RESUMO
The innate immune system relies on a variety of pathogen recognition receptors (PRRs) as the first line of defense against pathogenic invasions. Viruses have evolved multiple strategies to evade the host immune system through coevolution with hosts. The CRISPR-Cas system is an adaptive immune system in bacteria or archaea that defends against viral reinvasion by targeting nucleic acids for cleavage. Based on the characteristics of Cas proteins and their variants, the CRISPR-Cas system has been developed into a versatile gene-editing tool capable of gene knockout or knock-in operations to achieve genetic variations in organisms. It is now widely used in the study of viral immune evasion mechanisms. This chapter will introduce the use of the CRISPR-Cas9 system for editing herpes simplex virus 1 (HSV-1) genes to explore the mechanisms by which HSV-1 evades host innate immunity and the experimental procedures involved.
Assuntos
Sistemas CRISPR-Cas , Técnicas de Inativação de Genes , Herpesvirus Humano 1 , Evasão da Resposta Imune , Imunidade Inata , Sistemas CRISPR-Cas/genética , Imunidade Inata/genética , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/genética , Evasão da Resposta Imune/genética , Humanos , Edição de Genes/métodos , Animais , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/genética , Herpes Simples/imunologia , Herpes Simples/virologia , Herpes Simples/genéticaRESUMO
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/imunologiaRESUMO
The persistent challenge posed by viruses that infect the central nervous system lies in their sophisticated ability to evade the host immune system. This review explores into the complex mechanisms of immune evasion employed by these neurotropic viruses, focussing on their modulation of host immune responses, evasion of adaptive immunity, and the cellular and molecular strategies that enable their persistence. Key areas explored include viral latency and reactivation, the inhibition of apoptosis, and antigenic variation, with a detailed examination of viral proteins and their interactions with host cellular processes.
Assuntos
Evasão da Resposta Imune , Humanos , Animais , Interações Hospedeiro-Patógeno/imunologia , Latência Viral/imunologia , Imunidade Adaptativa , Apoptose , Viroses do Sistema Nervoso Central/virologia , Viroses do Sistema Nervoso Central/imunologia , Ativação Viral/imunologia , Vírus/imunologia , Vírus/patogenicidade , Variação AntigênicaRESUMO
Introduction: Due to the widespread presence of susceptible populations, the pandemic caused by BA.5 subbranches swiftly disseminated China, impacting the majority of individuals within a span of 1 to 2 months. Subsequently, XBB and its subbranches became the dominant variants in China. Methods: We tracked the immune landscape in the population after the SARS-CoV-2 pandemic in late 2022 in China. Results: Our findings suggested that low levels of neutralizing antibodies against BA.5 subbranches before the pandemic might have contributed to the national outbreak at the end of 2022. The widespread breakthrough infections subsequently increased immunity to BA.5, XBB.1.5/1.9.1, and JN.1, inhibiting a new wave of large-scale infections caused by XBB subbranches in China. Additionally, JN.1 demonstrated enhanced immune evasion capabilities; however, Chinese residents had comparable levels of neutralizing antibodies against JN.1 as those observed for XBB.1.5 among confirmed cases at the end of 2022. Discussion: We anticipate that JN.1 will replace XBB subbranches as the predominant epidemic variant in subsequent transmissions within China. However, it is unlikely to cause a large-scale spread comparable to that witnessed at the end of 2022, with transmission patterns potentially resembling those observed for XBB post-pandemic.
Assuntos
Anticorpos Neutralizantes , COVID-19 , Evasão da Resposta Imune , SARS-CoV-2 , Humanos , China/epidemiologia , COVID-19/imunologia , COVID-19/epidemiologia , SARS-CoV-2/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Pandemias , MasculinoRESUMO
BACKGROUND: The high prevalence and detrimental effects on patient outcomes make gastric cancer (GC) a significant health issue that persists internationally. Existing treatment modalities exhibit limited efficacy, prompting the exploration of immune checkpoint inhibitors as a novel therapeutic approach. However, resistance to immunotherapy poses a significant challenge in GC management, necessitating a profound grasp of the intrinsic molecular pathways. METHODS: This study focuses on investigating the immunosuppressive mechanisms of quiescent cancer cells (QCCs) in GC, particularly their resistance to T-cell-mediated immune responses. Utilizing mouse models, gene editing techniques, and transcriptome sequencing, we aim to elucidate the interactions between QCCs, immune cells, and key regulatory factors like HIF1A. Functional enrichment analysis will further underscore the role of glycolysis-related genes in mediating immunosuppression by QCCs. RESULTS: The cancer cells that survived GC treated with T-cell therapy lost their proliferative ability. QCCs, as the main resistance force to immunotherapy, exhibit stronger resistance to CD8+ T-cell attack and possess higher cancer-initiating potential. Single-cell sequencing analysis revealed that the microenvironment in the QCCs region harbors more M2-type tumor-associated macrophages and fewer T cells. This microenvironment in the QCCs region leads to the downregulation of T-cell immune activation and alters macrophage metabolic function. Transcriptome sequencing of QCCs identified upregulated genes related to chemo-resistance, hypoxia, and glycolysis. In vitro cell experiments illustrated that HIF1A promotes the transcription of glycolysis-related genes, and silencing HIF1A in QCCs enhances T-cell proliferation and activation in co-culture systems, induces apoptosis in QCCs, and increases QCCs' sensitivity to immune checkpoint inhibitors. In vivo, animal experiments showed that silencing HIF1A in QCCs can inhibit GC growth and metastasis. CONCLUSION: Unraveling the molecular mechanisms by which QCCs resist T-cell-mediated immune responses through immunosuppression holds promising implications for refining treatment strategies and enhancing patient outcomes in GC. By delineating these intricate interactions, this study contributes crucial insights into precision medicine and improved therapeutic outcomes in GC management.
Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia , Neoplasias Gástricas , Microambiente Tumoral , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/genética , Animais , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Humanos , Microambiente Tumoral/imunologia , Linhagem Celular Tumoral , Imunoterapia/métodos , Glicólise/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Evasão Tumoral/efeitos dos fármacos , Evasão da Resposta Imune , Camundongos Endogâmicos C57BLRESUMO
Despite effective countermeasures, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) persists worldwide because of its ability to diversify and evade human immunity. This evasion stems from amino acid substitutions, particularly in the receptor binding domain (RBD) of the spike protein that confers resistance to vaccine-induced antibodies and antibody therapeutics. To constrain viral escape through resistance mutations, we combined antibody variable regions that recognize different RBD sites into multispecific antibodies. Here, we describe multispecific antibodies, including a trivalent trispecific antibody that potently neutralized diverse SARS-CoV-2 variants and prevented virus escape more effectively than single antibodies or mixtures of the parental antibodies. Despite being generated before the appearance of Omicron, this trispecific antibody neutralized all major Omicron variants through BA.4/BA.5 at nanomolar concentrations. Negative stain electron microscopy suggested that synergistic neutralization was achieved by engaging different epitopes in specific orientations that facilitated binding across more than one spike protein. Moreover, a tetravalent trispecific antibody containing the same variable regions as the trivalent trispecific antibody also protected Syrian hamsters against Omicron variants BA.1, BA.2, and BA.5 challenge, each of which uses different amino acid substitutions to mediate escape from therapeutic antibodies. These results demonstrated that multispecific antibodies have the potential to provide broad SARS-CoV-2 coverage, decrease the likelihood of escape, simplify treatment, and provide a strategy for antibody therapies that could help eliminate pandemic spread for this and other pathogens.
Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19 , Evasão da Resposta Imune , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , SARS-CoV-2/imunologia , Animais , COVID-19/imunologia , COVID-19/prevenção & controle , COVID-19/virologia , Humanos , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/uso terapêutico , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/química , Anticorpos Antivirais/imunologia , Camundongos , Epitopos/imunologia , Mesocricetus , Cricetinae , Anticorpos Biespecíficos/imunologia , Anticorpos Biespecíficos/farmacologiaRESUMO
Life-threatening systemic fungal infections caused by Candida albicans are significant contributors to clinical mortality, particularly among cancer patients and immunosuppressed individuals. The evasion of the immune response facilitated by fungal surface components enables fungal pathogens to evade macrophage attacks and to establish successful infections. This study developed a mesoporous silica nanoplatform, i.e., MSNP-EAP1Ab, which is composed of mesoporous silica nanoparticles grafted with the antibody of C. albicans surface adhesin Eap1. The activity of MSNP-EAP1Ab against C. albicans immune escape and infection was then evaluated by using the cell interaction model and the mouse systemic infection model. During interaction between C. albicans cells and macrophages, MSNP-EAP1Ab significantly inhibited fungal immune escape, leading to the enhanced phagocytosis of fungal cells by macrophages, with phagocytosis rates increasing from less than 8% to 14%. Furthermore, after treatment of the C. albicans-infected mice, MSNP-EAP1Ab drastically prolonged the mouse survival time and decreased the kidney fungal burden from >30,0000 CFU/g kidney to <100 CFU/g kidney, indicating the rapid recognition and killing of the pathogens by immune cells. Moreover, MSNP-EAP1Ab attenuated kidney tissue inflammation, with remarkable attenuation of renal immune cell accumulation. This study presents an innovative nanoplatform that targets the C. albicans adhesin, offering a promising approach for combatting systemic fungal infections.
Assuntos
Candida albicans , Candidíase , Nanopartículas , Dióxido de Silício , Animais , Nanopartículas/química , Dióxido de Silício/química , Camundongos , Candida albicans/imunologia , Candida albicans/efeitos dos fármacos , Candidíase/tratamento farmacológico , Candidíase/imunologia , Fagocitose/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Modelos Animais de Doenças , Anticorpos Antifúngicos/imunologia , Evasão da Resposta Imune , Proteínas Fúngicas/imunologia , Proteínas Fúngicas/químicaRESUMO
Varicella-zoster virus (VZV) encephalitis and meningitis are potential central nervous system (CNS) complications following primary VZV infection or reactivation. With Type-I interferon (IFN) signalling being an important first line cellular defence mechanism against VZV infection by the peripheral tissues, we here investigated the triggering of innate immune responses in a human neural-like environment. For this, we established and characterised 5-month matured hiPSC-derived neurospheroids (NSPHs) containing neurons and astrocytes. Subsequently, NSPHs were infected with reporter strains of VZV (VZVeGFP-ORF23) or Sendai virus (SeVeGFP), with the latter serving as an immune-activating positive control. Live cell and immunocytochemical analyses demonstrated VZVeGFP-ORF23 infection throughout the NSPHs, while SeVeGFP infection was limited to the outer NSPH border. Next, NanoString digital transcriptomics revealed that SeVeGFP-infected NSPHs activated a clear Type-I IFN response, while this was not the case in VZVeGFP-ORF23-infected NSPHs. Moreover, the latter displayed a strong suppression of genes related to IFN signalling and antigen presentation, as further demonstrated by suppression of IL-6 and CXCL10 production, failure to upregulate Type-I IFN activated anti-viral proteins (Mx1, IFIT2 and ISG15), as well as reduced expression of CD74, a key-protein in the MHC class II antigen presentation pathway. Finally, even though VZVeGFP-ORF23-infection seems to be immunologically ignored in NSPHs, its presence does result in the formation of stress granules upon long-term infection, as well as disruption of cellular integrity within the infected NSPHs. Concluding, in this study we demonstrate that 5-month matured hiPSC-derived NSPHs display functional innate immune reactivity towards SeV infection, and have the capacity to recapitulate the strong immune evasive behaviour towards VZV.
Assuntos
Herpesvirus Humano 3 , Células-Tronco Pluripotentes Induzidas , Humanos , Herpesvirus Humano 3/imunologia , Células-Tronco Pluripotentes Induzidas/imunologia , Células-Tronco Pluripotentes Induzidas/virologia , Imunidade Inata , Neurônios/imunologia , Neurônios/virologia , Infecção pelo Vírus da Varicela-Zoster/imunologia , Infecção pelo Vírus da Varicela-Zoster/virologia , Células Cultivadas , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Evasão da Resposta Imune , Citocinas/metabolismo , Citocinas/imunologia , Astrócitos/imunologia , Astrócitos/virologia , Astrócitos/metabolismo , Transdução de Sinais/imunologiaRESUMO
The continuous emergence of SARS-CoV-2 variants of concern has rendered many therapeutic monoclonal antibodies (mAbs) ineffective. To date, there are no clinically authorized therapeutic antibodies effective against the recently circulating Omicron sub-lineages BA.2.86 and JN.1. Here, we report the isolation of broad and potent neutralizing human mAbs (HuMabs) from a healthcare worker infected with SARS-CoV-2 early in the pandemic. These include a genetically unique HuMab, named K501SP6, which can neutralize different Omicron sub-lineages, including BQ.1, XBB.1, BA.2.86 and JN.1, by targeting a highly conserved epitope on the N terminal domain, as well as an RBD-specific HuMab (K501SP3) with high potency towards earlier circulating variants that was escaped by the more recent Omicron sub-lineages through spike F486 and E484 substitutions. Characterizing SARS-CoV-2 spike-specific HuMabs, including broadly reactive non-RBD-specific HuMabs, can give insight into the immune mechanisms involved in neutralization and immune evasion, which can be a valuable addition to already existing SARS-CoV-2 therapies.
Assuntos
Anticorpos Monoclonais , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , SARS-CoV-2/imunologia , Humanos , Glicoproteína da Espícula de Coronavírus/imunologia , COVID-19/imunologia , COVID-19/virologia , Anticorpos Antivirais/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/imunologia , Epitopos/imunologia , Evasão da Resposta Imune , Testes de NeutralizaçãoRESUMO
The emergence of Singapore grouper iridovirus (SGIV) has caused huge losses to grouper farming. SGIV is a DNA virus and belongs to the genus Ranavirus. Groupers infected with SGIV showed haemorrhaging and swelling of the spleen, with a mortality rate of more than 90% within a week. Therefore, it is of great significance to study the escape mechanism of SGIV from host innate immunity for the prevention and treatment of viral diseases in grouper. In this study, the viral proteins that interact with EccGAS were identified by mass spectrometry, and the SGIV VP12 protein that inhibits cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-mediated antiviral innate immunity was screened by the dual-luciferase reporter gene assay. VP12 belongs to the late gene of the virus. The immunofluorescence analysis demonstrated that VP12 was aggregated and distributed in the cytoplasm during the early stage of virus infection and translocated into the nucleus at the late stage of virus infection. VP12 inhibited the activation of IFN3, ISRE and NF-κB promoter activities mediated by cGAS-STING, EcTBK1 and EcIRF3. Quantitative real-time PCR analysis showed that VP12 inhibited the expression of interferon-related genes, including those mediated by cGAS-STING. VP12 enhanced the inhibition of IFN3, ISRE and NF-κB promoter activity by EccGAS, EccGAS-mab-21 and EccGAS-delete-mab21. The interaction between VP12 and EccGAS was found to be domain independent. The immunoprecipitation results demonstrated that VP12 interacted and co-localized with EccGAS, EcTBK1 and EcIRF3. VP12 degraded the protein levels of EcTBK1 and EcIRF3 and degraded EcIRF3 through the protease pathway. These results suggest that SGIV VP12 protein escapes the cGAS-STING signalling pathway and degrades EcIRF3 protein expression through the protease pathway.
Assuntos
Infecções por Vírus de DNA , Imunidade Inata , Proteínas de Membrana , Nucleotidiltransferases , Ranavirus , Transdução de Sinais , Animais , Ranavirus/imunologia , Ranavirus/fisiologia , Infecções por Vírus de DNA/imunologia , Infecções por Vírus de DNA/virologia , Infecções por Vírus de DNA/veterinária , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Proteínas Virais/metabolismo , Proteínas Virais/genética , Proteínas Virais/imunologia , Evasão da Resposta Imune , Interações Hospedeiro-Patógeno/imunologiaRESUMO
Mpox, the zoonotic disease caused by Monkeypox virus (MPXV), is currently a global health emergency. This review (Part I) aims to provide insights into the virus life cycle, epidemiology, host immune responses, and immune evasion mechanisms. Mpox symptoms is similar to smallpox but with lower mortality rates and lower transmissibility. In the past, the virus has been endemic in Central (Clade I) and West (Clade II) African countries. The first outbreak in outside Africa is reported in the United States in 2003. A multi-country outbreak across all continents occurred in 2022, predominantly driven by Clade II. Recently, the emergence of Clade Ib with sustained person-to-person transmission characteristic in the 2023-2024 outbreaks has raised significant public health concerns. Its apparent capacity for rapid spread and potential for causing severe disease highlight the need for enhanced surveillance, especially in regions not traditionally affected by Mpox. Immune responses induced by MPXV infection in humans and animal models provide the insights into the key step in which the host immune response recognizes and responds to the infection. The sophisticated immune evasion strategy by MPXV at both innate and adaptive arms also emerges that are useful for vaccine-based control measures. Taken together, understanding MPXV life cycle, epidemiology and immune response will facilitate better control, limit viral spread, and provide important insights for vaccine development.
Assuntos
Saúde Global , Mpox , Humanos , Animais , Mpox/imunologia , Mpox/epidemiologia , Mpox/prevenção & controle , Monkeypox virus/imunologia , Vacinas Virais/imunologia , Surtos de Doenças , Evasão da Resposta ImuneRESUMO
Immune evasion represents a crucial milestone in the progression of cancer and serves as the theoretical foundation for tumor immunotherapy. In this study, we reveal a negative association between Human Papillomavirus (HPV)-encoded circular RNA, circE7, and the infiltration of CD8+ T cells in head and neck squamous cell carcinoma (HNSCC). Both in vitro and in vivo experiments demonstrate that circE7 suppresses the function and activity of T cells by downregulating the transcription of LGALS9, which encodes the galectin-9 protein. The molecular mechanism involves circE7 binding to acetyl-CoA carboxylase 1 (ACC1), promoting its dephosphorylation and thereby activating ACC1. Activated ACC1 reduces H3K27 acetylation at the LGALS9 gene promoter, leading to decreased galectin-9 expression. Notably, galectin-9 interacts with immune checkpoint molecules TIM-3 and PD-1, inhibiting the secretion of cytotoxic cytokines by T cells and promoting T cell apoptosis. Here, we demonstrate a mechanism by which HPV promotes immune evasion in HNSCC through a circE7-driven epigenetic modification and propose a potential immunotherapy strategy for HNSCC that involves the combined use of anti-PD-1 and anti-TIM-3 inhibitors.
Assuntos
Linfócitos T CD8-Positivos , Galectinas , Neoplasias de Cabeça e Pescoço , Evasão da Resposta Imune , RNA Circular , Carcinoma de Células Escamosas de Cabeça e Pescoço , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/imunologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/virologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/virologia , Neoplasias de Cabeça e Pescoço/genética , Animais , Galectinas/genética , Galectinas/metabolismo , Galectinas/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , RNA Circular/genética , RNA Circular/metabolismo , RNA Circular/imunologia , Evasão da Resposta Imune/genética , Camundongos , Papillomaviridae/genética , Infecções por Papillomavirus/imunologia , Infecções por Papillomavirus/virologia , Infecções por Papillomavirus/genética , Regulação Neoplásica da Expressão Gênica , Evasão Tumoral/genética , Receptor Celular 2 do Vírus da Hepatite A/genética , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Masculino , Apoptose/genética , Feminino , Epigênese Genética , RNA Viral/genética , RNA Viral/imunologia , Papillomavirus HumanoRESUMO
Listeria monocytogenes (LM) possesses the ability to breach multiple barriers and elicit intricate immune responses. However, there remains a lack of explicit understanding regarding how LM evades innate immune surveillance within the body. Here, we utilized liver intravital imaging to elucidate the dynamic process of LM during infection in the liver. We discovered that LM can rapidly escape from Kupffer cells (KCs) through listeriolysin O (LLO) and proliferate within hepatocytes. Upon LM exposure to the hepatic sinusoids, neutrophils rapidly aggregate at the site of infection. Subsequently, LM can induce type I interferon (IFN-I) production primarily in the spleen, which acts systemically on neutrophils to hamper their swarming by deactivating the ERK pathway, thus evading neutrophil-mediated eradication. Furthermore, our findings suggest that virus-induced IFN-I suppresses neutrophil swarming, and COVID-19 patients exhibit impaired neutrophil aggregation function. In conclusion, our findings provide compelling evidence demonstrating that intracellular bacteria represented by LM can hijack host defense mechanisms against viral infections to evade immune surveillance. Additionally, impaired neutrophil swarming caused by IFN-I is one of the significant factors contributing to the increased susceptibility to bacterial infections following viral infections.
Assuntos
COVID-19 , Interferon Tipo I , Células de Kupffer , Listeria monocytogenes , Listeriose , Neutrófilos , Animais , Feminino , Humanos , Masculino , Camundongos , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/imunologia , COVID-19/imunologia , COVID-19/virologia , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Hepatócitos/virologia , Hepatócitos/imunologia , Evasão da Resposta Imune , Imunidade Inata , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Células de Kupffer/imunologia , Listeria monocytogenes/imunologia , Listeria monocytogenes/fisiologia , Listeriose/imunologia , Listeriose/microbiologia , Fígado/imunologia , Fígado/virologia , Fígado/microbiologia , Sistema de Sinalização das MAP Quinases/imunologia , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , Baço/imunologiaRESUMO
SARS-CoV-2 JN.1 with an additional L455S mutation on spike when compared with its parental variant BA.2.86 has outcompeted all earlier variants to become the dominant circulating variant. Recent studies investigated the immune resistance of SARS-CoV-2 JN.1 but additional factors are speculated to contribute to its global dominance, which remain elusive until today. Here, we find that SARS-CoV-2 JN.1 has a higher infectivity than BA.2.86 in differentiated primary human nasal epithelial cells (hNECs). Mechanistically, we demonstrate that the gained infectivity of SARS-CoV-2 JN.1 over BA.2.86 associates with increased entry efficiency conferred by L455S and better spike cleavage in hNECs. Structurally, S455 altered the mode of binding of JN.1 spike protein to ACE2 when compared to BA.2.86 spike at ACE2H34, and modified the internal structure of JN.1 spike protein by increasing the number of hydrogen bonds with neighboring residues. These findings indicate that a single mutation (L455S) enhances virus entry in hNECs and increases immune evasiveness, which contribute to the robust transmissibility of SARS-CoV-2 JN.1. We further evaluate the in vitro and in vivo virological characteristics between SARS-CoV-2 BA.2.86/JN.1 and EG.5.1/HK.3, and identify key lineage-specific features of the two Omicron sublineages that contribute to our understanding on Omicron antigenicity, transmissibility, and pathogenicity.
Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Evasão da Resposta Imune , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Humanos , SARS-CoV-2/genética , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/química , Evasão da Resposta Imune/genética , COVID-19/virologia , COVID-19/imunologia , Animais , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Internalização do Vírus , Mutação , Camundongos , Mucosa Nasal/virologia , Mucosa Nasal/imunologia , Células Epiteliais/virologia , Células Epiteliais/imunologia , Chlorocebus aethiops , Feminino , Células VeroRESUMO
The SARS-CoV-2 BA.2.86 lineage, and its sublineage JN.1 in particular, achieved widespread transmission in the US during winter 2023-24. However, this surge in infections was not accompanied by COVID-19 hospitalizations and mortality commensurate with prior waves. To understand shifts in COVID-19 epidemiology associated with JN.1 emergence, we compared characteristics and clinical outcomes of time-matched cases infected with BA.2.86 lineages (predominantly representing JN.1) versus co-circulating XBB-derived lineages in December, 2023 and January, 2024. Cases infected with BA.2.86 lineages received greater numbers of COVID-19 vaccine doses, including XBB.1.5-targeted boosters, in comparison to cases infected with XBB-derived lineages. Additionally, cases infected with BA.2.86 lineages experienced greater numbers of documented prior SARS-CoV-2 infections. Cases infected with BA.2.86 lineages also experienced lower risk of progression to severe clinical outcomes requiring emergency department consultations or hospital admission. Sensitivity analyses suggested under-ascertainment of prior infections could not explain this apparent attenuation of severity. Our findings implicate escape from immunity acquired from prior vaccination or infection in the emergence of the JN.1 lineage and suggest infections with this lineage are less likely to experience clinically-severe disease. Monitoring of immune escape and clinical severity in emerging SARS-CoV-2 variants remains a priority to inform responses.
Assuntos
Vacinas contra COVID-19 , COVID-19 , Evasão da Resposta Imune , SARS-CoV-2 , Humanos , COVID-19/imunologia , COVID-19/virologia , COVID-19/epidemiologia , SARS-CoV-2/imunologia , Masculino , Feminino , Pessoa de Meia-Idade , Vacinas contra COVID-19/imunologia , Adulto , Idoso , Índice de Gravidade de Doença , Hospitalização/estatística & dados numéricos , Estados Unidos/epidemiologiaRESUMO
The Staphylococcus aureus (S. aureus) SaeRS two-component system (TCS) regulates over 20 virulence factors. While its impact on chronic infection has been thoroughly discussed, its role in the early stage of infection remains elusive. Since macrophages serve as the primary immune defenders at the onset of infection, this study investigates the influence of SaeRS on macrophage functions and elucidates the underlying mechanisms. Macrophage expression of inflammatory and chemotactic factors, phagocytosis, and bactericidal activity against S. aureus were assessed, along with the evaluation of cellular oxidative stress. SaeRS was found to impair macrophage function. Mechanistically, SaeRS inhibited NF-κB pathway activation via toll-like receptor 2 (TLR2). Its immune-modulating effect could partially be explained by the strengthened biofilm formation. More importantly, we found SaeRS compromised macrophage immune functions at early infection stages even prior to biofilm formation. These early immune evasion effects were dependent on bacterial clumping as cytokine secretion, phagocytosis, and bactericidal activity were repaired when clumping was inhibited. We speculate that the bacterial clumping-mediated antigen mask is responsible for SaeRS-mediated immune evasion at the early infection stage. In vivo, ΔsaeRS infection was cleared earlier, accompanied by early pro-inflammatory cytokines production, and increased tissue oxidative stress. Subsequently, macrophages transitioned to an anti-inflammatory state, thereby promoting tissue repair. In summary, our findings underscore the critical role of the SaeRS TCS in S. aureus pathogenicity, particularly during early infection, which is likely initiated by SaeRS-mediated bacterial clumping.
Assuntos
Proteínas de Bactérias , Biofilmes , Evasão da Resposta Imune , Macrófagos , Fagocitose , Infecções Estafilocócicas , Staphylococcus aureus , Staphylococcus aureus/patogenicidade , Staphylococcus aureus/imunologia , Staphylococcus aureus/fisiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Animais , Camundongos , Biofilmes/crescimento & desenvolvimento , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , NF-kappa B/metabolismo , Receptor 2 Toll-Like/metabolismo , Fatores de Virulência/metabolismo , Fatores de Virulência/genética , Citocinas/metabolismo , Estresse Oxidativo , Fatores de TranscriçãoRESUMO
Herpes simplex virus type 2 (HSV-2) is a sexually transmitted virus, the cause of genital herpes, and its infection can increase the risk of HIV-1 infection. After initial infection, HSV-2 can establish lifelong latency within the nervous system, which is likely associated with the virus-mediated immune evasion. In this study, we found that HSV-2 UL24 significantly inhibited the activation of the IFN-ß promoter and the production of IFN-ß at both mRNA and protein levels. Of importance, the inhibitory effect of HSV-2 on IFN-ß production was significantly impaired in the context of HSV-2 infection when UL24 was knocked down. Additional studies revealed that, although the full-length HSV-2 UL24 affected cell cycle and viability to some extent, its N-terminal 1-202AA domain showed no obvious cytotoxicity while its C-terminal 201-281 AA domain had a minimal impact on cell viability. Further studies showed that the N-terminal 1-202 AA domain of HSV-2 UL24 (HSV-2 UL24-N) was the main functional region responsible for the inhibition of IFN-ß production mediated by HSV-2 UL24. This domain significantly suppressed the activity of RIG-IN, MAVS, TBK-1, IKK-ε, or the IRF-3/5D-activated IFN-ß promoter. Mechanistically, HSV-2 UL24-N suppressed IRF-3 phosphorylation, resulting in the inhibition of IFN-ß production. The findings of this study highlight the significance of HSV-2 UL24 in inhibiting IFN-ß production, revealing two potential roles of UL24 during HSV-2 infection: facilitating immune evasion and inducing cell cycle arrest.
Assuntos
Herpesvirus Humano 2 , Fator Regulador 3 de Interferon , Interferon beta , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/imunologia , Herpesvirus Humano 2/fisiologia , Interferon beta/metabolismo , Interferon beta/genética , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Humanos , Fosforilação , Proteínas Virais/metabolismo , Proteínas Virais/genética , Regiões Promotoras Genéticas , Evasão da Resposta Imune , Animais , Transdução de Sinais , Células HEK293 , Chlorocebus aethiops , Linhagem Celular , Interações Hospedeiro-Patógeno , Células VeroRESUMO
The complement system is a vital anti-microbial defence mechanism against circulating pathogens. Excessive complement activation can have deleterious outcomes for the host and is consequently tightly modulated by a set of membrane-associated and fluid-phase regulators of complement activation (RCAs). Here, we demonstrate that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) hijacks host cellular RCA members CD55 and CD59 and serum-derived Factor H (FH) to resist antibody-dependent complement-mediated lysis triggered by immunized human sera. Blockage of the biological functions of virion-associated CD55 and CD59 and competition of FH recruitment with functionally inactive recombinant FH-derived short consensus repeats SCR18-20 restore SARS-CoV-2 complement sensitivity in a synergistic manner. Moreover, complement-mediated virolysis is dependent on classical pathway activation and does not occur in the absence of virus-specific antibodies. Altogether, our findings present an intriguing immune escape mechanism that provides novel insights into the immunopathology observed in severe coronavirus disease 2019 (COVID-19).
Assuntos
Antígenos CD55 , Antígenos CD59 , COVID-19 , Ativação do Complemento , Fator H do Complemento , SARS-CoV-2 , Humanos , SARS-CoV-2/imunologia , Antígenos CD59/imunologia , Antígenos CD55/imunologia , Antígenos CD55/metabolismo , COVID-19/imunologia , COVID-19/virologia , Fator H do Complemento/imunologia , Fator H do Complemento/metabolismo , Anticorpos Antivirais/imunologia , Evasão da Resposta ImuneRESUMO
Aerobic glycolysis and immune evasion are two key hallmarks of cancer. However, how these two features are mechanistically linked to promote tumor growth is not well understood. Here, we show that the glycolytic enzyme enolase-1 (ENO1) is dynamically modified with an O-linked ß-N-acetylglucosamine (O-GlcNAcylation), and simultaneously regulates aerobic glycolysis and immune evasion via differential glycosylation. Glycosylation of threonine 19 (T19) on ENO1 promotes its glycolytic activity via the formation of active dimers. On the other hand, glycosylation of serine 249 (S249) on ENO1 inhibits its interaction with PD-L1, decreases association of PD-L1 with the E3 ligase STUB1, resulting in stabilization of PD-L1. Consequently, blockade of T19 glycosylation on ENO1 inhibits glycolysis, and decreases cell proliferation and tumor growth. Blockade of S249 glycosylation on ENO1 reduces PD-L1 expression and enhances T cell-mediated immunity against tumor cells. Notably, elimination of glycosylation at both sites synergizes with PD-L1 monoclonal antibody therapy to promote antitumor immune response. Clinically, ENO1 glycosylation levels are up-regulated and show a positive correlation with PD-L1 levels in human colorectal cancers. Thus, our findings provide a mechanistic understanding of how O-GlcNAcylation bridges aerobic glycolysis and immune evasion to promote tumor growth, suggesting effective therapeutic opportunities.
Assuntos
Antígeno B7-H1 , Neoplasias Colorretais , Proteínas de Ligação a DNA , Glicólise , Fosfopiruvato Hidratase , Proteínas Supressoras de Tumor , Fosfopiruvato Hidratase/metabolismo , Fosfopiruvato Hidratase/imunologia , Fosfopiruvato Hidratase/genética , Humanos , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Glicosilação , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Camundongos , Antígeno B7-H1/metabolismo , Animais , Evasão da Resposta Imune , Linhagem Celular Tumoral , Acetilglucosamina/metabolismo , Proliferação de Células , Aerobiose , Evasão Tumoral , Biomarcadores TumoraisRESUMO
Despite advances in vaccination and therapies for coronavirus disease, challenges remain due to reduced antibody longevity and the emergence of virulent variants like Omicron (BA.1) and its subvariants (BA.1.1, BA.2, BA.3, and BA.5). This study explored the potential of adoptive immunotherapy and harnessing the protective abilities using virus-specific T cells (VSTs). Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) VSTs were generated by stimulating donor-derived peripheral blood mononuclear cells with spike, nucleocapsid, and membrane protein peptide mixtures. Phenotypic characterization, including T-cell receptor (TCR) vß and pentamer analyses, was performed on the ex vivo-expanded cells. We infected human leukocyte antigen (HLA)-partially matched human Calu-3 cells with various authentic SARS-CoV-2 strains in a Biosafety Level 3 facility and co-cultured them with VSTs. VSTs exhibited a diverse TCR vß repertoire, confirming their ability to target a broad range of SARS-CoV-2 antigens from both the ancestral and mutant strains, including Omicron BA.1 and BA.5. These ex vivo-expanded cells exhibited robust cytotoxicity and low alloreactivity against HLA-partially matched SARS-CoV-2-infected cells. Their cytotoxic effects were consistent across variants, targeting conserved spike and nucleocapsid epitopes. Our findings suggest that third-party partial HLA-matching VSTs could counter immune-escape mechanisms posed by emerging variants of concern.