Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 7.266
1.
J Immunol ; 212(10): 1523-1529, 2024 May 15.
Article En | MEDLINE | ID: mdl-38709994

The study of S100A9 in viral infections has seen increased interest since the COVID-19 pandemic. S100A8/A9 levels were found to be correlated with the severity of COVID-19 disease, cytokine storm, and changes in myeloid cell subsets. These data led to the hypothesis that S100A8/A9 proteins might play an active role in COVID-19 pathogenesis. This review explores the structures and functions of S100A8/9 and the current knowledge on the involvement of S100A8/A9 and its constituents in viral infections. The potential roles of S100A9 in SARS-CoV-2 infections are also discussed.


COVID-19 , Calgranulin A , Calgranulin B , Inflammation , SARS-CoV-2 , Humans , COVID-19/immunology , SARS-CoV-2/immunology , Inflammation/immunology , Cytokine Release Syndrome/immunology , Virus Diseases/immunology
2.
Sci Immunol ; 9(95): eadq0015, 2024 May 03.
Article En | MEDLINE | ID: mdl-38701192

Initial imprinting by type 1 interferons shapes memory B cell generation in chronic viral infection.


B-Lymphocytes , Humans , Animals , B-Lymphocytes/immunology , Interferon Type I/immunology , Memory B Cells/immunology , Virus Diseases/immunology
4.
Clin Exp Med ; 24(1): 91, 2024 May 02.
Article En | MEDLINE | ID: mdl-38693436

The ubiquitous RNA-processing molecule TDP-43 is involved in neuromuscular diseases such as inclusion body myositis, a late-onset acquired inflammatory myopathy. TDP-43 solubility and function are disrupted in certain viral infections. Certain viruses, high viremia, co-infections, reactivation of latent viruses, and post-acute expansion of cytotoxic T cells may all contribute to inclusion body myositis, mainly in an age-shaped immune landscape. The virally induced senescent, interferon gamma-producing cytotoxic CD8+ T cells with increased inflammatory, and cytotoxic features are involved in the occurrence of inclusion body myositis in most such cases, in a genetically predisposed host. We discuss the putative mechanisms linking inclusion body myositis, TDP-43, and viral infections untangling the links between viruses, interferon, and neuromuscular degeneration could shed a light on the pathogenesis of the inclusion body myositis and other TDP-43-related neuromuscular diseases, with possible therapeutic implications.


DNA-Binding Proteins , Myositis, Inclusion Body , Virus Diseases , Myositis, Inclusion Body/virology , Humans , Virus Diseases/immunology , Virus Diseases/virology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
5.
Int J Mol Sci ; 25(8)2024 Apr 18.
Article En | MEDLINE | ID: mdl-38674036

CX3CL1, also named fractalkine or neurotactin, is the only known member of the CX3C chemokine family that can chemoattract several immune cells. CX3CL1 exists in both membrane-anchored and soluble forms, with each mediating distinct biological activities. CX3CL1 signals are transmitted through its unique receptor, CX3CR1, primarily expressed in the microglia of the central nervous system (CNS). In the CNS, CX3CL1 acts as a regulator of microglia activation in response to brain disorders or inflammation. Recently, there has been a growing interest in the role of CX3CL1 in regulating cell adhesion, chemotaxis, and host immune response in viral infection. Here, we provide a comprehensive review of the changes and function of CX3CL1 in various viral infections, such as human immunodeficiency virus (HIV), SARS-CoV-2, influenza virus, and cytomegalovirus (CMV) infection, to highlight the emerging roles of CX3CL1 in viral infection and associated diseases.


Chemokine CX3CL1 , Virus Diseases , Chemokine CX3CL1/metabolism , Humans , Virus Diseases/metabolism , Virus Diseases/immunology , Virus Diseases/virology , Animals , COVID-19/virology , COVID-19/metabolism , COVID-19/immunology , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , Microglia/metabolism , Microglia/virology , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics
6.
mBio ; 15(5): e0069224, 2024 May 08.
Article En | MEDLINE | ID: mdl-38567955

Defective viral genomes (DVGs) are truncated derivatives of their parental viral genomes generated during an aberrant round of viral genomic replication. Distinct classes of DVGs have been identified in most families of both positive- and negative-sense RNA viruses. Importantly, DVGs have been detected in clinical samples from virally infected individuals and an emerging body of association studies implicates DVGs in shaping the severity of disease caused by viral infections in humans. Consequently, there is growing interest in understanding the molecular mechanisms of de novo DVG generation, how DVGs interact with the innate immune system, and harnessing DVGs as novel therapeutics and vaccine adjuvants to attenuate viral pathogenesis. This minireview focuses on single-stranded RNA viruses (excluding retroviridae), and summarizes the current knowledge of DVG generation, the functions and diversity of DVG species, the roles DVGs play in influencing disease progression, and their application as antivirals and vaccine adjuvants.


Defective Viruses , Genome, Viral , Humans , Defective Viruses/genetics , Virus Replication , Animals , RNA Viruses/genetics , Immunity, Innate , Virus Diseases/virology , Virus Diseases/genetics , Virus Diseases/immunology
8.
Sci Data ; 11(1): 328, 2024 Apr 02.
Article En | MEDLINE | ID: mdl-38565538

Human infections caused by viral pathogens trigger a complex gamut of host responses that limit disease, resolve infection, generate immunity, and contribute to severe disease or death. Here, we present experimental methods and multi-omics data capture approaches representing the global host response to infection generated from 45 individual experiments involving human viruses from the Orthomyxoviridae, Filoviridae, Flaviviridae, and Coronaviridae families. Analogous experimental designs were implemented across human or mouse host model systems, longitudinal samples were collected over defined time courses, and global multi-omics data (transcriptomics, proteomics, metabolomics, and lipidomics) were acquired by microarray, RNA sequencing, or mass spectrometry analyses. For comparison, we have included transcriptomics datasets from cells treated with type I and type II human interferon. Raw multi-omics data and metadata were deposited in public repositories, and we provide a central location linking the raw data with experimental metadata and ready-to-use, quality-controlled, statistically processed multi-omics datasets not previously available in any public repository. This compendium of infection-induced host response data for reuse will be useful for those endeavouring to understand viral disease pathophysiology and network biology.


Multiomics , Virus Diseases , Viruses , Animals , Humans , Mice , Gene Expression Profiling/methods , Metabolomics , Proteomics/methods , Virus Diseases/immunology , Host-Pathogen Interactions
9.
J Med Virol ; 96(5): e29622, 2024 May.
Article En | MEDLINE | ID: mdl-38682614

RNA capping is an essential trigger for protein translation in eukaryotic cells. Many viruses have evolved various strategies for initiating the translation of viral genes and generating progeny virions in infected cells via synthesizing cap structure or stealing the RNA cap from nascent host messenger ribonucleotide acid (mRNA). In addition to protein translation, a new understanding of the role of the RNA cap in antiviral innate immunity has advanced the field of mRNA synthesis in vitro and therapeutic applications. Recent studies on these viral RNA capping systems have revealed startlingly diverse ways and molecular machinery. A comprehensive understanding of how viruses accomplish the RNA capping in infected cells is pivotal for designing effective broad-spectrum antiviral therapies. Here we systematically review the contemporary insights into the RNA-capping mechanisms employed by viruses causing human and animal infectious diseases, while also highlighting its impact on host antiviral innate immune response. The therapeutic applications of targeting RNA capping against viral infections and the development of RNA-capping inhibitors are also summarized.


Antiviral Agents , Immunity, Innate , RNA Caps , RNA, Viral , Virus Diseases , Humans , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , RNA, Viral/genetics , Animals , RNA Caps/metabolism , Virus Diseases/drug therapy , Virus Diseases/immunology , Virus Replication/drug effects , Viruses/genetics , Viruses/drug effects , Viruses/immunology
10.
Nutrients ; 16(8)2024 Apr 19.
Article En | MEDLINE | ID: mdl-38674902

Fermented foods have long been known to have immunomodulatory capabilities, and fermentates derived from the lactic acid bacteria of dairy products can modulate the immune system. We have used skimmed milk powder to generate novel fermentates using Lb. helveticus strains SC234 and SC232 and we demonstrate here that these fermentates can enhance key immune mechanisms that are critical to the immune response to viruses. We show that our novel fermentates, SC234 and SC232, can positively impact on cytokine and chemokine secretion, nitric oxide (NO) production, cell surface marker expression, and phagocytosis in macrophage models. We demonstrate that the fermentates SC234 and SC232 increase the secretion of cytokines IL-1ß, IL-6, TNF-α, IL-27, and IL-10; promote an M1 pro-inflammatory phenotype for viral immunity via NO induction; decrease chemokine expression of Monocyte Chemoattractant Protein (MCP); increase cell surface marker expression; and enhance phagocytosis in comparison to their starting material. These data suggest that these novel fermentates have potential as novel functional food ingredients for the treatment, management, and control of viral infection.


Cytokines , Fermentation , Nitric Oxide , Phagocytosis , Cytokines/metabolism , Animals , Nitric Oxide/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , RAW 264.7 Cells , Virus Diseases/immunology
11.
Viruses ; 16(4)2024 Apr 01.
Article En | MEDLINE | ID: mdl-38675895

Macrophages play multiple roles in innate immunity including phagocytosing pathogens, modulating the inflammatory response, presenting antigens, and recruiting other immune cells. Tissue-resident macrophages (TRMs) adapt to the local microenvironment and can exhibit different immune responses upon encountering distinct pathogens. In this study, we generated induced macrophages (iMACs) derived from human pluripotent stem cells (hPSCs) to investigate the interactions between the macrophages and various human pathogens, including the hepatitis C virus (HCV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and Streptococcus pneumoniae. iMACs can engulf all three pathogens. A comparison of the RNA-seq data of the iMACs encountering these pathogens revealed that the pathogens activated distinct gene networks related to viral response and inflammation in iMACs. Interestingly, in the presence of both HCV and host cells, iMACs upregulated different sets of genes involved in immune cell migration and chemotaxis. Finally, we constructed an image-based high-content analysis system consisting of iMACs, recombinant GFP-HCV, and hepatic cells to evaluate the effect of a chemical inhibitor on HCV infection. In summary, we developed a human cell-based in vitro model to study the macrophage response to human viral and bacterial infections; the results of the transcriptome analysis indicated that the iMACs were a useful resource for modeling pathogen-macrophage-tissue microenvironment interactions.


Hepacivirus , Macrophages , Pluripotent Stem Cells , SARS-CoV-2 , Humans , Macrophages/immunology , Macrophages/virology , Hepacivirus/immunology , Hepacivirus/physiology , SARS-CoV-2/immunology , Pluripotent Stem Cells/immunology , Streptococcus pneumoniae/immunology , COVID-19/immunology , COVID-19/virology , Hepatitis C/immunology , Hepatitis C/virology , Phagocytosis , Virus Diseases/immunology , Immunity, Innate
12.
Viruses ; 16(4)2024 Apr 11.
Article En | MEDLINE | ID: mdl-38675930

Inflammation is a protective host response essential for controlling viral replication and promoting tissue repair [...].


Inflammation , Virus Diseases , Inflammation/virology , Humans , Virus Diseases/immunology , Virus Diseases/virology , Animals , Viruses/immunology , Viruses/pathogenicity , Virus Replication , Host-Pathogen Interactions/immunology
13.
Viruses ; 16(4)2024 Apr 13.
Article En | MEDLINE | ID: mdl-38675942

The epitranscriptomic modification m6A is a prevalent RNA modification that plays a crucial role in the regulation of various aspects of RNA metabolism. It has been found to be involved in a wide range of physiological processes and disease states. Of particular interest is the role of m6A machinery and modifications in viral infections, serving as an evolutionary marker for distinguishing between self and non-self entities. In this review article, we present a comprehensive overview of the epitranscriptomic modification m6A and its implications for the interplay between viruses and their host, focusing on immune responses and viral replication. We outline future research directions that highlight the role of m6A in viral nucleic acid recognition, initiation of antiviral immune responses, and modulation of antiviral signaling pathways. Additionally, we discuss the potential of m6A as a prognostic biomarker and a target for therapeutic interventions in viral infections.


Immunity, Innate , Virus Diseases , Humans , Virus Diseases/immunology , Virus Diseases/virology , Methylation , Virus Replication , Viruses/immunology , Viruses/genetics , Animals , RNA, Viral/genetics , RNA, Viral/immunology , Signal Transduction , Host-Pathogen Interactions/immunology
14.
BMC Infect Dis ; 24(1): 431, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38654203

BACKGROUND: Vaccination is effective in preventing viral respiratory infectious diseases through protective antibodies and the gut microbiome has been proven to regulate human immunity. This study explores the causal correlations between gut microbial features and serum-specific antiviral immunoglobulin G (IgG) levels. METHODS: We conduct a two-sample bidirectional Mendelian randomization (MR) analysis using genome-wide association study (GWAS) summary data to explore the causal relationships between 412 gut microbial features and four antiviral IgG (for influenza A, measles, rubella, and mumps) levels. To make the results more reliable, we used four robust methods and performed comprehensive sensitivity analyses. RESULTS: The MR analyses revealed 26, 13, 20, and 18 causal associations of the gut microbial features influencing four IgG levels separately. ​Interestingly, ten microbial features, like genus Collinsella, species Bifidobacterium longum, and the biosynthesis of L-alanine have shown the capacity to regulate multiple IgG levels with consistent direction (rise or fall). The ​reverse MR analysis suggested several potential causal associations of IgG levels affecting microbial features. CONCLUSIONS: The human immune response against viral respiratory infectious diseases could be modulated by changing the abundance of gut microbes, which provided new approaches for the intervention of viral respiratory infections.


Gastrointestinal Microbiome , Immunoglobulin G , Mendelian Randomization Analysis , Respiratory Tract Infections , Humans , Immunoglobulin G/blood , Respiratory Tract Infections/immunology , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/microbiology , Genome-Wide Association Study , Antibodies, Viral/blood , Antibodies, Viral/immunology , Vaccination , Virus Diseases/immunology , Virus Diseases/prevention & control
15.
Cell Death Differ ; 31(3): 280-291, 2024 03.
Article En | MEDLINE | ID: mdl-38383887

Detection of cytosolic nucleic acids by pattern recognition receptors, including STING and RIG-I, leads to the activation of multiple signalling pathways that culminate in the production of type I interferons (IFNs) which are vital for host survival during virus infection. In addition to protective immune modulatory functions, type I IFNs are also associated with autoimmune diseases. Hence, it is important to elucidate the mechanisms that govern their expression. In this study, we identified a critical regulatory function of the DUSP4 phosphatase in innate immune signalling. We found that DUSP4 regulates the activation of TBK1 and ERK1/2 in a signalling complex containing DUSP4, TBK1, ERK1/2 and IRF3 to regulate the production of type I IFNs. Mice deficient in DUSP4 were more resistant to infections by both RNA and DNA viruses but more susceptible to malaria parasites. Therefore, our study establishes DUSP4 as a regulator of nucleic acid sensor signalling and sheds light on an important facet of the type I IFN regulatory system.


Interferon Type I , Membrane Proteins , Protein Tyrosine Phosphatases , Receptors, Cell Surface , Roundabout Proteins , Virus Diseases , Animals , Mice , Immunity, Innate , Interferon Type I/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Virus Diseases/immunology , Virus Diseases/metabolism , Membrane Proteins/metabolism , Roundabout Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Receptors, Cell Surface/metabolism
16.
J Virol ; 98(3): e0008824, 2024 Mar 19.
Article En | MEDLINE | ID: mdl-38386781

Type I and type II IFNs are important immune modulators in both innate and adaptive immunity. They transmit signaling by activating JAK-STAT pathways. Sirtuin 1 (SIRT1), a class III NAD+-dependent deacetylase, has multiple functions in a variety of physiological processes. Here, we characterized the novel functions of SIRT1 in the regulation of type I and type II IFN-induced signaling. Overexpression of SIRT1 inhibited type I and type II IFN-induced interferon-stimulated response element activation. In contrast, knockout of SIRT1 promoted type I and type II IFN-induced expression of ISGs and inhibited viral replication. Treatment with SIRT1 inhibitor EX527 had similar positive effects. SIRT1 physically associated with STAT1 or STAT3, and this interaction was enhanced by IFN stimulation or viral infection. By deacetylating STAT1 at K673 and STAT3 at K679/K685/K707/K709, SIRT1 downregulated the phosphorylation of STAT1 (Y701) and STAT3 (Y705). Sirt1+/- primary peritoneal macrophages and Sirt1+/- mice exhibited enhanced IFN-induced signaling and antiviral activity. Thus, SIRT1 is a novel negative regulator of type I and type II IFN-induced signaling through its deacetylase activity.IMPORTANCESIRT1 has been reported in the precise regulation of antiviral (RNA and DNA) immunity. However, its functions in type I and type II IFN-induced signaling are still unclear. In this study, we deciphered the important functions of SIRT1 in both type I and type II IFN-induced JAK-STAT signaling and explored the potential acting mechanisms. It is helpful for understanding the regulatory roles of SIRT1 at different levels of IFN signaling. It also consolidates the notion that SIRT1 is an important target for intervention in viral infection, inflammatory diseases, or even interferon-related therapies.


Interferon Type I , Sirtuin 1 , Virus Diseases , Animals , Mice , Immunity, Innate , Interferon Type I/metabolism , Interferon-gamma , Phosphorylation , Signal Transduction , Sirtuin 1/genetics , Sirtuin 1/metabolism , STAT1 Transcription Factor/metabolism , Virus Diseases/immunology
17.
SLAS Discov ; 29(3): 100140, 2024 Apr.
Article En | MEDLINE | ID: mdl-38182043

The use of recombinant antibodies developed through phage display technology offers a promising approach for combating viral infectious diseases. By specifically targeting antigens on viral surfaces, these antibodies have the potential to reduce the severity of infections or even prevent them altogether. With the emergence of new and more virulent strains of viruses, it is crucial to develop innovative methods to counteract them. Phage display technology has proven successful in generating recombinant antibodies capable of targeting specific viral antigens, thereby providing a powerful tool to fight viral infections. In this mini-review article, we examine the development of these antibodies using phage display technology, and discuss the associated challenges and opportunities in developing novel treatments for viral infectious diseases. Furthermore, we provide an overview of phage display technology. As these methods continue to evolve and improve, novel and sophisticated tools based on phage display and peptide display systems are constantly emerging, offering exciting prospects for solving scientific, medical, and technological problems related to viral infectious diseases in the near future.


Cell Surface Display Techniques , Recombinant Proteins , Virus Diseases , Humans , Virus Diseases/immunology , Virus Diseases/therapy , Cell Surface Display Techniques/methods , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Antibodies, Neutralizing/immunology , Peptide Library , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , Animals , Antigens, Viral/immunology , Antigens, Viral/genetics
18.
Nature ; 624(7992): 645-652, 2023 Dec.
Article En | MEDLINE | ID: mdl-38093014

People with diabetes feature a life-risking susceptibility to respiratory viral infection, including influenza and SARS-CoV-2 (ref. 1), whose mechanism remains unknown. In acquired and genetic mouse models of diabetes, induced with an acute pulmonary viral infection, we demonstrate that hyperglycaemia leads to impaired costimulatory molecule expression, antigen transport and T cell priming in distinct lung dendritic cell (DC) subsets, driving a defective antiviral adaptive immune response, delayed viral clearance and enhanced mortality. Mechanistically, hyperglycaemia induces an altered metabolic DC circuitry characterized by increased glucose-to-acetyl-CoA shunting and downstream histone acetylation, leading to global chromatin alterations. These, in turn, drive impaired expression of key DC effectors including central antigen presentation-related genes. Either glucose-lowering treatment or pharmacological modulation of histone acetylation rescues DC function and antiviral immunity. Collectively, we highlight a hyperglycaemia-driven metabolic-immune axis orchestrating DC dysfunction during pulmonary viral infection and identify metabolic checkpoints that may be therapeutically exploited in mitigating exacerbated disease in infected diabetics.


Dendritic Cells , Diabetes Complications , Diabetes Mellitus , Disease Susceptibility , Hyperglycemia , Lung , Virus Diseases , Animals , Mice , Acetyl Coenzyme A/metabolism , Acetylation , Chromatin/genetics , Chromatin/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Diabetes Complications/immunology , Diabetes Complications/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Glucose/metabolism , Histones/metabolism , Hyperglycemia/complications , Hyperglycemia/immunology , Hyperglycemia/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , T-Lymphocytes/immunology , Virus Diseases/complications , Virus Diseases/immunology , Virus Diseases/mortality , Viruses/immunology , Disease Models, Animal , Humans
19.
J Virol ; 97(12): e0150123, 2023 Dec 21.
Article En | MEDLINE | ID: mdl-37982618

IMPORTANCE: The type-I interferon (IFN-I) signaling pathway is the first line of antiviral innate immunity. It must be precisely regulated against virus-induced damage. The tightly regulated mechanisms of action of host genes in the antiviral innate immune signaling pathway are still worth studying. Here, we report a novel role of DLG1 in positively regulating the IκB kinase epsilon (IKKε)-mediated IFN-I signaling response against negative-stranded RNA virus replication, whereas the RNA virus inhibits the expression of DLG1 for immune escape. Importantly, the E3 ligase March2 interacts with and promotes K27-linked polyubiquitination of IKKε, and p62 is a cargo receptor that recognizes ubiquitinated IKKε for eventual autophagic degradation. Together, the current findings elucidate the role of DLG1 in the antiviral IFN-I signaling pathway and viral infection repression.


Autophagy , Discs Large Homolog 1 Protein , I-kappa B Kinase , Immunity, Innate , Negative-Sense RNA Viruses , Sequestosome-1 Protein , Virus Diseases , Humans , Discs Large Homolog 1 Protein/metabolism , I-kappa B Kinase/metabolism , Immunity, Innate/immunology , Negative-Sense RNA Viruses/growth & development , Negative-Sense RNA Viruses/immunology , Polyubiquitin/metabolism , Sequestosome-1 Protein/antagonists & inhibitors , Signal Transduction , Virus Diseases/immunology , Animals , Cell Line
20.
J Virol ; 97(11): e0088623, 2023 Nov 30.
Article En | MEDLINE | ID: mdl-37843373

IMPORTANCE: The expression of circVPS13D was upregulated with SCRV invasion, which proved that circVPS13D was involved in the regulation of the antiviral immune response. Our study revealed that the existence of circVPS13D promoted the replication of SCRV. Functionally, circVPS13D negatively regulates the antiviral responses of fish. Mechanistically, we confirmed that circVPS13D inhibited RLRs antiviral signaling pathway via the encoded protein VPS13D-170aa by targeting MAVS. Our study provided novel insights into the roles of protein-coding circRNAs and supported VPS13D-170aa as a negative regulator in the antiviral immune responses of teleost fish.


Fish Proteins , RNA, Circular , Vesicular Transport Proteins , Virus Diseases , Animals , Fish Proteins/genetics , Fishes/immunology , Fishes/virology , Immunity, Innate , RNA, Circular/metabolism , Signal Transduction , Vesicular Transport Proteins/genetics , Virus Diseases/immunology , Virus Diseases/metabolism , Virus Diseases/veterinary , Virus Diseases/virology , Fish Diseases/immunology , Fish Diseases/metabolism , Fish Diseases/virology
...