS1 is evident in its ability to modulate various aspects of the infectious process, from immune evasion to pathogenesis. As an intracellular protein, it disrupts many processes, interfering with signaling pathways and facilitating viral replication in concert with other viral proteins. As a secreted protein, NS1 actively participates in immune evasion, interfering with the host immune system, inhibiting the complement system, facilitating viral dissemination, and disrupting the integrity of endothelial barriers. This review primarily aims to address the role of NS1 in viral pathogenesis associated with orthoflaviviruses.


Assuntos
Proteínas não Estruturais Virais , Replicação Viral , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/fisiologia , Humanos , Animais , Infecções por Flavivirus/virologia , Evasão da Resposta Imune , Flavivirus/fisiologia , Flavivirus/patogenicidade , Zika virus/fisiologia , Zika virus/patogenicidade , Vírus da Dengue/fisiologia
3.
Oncoimmunology ; 13(1): 2376264, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38988824

RESUMO

Functional roles of SIGLEC15 in hepatocellular carcinoma (HCC) were not clear, which was recently found to be an immune inhibitor with similar structure of inhibitory B7 family members. SIGLEC15 expression in HCC was explored in public databases and further examined by PCR analysis. SIGLEC15 and PD-L1 expression patterns were examined in HCC samples through immunohistochemistry. SIGLEC15 expression was knocked-down or over-expressed in HCC cell lines, and CCK8 tests were used to examine cell proliferative ability in vitro. Influences of SIGLEC15 expression on tumor growth were examined in immune deficient and immunocompetent mice respectively. Co-culture system of HCC cell lines and Jurkat cells, flow cytometry analysis of tumor infiltrated immune cells and further sequencing analyses were performed to investigate how SIGLEC15 could affect T cells in vitro and in vivo. We found SIGLEC15 was increased in HCC tumor tissues and was negatively correlated with PD-L1 in HCC samples. In vitro and in vivo models demonstrated inhibition of SIGLEC15 did not directly influence tumor proliferation. However, SIGLEC15 could promoted HCC immune evasion in immune competent mouse models. Knock-out of Siglec15 could inhibit tumor growth and reinvigorate CD8+ T cell cytotoxicity. Anti-SIGLEC15 treatment could effectively inhibit tumor growth in mouse models with or without mononuclear phagocyte deletion. Bulk and single-cell RNA sequencing data of treated mouse tumors demonstrated SIGLEC15 could interfere CD8+ T cell viability and induce cell apoptosis. In all, SIGLEC15 was negatively correlated with PD-L1 in HCC and mainly promote HCC immune evasion through inhibition of CD8+ T cell viability and cytotoxicity.


Assuntos
Apoptose , Antígeno B7-H1 , Linfócitos T CD8-Positivos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Animais , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Antígeno B7-H1/imunologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Camundongos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Evasão Tumoral/genética , Linhagem Celular Tumoral , Proliferação de Células , Masculino , Feminino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Evasão da Resposta Imune , Imunoglobulinas
4.
Commun Biol ; 7(1): 824, 2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-38971948

RESUMO

The expression dysregulation of microRNAs (miRNA) has been widely reported during cancer development, however, the underling mechanism remains largely unanswered. In the present work, we performed a systematic integrative study for genome-wide DNA methylation, copy number variation and miRNA expression data to identify mechanisms underlying miRNA dysregulation in lower grade glioma. We identify 719 miRNAs whose expression was associated with alterations of copy number variation or promoter methylation. Integrative multi-omics analysis revealed four subtypes with differing prognoses. These glioma subtypes exhibited distinct immune-related characteristics as well as clinical and genetic features. By construction of a miRNA regulatory network, we identified candidate miRNAs associated with immune evasion and response to immunotherapy. Finally, eight prognosis related miRNAs were validated to promote cell migration, invasion and proliferation through in vitro experiments. Our study reveals the crosstalk among DNA methylation, copy number variation and miRNA expression for immune regulation in glioma, and could have important implications for patient stratification and development of biomarkers for immunotherapy approaches.


Assuntos
Neoplasias Encefálicas , Variações do Número de Cópias de DNA , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Glioma , MicroRNAs , Humanos , Glioma/genética , Glioma/imunologia , Glioma/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Epigenômica , Genômica , Redes Reguladoras de Genes , Linhagem Celular Tumoral , Evasão da Resposta Imune/genética , Epigênese Genética , Feminino , Masculino , Prognóstico , Gradação de Tumores
6.
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
7.
J Extracell Vesicles ; 13(7): e12484, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39041344

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is characterised by immune evasion that contribute to poor prognosis. Cancer-associated fibroblasts (CAFs) play a pivotal role in orchestrating the PDAC tumour microenvironment. We investigated the role of CAF-derived extracellular vesicle (EV)-packaged long non-coding RNAs (lncRNAs) in immune evasion and explored gene therapy using engineered EVs loading small interfering RNAs (siRNAs) as a potential therapeutic strategy. Our findings highlight the significance of EV-packaged lncRNA RP11-161H23.5 from CAF in promoting PDAC immune evasion by downregulating HLA-A expression, a key component of antigen presentation. Mechanistically, RP11-161H23.5 forms a complex with CNOT4, a subunit of the mRNA deadenylase CCR4-NOT complex, enhancing the degradation of HLA-A mRNA by shortening its poly(A) tail. This immune evasion mechanism compromises the anti-tumour immune response. To combat this, we propose an innovative approach utilising engineered EVs as natural and biocompatible nanocarriers for siRNA-based gene therapy and this strategy holds promise for enhancing the effectiveness of immunotherapy in PDAC. Overall, our study sheds light on the critical role of CAF-derived EV-packaged lncRNA RP11-161H23.5/CNOT4/HLA-A axis in PDAC immune evasion and presents a novel avenue for therapeutic intervention.


Assuntos
Fibroblastos Associados a Câncer , Carcinoma Ductal Pancreático , Vesículas Extracelulares , Antígenos HLA-A , Neoplasias Pancreáticas , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/imunologia , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/imunologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/terapia , Linhagem Celular Tumoral , Antígenos HLA-A/genética , Antígenos HLA-A/imunologia , Antígenos HLA-A/metabolismo , Evasão da Resposta Imune , Regulação Neoplásica da Expressão Gênica , Regulação para Baixo , RNA Interferente Pequeno , Microambiente Tumoral/imunologia , Animais , Evasão Tumoral , Camundongos
8.
Med Microbiol Immunol ; 213(1): 13, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38967888

RESUMO

Candida auris is an emerging pathogenic yeast that has been categorized as a global public health threat and a critical priority among fungal pathogens. Despite this, the immune response against C. auris infection is still not well understood. Hosts fight Candida infections through the immune system that recognizes pathogen-associated molecular patterns such as ß-glucan, mannan, and chitin on the fungal cell wall. In this study, levels of ß-glucan and mannan exposures in C. auris grown under different physiologically relevant stimuli were quantified by flow cytometry-based analysis. Lactate, hypoxia, and sublethal concentration of fluconazole trigger a decrease in surface ß-glucan while low pH triggers an increase in ß-glucan. There is no inverse pattern between exposure levels of ß-glucan and mannan in the cell wall architecture among the three clades. To determine the effect of cell wall remodeling on the immune response, a phagocytosis assay was performed, followed by quantification of released cytokines by ELISA. Lactate-induced decrease in ß-glucan leads to reduced uptake of C. auris by PMA-differentiated THP-1 and RAW 264.7 macrophages. Furthermore, reduced production of CCL3/MIP-1⍺ but not TNF-⍺ and IL-10 were observed. An in vivo infection analysis using silkworms reveals that a reduction in ß-glucan triggers an increase in the virulence of C. auris. This study demonstrates that ß-glucan alteration occurs in C. auris and serves as an escape mechanism from immune cells leading to increased virulence.


Assuntos
Candida auris , Parede Celular , Evasão da Resposta Imune , beta-Glucanas , beta-Glucanas/metabolismo , Animais , Virulência , Camundongos , Parede Celular/imunologia , Parede Celular/química , Parede Celular/metabolismo , Humanos , Candida auris/patogenicidade , Células RAW 264.7 , Candidíase/microbiologia , Candidíase/imunologia , Citocinas/metabolismo , Fagocitose , Macrófagos/imunologia , Macrófagos/microbiologia , Mananas/farmacologia , Ácido Láctico/metabolismo , Modelos Animais de Doenças , Células THP-1
9.
Science ; 385(6705): eadl6173, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-38991060

RESUMO

Isocitrate dehydrogenase 1 (IDH1) is the most commonly mutated metabolic gene across human cancers. Mutant IDH1 (mIDH1) generates the oncometabolite (R)-2-hydroxyglutarate, disrupting enzymes involved in epigenetics and other processes. A hallmark of IDH1-mutant solid tumors is T cell exclusion, whereas mIDH1 inhibition in preclinical models restores antitumor immunity. Here, we define a cell-autonomous mechanism of mIDH1-driven immune evasion. IDH1-mutant solid tumors show selective hypermethylation and silencing of the cytoplasmic double-stranded DNA (dsDNA) sensor CGAS, compromising innate immune signaling. mIDH1 inhibition restores DNA demethylation, derepressing CGAS and transposable element (TE) subclasses. dsDNA produced by TE-reverse transcriptase (TE-RT) activates cGAS, triggering viral mimicry and stimulating antitumor immunity. In summary, we demonstrate that mIDH1 epigenetically suppresses innate immunity and link endogenous RT activity to the mechanism of action of a US Food and Drug Administration-approved oncology drug.


Assuntos
Evasão da Resposta Imune , Imunidade Inata , Isocitrato Desidrogenase , Neoplasias , Animais , Humanos , Camundongos , Linhagem Celular Tumoral , DNA/metabolismo , Desmetilação do DNA , Metilação de DNA , Elementos de DNA Transponíveis , Epigênese Genética , Glutaratos/metabolismo , Imunidade Inata/genética , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Mutação , Neoplasias/imunologia , Neoplasias/genética , Nucleotidiltransferases/genética , Evasão Tumoral , Evasão da Resposta Imune/genética
10.
Nature ; 631(8021): 617-626, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38961298

RESUMO

SARS-CoV-2 variants acquire mutations in the spike protein that promote immune evasion1 and affect other properties that contribute to viral fitness, such as ACE2 receptor binding and cell entry2,3. Knowledge of how mutations affect these spike phenotypes can provide insight into the current and potential future evolution of the virus. Here we use pseudovirus deep mutational scanning4 to measure how more than 9,000 mutations across the full XBB.1.5 and BA.2 spikes affect ACE2 binding, cell entry or escape from human sera. We find that mutations outside the receptor-binding domain (RBD) have meaningfully affected ACE2 binding during SARS-CoV-2 evolution. We also measure how mutations to the XBB.1.5 spike affect neutralization by serum from individuals who recently had SARS-CoV-2 infections. The strongest serum escape mutations are in the RBD at sites 357, 420, 440, 456 and 473; however, the antigenic effects of these mutations vary across individuals. We also identify strong escape mutations outside the RBD; however, many of them decrease ACE2 binding, suggesting they act by modulating RBD conformation. Notably, the growth rates of human SARS-CoV-2 clades can be explained in substantial part by the measured effects of mutations on spike phenotypes, suggesting our data could enable better prediction of viral evolution.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Mutação , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/imunologia , Humanos , SARS-CoV-2/genética , SARS-CoV-2/imunologia , SARS-CoV-2/classificação , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , COVID-19/virologia , COVID-19/imunologia , COVID-19/genética , Ligação Proteica , Evasão da Resposta Imune/genética , Anticorpos Neutralizantes/imunologia , Internalização do Vírus , Evolução Molecular , Modelos Moleculares , Sítios de Ligação , Domínios Proteicos , Testes de Neutralização , Análise Mutacional de DNA
11.
Front Immunol ; 15: 1403070, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39015575

RESUMO

Background: The cGAS-STING axis-mediated type I interferon pathway is a crucial strategy for host defense against DNA virus infection. Numerous evasion strategies developed by the pseudorabies virus (PRV) counteract host antiviral immunity. To what extent PRV-encoded proteins evade the cGAS-STING signaling pathway is unknown. Methods: Using US2 stably expressing cell lines and US2-deficient PRV model, we revealed that the PRV tegument protein US2 reduces STING protein stability and downregulates STING-mediated antiviral signaling. Results: To promote K48-linked ubiquitination and STING degradation, US2 interacts with the LBD structural domain of STING and recruits the E3 ligase TRIM21. TRIM21 deficiency consistently strengthens the host antiviral immune response brought on by PRV infection. Additionally, US2-deficient PRV is less harmful in mice. Conclusions: Our study implies that PRV US2 inhibits IFN signaling by a new mechanism that selectively targets STING while successfully evading the host antiviral response. As a result, the present study reveals a novel strategy by which PRV evades host defense and offers explanations for why the Bartha-K61 classical vaccine strain failed to offer effective defense against PRV variant strains in China, indicating that US2 may be a key target for developing gene-deficient PRV vaccines.


Assuntos
Herpesvirus Suídeo 1 , Imunidade Inata , Proteínas de Membrana , Nucleotidiltransferases , Pseudorraiva , Transdução de Sinais , Animais , Proteínas de Membrana/metabolismo , Proteínas de Membrana/imunologia , Proteínas de Membrana/genética , Transdução de Sinais/imunologia , Herpesvirus Suídeo 1/imunologia , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/imunologia , Camundongos , Pseudorraiva/imunologia , Pseudorraiva/virologia , Humanos , Ubiquitinação , Ribonucleoproteínas/imunologia , Ribonucleoproteínas/metabolismo , Evasão da Resposta Imune , Interações Hospedeiro-Patógeno/imunologia , Células HEK293
12.
Cell Death Dis ; 15(7): 511, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39019859

RESUMO

Immune checkpoint inhibitors, particularly PD-1/PD-L1 blockades, have been approved for unresectable hepatocellular carcinoma (HCC). However, high resistance rates still limit their efficacy, highlighting the urgent need to understand the underlying mechanisms and develop strategies for overcoming the resistance. In this study, tankyrasel binding protein 1 (TNKS1BP1) was found to interact with tripartite motif containing 21 (TRIM21) and mediated the ubiquitination of CCR4-NOT transcription complex subunit 4 (CNOT4) at the K239 residue via K48 and K6 linkage, which was essential for its tumorigenesis function. Autophagy and lipid reprogramming were identified as two possible mechanisms underlying the pro-tumor effect of TNKS1BP1. Upregulated TNKS1BP1 inhibited autophagy while induced lipid accumulation by inhibiting the JAK2/STAT3 pathway upon the degradation of CNOT4 in HCC. Importantly, knocking down TNKS1BP1 synergized with anti-PD-L1 treatment by upregulating PD-L1 expression on tumor cells via the JAK2/STAT3 pathway, and remodeling the tumor microenvironment by increasing infiltration of tumor-infiltrating lymphocytes as well as augmenting the effect of cytotoxic T lymphocytes. In conclusion, this study identified TNKS1BP1 as a predictive biomarker for patient prognosis and a promising therapeutic target to overcome anti-PD-L1 resistance in HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Ribonucleoproteínas , Ubiquitinação , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/genética , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/imunologia , Ribonucleoproteínas/metabolismo , Animais , Camundongos , Progressão da Doença , Linhagem Celular Tumoral , Camundongos Nus , Evasão da Resposta Imune , Fatores de Transcrição/metabolismo , Regulação Neoplásica da Expressão Gênica , Camundongos Endogâmicos BALB C
14.
Viruses ; 16(6)2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38932162

RESUMO

Vaccinia virus is the most successful vaccine in human history and functions as a protective vaccine against smallpox and monkeypox, highlighting the importance of ongoing research into vaccinia due to its genetic similarity to other emergent poxviruses. Moreover, vaccinia's ability to accommodate large genetic insertions makes it promising for vaccine development and potential therapeutic applications, such as oncolytic agents. Thus, understanding how superior immunity is generated by vaccinia is crucial for designing other effective and safe vaccine strategies. During vaccinia inoculation by scarification, the skin serves as a primary site for the virus-host interaction, with various cell types playing distinct roles. During this process, hematopoietic cells undergo abortive infections, while non-hematopoietic cells support the full viral life cycle. This differential permissiveness to viral replication influences subsequent innate and adaptive immune responses. Dendritic cells (DCs), key immune sentinels in peripheral tissues such as skin, are pivotal in generating T cell memory during vaccinia immunization. DCs residing in the skin capture viral antigens and migrate to the draining lymph nodes (dLN), where they undergo maturation and present processed antigens to T cells. Notably, CD8+ T cells are particularly significant in viral clearance and the establishment of long-term protective immunity. Here, we will discuss vaccinia virus, its continued relevance to public health, and viral strategies permissive to immune escape. We will also discuss key events and populations leading to long-term protective immunity and remaining key gaps.


Assuntos
Evasão da Resposta Imune , Vaccinia virus , Vacínia , Vaccinia virus/imunologia , Vaccinia virus/genética , Humanos , Animais , Vacínia/imunologia , Vacínia/virologia , Células Dendríticas/imunologia , Replicação Viral , Imunidade Adaptativa , Linfócitos T CD8-Positivos/imunologia
15.
Viruses ; 16(6)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38932192

RESUMO

Currently, SARS-CoV-2 has evolved into various variants, including the numerous highly mutated Omicron sub-lineages, significantly increasing immune evasion ability. The development raises concerns about the possibly diminished effectiveness of available vaccines and antibody-based therapeutics. Here, we describe those representative categories of broadly neutralizing antibodies (bnAbs) that retain prominent effectiveness against emerging variants including Omicron sub-lineages. The molecular characteristics, epitope conservation, and resistance mechanisms of these antibodies are further detailed, aiming to offer suggestion or direction for the development of therapeutic antibodies, and facilitate the design of vaccines with broad-spectrum potential.


Assuntos
Anticorpos Antivirais , Anticorpos Amplamente Neutralizantes , COVID-19 , Epitopos , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , SARS-CoV-2/imunologia , Humanos , Anticorpos Antivirais/imunologia , COVID-19/imunologia , COVID-19/virologia , Anticorpos Amplamente Neutralizantes/imunologia , Epitopos/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Anticorpos Neutralizantes/imunologia , Evasão da Resposta Imune , Vacinas contra COVID-19/imunologia
16.
Viruses ; 16(6)2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38932205

RESUMO

African swine fever (ASF) is a contagious viral disease affecting pigs and wild boars. It typically presents as a hemorrhagic fever but can also manifest in various forms, ranging from acute to asymptomatic. ASF has spread extensively globally, significantly impacting the swine industry. The complex and highly variable character of the ASFV genome makes vaccine development and disease surveillance extremely difficult. The overall trend in ASFV evolution is towards decreased virulence and increased transmissibility. Factors such as gene mutation, viral recombination, and the strain-specificity of virulence-associated genes facilitate viral variations. This review deeply discusses the influence of these factors on viral immune evasion, pathogenicity, and the ensuing complexities encountered in vaccine development, disease detection, and surveillance. The ultimate goal of this review is to thoroughly explore the genetic evolution patterns and variation mechanisms of ASFV, providing a theoretical foundation for advancement in vaccine and diagnostic technologies.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Variação Genética , Genoma Viral , Vírus da Febre Suína Africana/genética , Animais , Suínos , Febre Suína Africana/virologia , Virulência , Vacinas Virais/imunologia , Vacinas Virais/genética , Evolução Molecular , Evasão da Resposta Imune/genética , Mutação , Desenvolvimento de Vacinas
17.
Annu Rev Immunol ; 42(1): 551-584, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38941604

RESUMO

Poxviruses have evolved a wide array of mechanisms to evade the immune response, and we provide an overview of the different immunomodulatory strategies. Poxviruses prevent the recognition of viral DNA that triggers the immune responses and inhibit signaling pathways within the infected cell. A unique feature of poxviruses is the production of secreted proteins that mimic cytokines and cytokine receptors, acting as decoy receptors to neutralize the activity of cytokines and chemokines. The capacity of these proteins to evade cellular immune responses by inhibiting cytokine activation is complemented by poxviruses' strategies to block natural killer cells and cytotoxic T cells, often through interfering with antigen presentation pathways. Mechanisms that target complement activation are also encoded by poxviruses. Virus-encoded proteins that target immune molecules and pathways play a major role in immune modulation, and their contribution to viral pathogenesis, facilitating virus replication or preventing immunopathology, is discussed.


Assuntos
Evasão da Resposta Imune , Infecções por Poxviridae , Poxviridae , Humanos , Poxviridae/imunologia , Poxviridae/fisiologia , Animais , Infecções por Poxviridae/imunologia , Citocinas/metabolismo , Transdução de Sinais , Proteínas Virais/metabolismo , Proteínas Virais/imunologia , Apresentação de Antígeno/imunologia , Interações Hospedeiro-Patógeno/imunologia
18.
Vet Res ; 55(1): 79, 2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38886840

RESUMO

Porcine deltacoronavirus (PDCoV) is an enteropathogenic coronavirus that has been reported to use various strategies to counter the host antiviral innate immune response. The cGAS-STING signalling pathway plays an important role in antiviral innate immunity. However, it remains unclear whether PDCoV achieves immune evasion by regulating the cGAS-STING pathway. Here, we demonstrated that the nonstructural protein 2 (nsp2) encoded by PDCoV inhibits cGAS-STING-mediated type I and III interferon (IFN) responses via the regulation of porcine STING (pSTING) stability. Mechanistically, ectopically expressed PDCoV nsp2 was found to interact with the N-terminal region of pSTING. Consequently, pSTING was degraded through K48-linked ubiquitination and the proteasomal pathway, leading to the disruption of cGAS-STING signalling. Furthermore, K150 and K236 of pSTING were identified as crucial residues for nsp2-mediated ubiquitination and degradation. In summary, our findings provide a basis for elucidating the immune evasion mechanism of PDCoV and will contribute to the development of targets for anti-coronavirus drugs.


Assuntos
Deltacoronavirus , Proteínas não Estruturais Virais , Animais , Suínos , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Deltacoronavirus/genética , Deltacoronavirus/fisiologia , Doenças dos Suínos/virologia , Doenças dos Suínos/imunologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/metabolismo , Interferon Tipo I/metabolismo , Interferon Tipo I/genética , Imunidade Inata , Células HEK293 , Evasão da Resposta Imune , Ubiquitinação
19.
Genome Biol ; 25(1): 168, 2024 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-38926878

RESUMO

BACKGROUND: Carcinogenesis is driven by interactions between genetic mutations and the local tumor microenvironment. Recent research has identified hundreds of cancer driver genes; however, these studies often include a mixture of different molecular subtypes and ecological niches and ignore the impact of the immune system. RESULTS: In this study, we compare the landscape of driver genes in tumors that escaped the immune system (escape +) versus those that did not (escape -). We analyze 9896 primary tumors from The Cancer Genome Atlas using the ratio of non-synonymous to synonymous mutations (dN/dS) and find 85 driver genes, including 27 and 16 novel genes, in escape - and escape + tumors, respectively. The dN/dS of driver genes in immune escaped tumors is significantly lower and closer to neutrality than in non-escaped tumors, suggesting selection buffering in driver genes fueled by immune escape. Additionally, we find that immune evasion leads to more mutated sites, a diverse array of mutational signatures and is linked to tumor prognosis. CONCLUSIONS: Our findings highlight the need for improved patient stratification to identify new therapeutic targets for cancer treatment.


Assuntos
Mutação , Neoplasias , Evasão Tumoral , Humanos , Neoplasias/genética , Neoplasias/imunologia , Evasão Tumoral/genética , Evasão da Resposta Imune/genética , Evolução Molecular , Microambiente Tumoral/genética
20.
Int J Oral Sci ; 16(1): 47, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38945975

RESUMO

Dysregulated Epiregulin (EREG) can activate epidermal growth factor receptor (EGFR) and promote tumor progression in head and neck squamous cell carcinoma (HNSCC). However, the mechanisms underlying EREG dysregulation remain largely unknown. Here, we showed that dysregulated EREG was highly associated with enhanced PDL1 in HNSCC tissues. Treatment of HNSCC cells with EREG resulted in upregulated PDL1 via the c-myc pathway. Of note, we found that N-glycosylation of EREG was essential for its stability, membrane location, biological function, and upregulation of its downstream target PDL1 in HNSCC. EREG was glycosylated at N47 via STT3B glycosyltransferases, whereas mutations at N47 site abrogated N-glycosylation and destabilized EREG. Consistently, knockdown of STT3B suppressed glycosylated EREG and inhibited PDL1 in HNSCC cells. Moreover, treatment of HNSCC cells with NGI-1, an inhibitor of STT3B, blocked STT3B-mediated glycosylation of EREG, leading to its degradation and suppression of PDL1. Finally, combination of NGI-1 treatment with anti-PDLl therapy synergistically enhanced the efficacy of immunotherapy of HNSCC in vivo. Taken together, STT3B-mediated N-glycosylation is essential for stabilization of EREG, which mediates PDL1 upregulation and immune evasion in HNSCC.


Assuntos
Antígeno B7-H1 , Neoplasias de Cabeça e Pescoço , Carcinoma de Células Escamosas de Cabeça e Pescoço , Regulação para Cima , Animais , Humanos , Camundongos , Antígeno B7-H1/metabolismo , Western Blotting , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Epirregulina , Glicosilação , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/metabolismo , Evasão da Resposta Imune , Sialiltransferases/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/imunologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA