Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.360
Filter
1.
Methods Mol Biol ; 2854: 1-7, 2025.
Article in English | MEDLINE | ID: mdl-39192112

ABSTRACT

Antiviral innate immunity is a complicated system initiated by the induction of type I interferon (IFN-I) and downstream interferon-stimulated genes (ISGs) and is finely regulated by numerous positive and negative factors at different signaling adaptors. During this process, posttranslational modifications, especially ubiquitination, are the most common regulatory strategy used by the host to switch the antiviral innate signaling pathway and are mainly controlled by E3 ubiquitin ligases from different protein families. A comprehensive understanding of the regulatory mechanisms and a novel discovery of regulatory factors involved in the IFN-I signaling pathway are important for researchers to identify novel therapeutic targets against viral infectious diseases based on innate immunotherapy. In this section, we use the E3 ubiquitin ligase as an example to guide the identification of a protein belonging to the RING Finger (RNF) family that regulates the RIG-I-mediated IFN-I pathway through ubiquitination.


Subject(s)
Immunity, Innate , Interferon Type I , Signal Transduction , Ubiquitin-Protein Ligases , Ubiquitination , Humans , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Interferon Type I/metabolism , Virus Diseases/immunology , Virus Diseases/genetics , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/genetics
2.
Methods Mol Biol ; 2854: 9-18, 2025.
Article in English | MEDLINE | ID: mdl-39192113

ABSTRACT

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.


Subject(s)
Immunity, Innate , Humans , Viral Proteins/immunology , Viral Proteins/genetics , Viruses/immunology , Viruses/genetics , Immune Evasion , Virus Diseases/immunology , Virus Diseases/virology , Animals , Genes, Viral , Autophagy/immunology , Host-Pathogen Interactions/immunology , Signal Transduction/immunology
3.
Methods Mol Biol ; 2854: 83-91, 2025.
Article in English | MEDLINE | ID: mdl-39192121

ABSTRACT

Transcriptomics is an extremely important area of molecular biology and is a powerful tool for studying all RNA molecules in an organism. Conventional transcriptomic technologies include microarrays and RNA sequencing, and the rapid development of single-cell sequencing and spatial transcriptomics in recent years has provided an enormous scope for research in this field. This chapter describes the application, significance, and experimental procedures of a variety of transcriptomic technologies in antiviral natural immunity.


Subject(s)
Gene Expression Profiling , Immunity, Innate , Transcriptome , Immunity, Innate/genetics , Humans , Gene Expression Profiling/methods , Animals , Virus Diseases/immunology , Virus Diseases/genetics , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Oligonucleotide Array Sequence Analysis/methods
4.
Methods Mol Biol ; 2854: 61-74, 2025.
Article in English | MEDLINE | ID: mdl-39192119

ABSTRACT

With the rapid development of CRISPR-Cas9 technology, gene editing has become a powerful tool for studying gene function. Specifically, in the study of the mechanisms by which natural immune responses combat viral infections, gene knockout mouse models have provided an indispensable platform. This article describes a detailed protocol for constructing gene knockout mice using the CRISPR-Cas9 system. This field focuses on the design of single-guide RNAs (sgRNAs) targeting the antiviral immune gene cGAS, embryo microinjection, and screening and verification of gene editing outcomes. Furthermore, this study provides methods for using cGAS gene knockout mice to analyze the role of specific genes in natural immune responses. Through this protocol, researchers can efficiently generate specific gene knockout mouse models, which not only helps in understanding the functions of the immune system but also offers a powerful experimental tool for exploring the mechanisms of antiviral innate immunity.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Immunity, Innate , Mice, Knockout , RNA, Guide, CRISPR-Cas Systems , Animals , Immunity, Innate/genetics , Mice , RNA, Guide, CRISPR-Cas Systems/genetics , Gene Editing/methods , Gene Knockout Techniques/methods , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Virus Diseases/immunology , Virus Diseases/genetics
5.
Methods Mol Biol ; 2854: 199-212, 2025.
Article in English | MEDLINE | ID: mdl-39192131

ABSTRACT

Antiviral innate immunity plays a critical role in the defense against viral infections, yet its complex interactions with viruses have been challenging to study using traditional models. Organoids, three-dimensional (3D) tissue-like structures derived from stem cells, have emerged as powerful tools for modeling human tissues and studying the complex interactions between viruses and the host innate immune system. This chapter summarizes relevant applications of organoids in antiviral innate immunity studies and provides detailed information and experimental procedures for using organoids to study antiviral innate immunity.


Subject(s)
Immunity, Innate , Organoids , Virus Diseases , Organoids/immunology , Organoids/virology , Humans , Virus Diseases/immunology , Virus Diseases/virology , Animals , Host-Pathogen Interactions/immunology , Viruses/immunology
6.
Methods Mol Biol ; 2854: 221-236, 2025.
Article in English | MEDLINE | ID: mdl-39192133

ABSTRACT

Zebrafish is a widely used model organism in genetics, developmental biology, pathology, and immunology research. Due to their fast reproduction, large numbers, transparent early embryos, and high genetic conservation with the human genome, zebrafish have been used as a model for studying human and fish viral diseases. In particular, the ability to easily perform forward and reverse genetics and lacking a functional adaptive immune response during the early period of development establish the zebrafish as a favored option to assess the functional implication of specific genes in the antiviral innate immune response and the pathogenesis of viral diseases. In this chapter, we detail protocols for the antiviral innate immunity analysis using the zebrafish model, including the generation of gene-overexpression zebrafish, generation of gene-knockout zebrafish by clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 technology, methods of viral infection in zebrafish larvae, analyzing the expression of antiviral genes in zebrafish larvae using qRT-PCR, Western blotting and transcriptome sequencing, and in vivo antiviral assays. These experimental protocols provide effective references for studying the antiviral immune response in the zebrafish model.


Subject(s)
CRISPR-Cas Systems , Disease Models, Animal , Immunity, Innate , Zebrafish , Animals , Zebrafish/immunology , Zebrafish/genetics , Zebrafish/virology , Immunity, Innate/genetics , Virus Diseases/immunology , Virus Diseases/genetics , Gene Knockout Techniques , Animals, Genetically Modified
7.
Methods Mol Biol ; 2854: 253-264, 2025.
Article in English | MEDLINE | ID: mdl-39192135

ABSTRACT

Innate immunity is an important defense barrier for the human body. After viral pathogen-associated molecular patterns (PAMPs) are detected by host-pathogen recognition receptors (PRRs), the associated signaling pathways trigger the activation of the interferon (IFN) regulatory factor (IRF) family members and the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). However, any gene defects among the signaling adaptors will compromise innate immune efficiency. Therefore, investigating genetic defects in the antiviral innate immune signaling pathway is important. We summarize the commonly used research methods related to antiviral immune gene defects and outline the relevant research protocols, which will help investigators study antiviral innate immunity.


Subject(s)
Immunity, Innate , Signal Transduction , Humans , Animals , Virus Diseases/immunology , Virus Diseases/genetics , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , NF-kappa B/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Receptors, Pattern Recognition/metabolism , Receptors, Pattern Recognition/genetics
8.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273355

ABSTRACT

Viruses utilize host cells at all stages of their life cycle, from the transcription of genes and translation of viral proteins to the release of viral copies. The human immune system counteracts viruses through a variety of complex mechanisms, including both innate and adaptive components. Viruses have an ability to evade different components of the immune system and affect them, leading to disruption. This review covers contemporary knowledge about the virus-induced complex interplay of molecular interactions, including regulation of transcription and translation in host cells resulting in the modulation of immune system functions. Thorough investigation of molecular mechanisms and signaling pathways that are involved in modulating of host immune response to viral infections can help to develop novel approaches for antiviral therapy. In this review, we consider new therapeutic approaches for antiviral treatment. Modern therapeutic strategies for the treatment and cure of human immunodeficiency virus (HIV) are considered in detail because HIV is a unique example of a virus that leads to host T lymphocyte deregulation and significant modulation of the host immune response. Furthermore, peculiarities of some promising novel agents for the treatment of various viral infections are described.


Subject(s)
Antiviral Agents , Humans , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Virus Diseases/drug therapy , Virus Diseases/immunology , Virus Diseases/virology , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/drug effects , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , Immunity, Innate/drug effects , Animals , Viruses/drug effects , Viruses/immunology
9.
Rev Med Virol ; 34(5): e2583, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39289528

ABSTRACT

Autoantibodies are immune system-produced antibodies that wrongly target the body's cells and tissues for attack. The COVID-19 pandemic has made it possible to link autoantibodies to both the severity of pathogenic infection and the emergence of several autoimmune diseases after recovery from the infection. An overview of autoimmune disorders and the function of autoantibodies in COVID-19 and other infectious diseases are discussed in this review article. We also investigated the different categories of autoantibodies found in COVID-19 and other infectious diseases including the potential pathways by which they contribute to the severity of the illness. Additionally, it also highlights the probable connection between vaccine-induced autoantibodies and their adverse outcomes. The review also discusses the therapeutic perspectives of autoantibodies. This paper advances our knowledge about the intricate interaction between autoantibodies and COVID-19 by thoroughly assessing the most recent findings.


Subject(s)
Autoantibodies , Autoimmune Diseases , COVID-19 , SARS-CoV-2 , Humans , COVID-19/immunology , COVID-19/virology , Autoantibodies/immunology , SARS-CoV-2/immunology , Autoimmune Diseases/immunology , Virus Diseases/immunology
10.
Int J Mol Sci ; 25(17)2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39273156

ABSTRACT

Mitochondria play pivotal roles in sustaining various biological functions including energy metabolism, cellular signaling transduction, and innate immune responses. Viruses exploit cellular metabolic synthesis to facilitate viral replication, potentially disrupting mitochondrial functions and subsequently eliciting a cascade of proinflammatory responses in host cells. Additionally, the disruption of mitochondrial membranes is involved in immune regulation. During viral infections, mitochondria orchestrate innate immune responses through the generation of reactive oxygen species (ROS) and the release of mitochondrial DNA, which serves as an effective defense mechanism against virus invasion. The targeting of mitochondrial damage may represent a novel approach to antiviral intervention. This review summarizes the regulatory mechanism underlying proinflammatory response induced by mitochondrial damage during viral infections, providing new insights for antiviral strategies.


Subject(s)
Immunity, Innate , Mitochondria , Reactive Oxygen Species , Virus Diseases , Humans , Mitochondria/metabolism , Virus Diseases/immunology , Virus Diseases/metabolism , Reactive Oxygen Species/metabolism , Animals , Inflammation/metabolism , Inflammation/immunology , DNA, Mitochondrial/metabolism , Signal Transduction
11.
J Transl Med ; 22(1): 847, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294611

ABSTRACT

SCOPE: The underlying medical conditions and gut dysbiosis is known to influence COVID-19 severity in high-risk patients. The current review proposed the optimal usage of nutraceuticals & pharmacological interventions can help regulate the protective immune response and balance the regulatory functionality of gut microbiota. Many studies have revealed that the probiotic interventions viz., Lactobacillus rhamnosus, L. plantarum & other bacterial spp. reduce IFNγ & TNF-α and increase IL-4 & IL-10 secretions to control the immunostimulatory effects in upper respiratory tract infection. Dietary fibres utilized by beneficial microbiota and microbial metabolites can control the NF-kB regulation. Vitamin C halts the propagation of pathogens and vitamin D and A modulate the GM. Selenium and Flavonoids also control the redox regulations. Interferon therapy can antagonize the viral replications, while corticosteroids may reduce the death rates. BCG vaccine reprograms the monocytes to build trained immunity. Bifidobacterium and related microbes were found to increase the vaccine efficacy. Vaccines against COVID-19 and season flu also boost the immunity profile for robust protection. Over all, the collective effects of these therapeutics could help increase the opportunities for faster recovery from infectious diseases. CONCLUSION: The nutraceutical supplements and pharmacological medicines mediate the modulatory functionalities among beneficial microbes of gut, which in turn eliminate pathogens, harmonize the activity of immune cells to secrete essential regulatory molecular receptors and adaptor proteins establishing the homeostasis in the body organs through essential microbiome. Therefore, the implementation of this methodology could control the severity events during clinical sickness and reduce the mortalities.


Subject(s)
COVID-19 , Dietary Supplements , Gastrointestinal Microbiome , SARS-CoV-2 , Humans , COVID-19/immunology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , SARS-CoV-2/immunology , COVID-19 Drug Treatment , Probiotics/therapeutic use , Virus Diseases/immunology , Virus Diseases/drug therapy , Immunity/drug effects
12.
Immunity ; 57(8): 1721-1723, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39142270

ABSTRACT

The interferon-stimulated gene OAS1 has well-defined antiviral properties. In two recent issues of Immunity, Harioudh et al. describe a non-canonical function of OAS1 that selectively protects the translation of proteins involved in defense against viral or bacterial infections.


Subject(s)
2',5'-Oligoadenylate Synthetase , Bacterial Infections , Virus Diseases , 2',5'-Oligoadenylate Synthetase/metabolism , 2',5'-Oligoadenylate Synthetase/genetics , Bacterial Infections/immunology , Humans , Virus Diseases/immunology , Animals , Mice
13.
Sci Prog ; 107(3): 368504241265182, 2024.
Article in English | MEDLINE | ID: mdl-39091074

ABSTRACT

Insights into mechanisms driving either activation or inhibition of immune response are crucial in understanding the pathology of various diseases. The differentiation of viral from endogenous RNA in the cytoplasm by pattern-recognition receptors, such as retinoic acid-inducible gene I (RIG-I), is one of the essential paths for timely activation of an antiviral immune response through induction of type I interferons (IFN). In this mini-review, we describe the most recent developments centered around RIG-I's structure and mechanism of action. We summarize the paradigm-changing work over the past few years that helped us better understand RIG-I's monomeric and oligomerization states and their role in conveying immune response. We also discuss potential applications of the modulation of the RIG-I pathway in preventing autoimmune diseases or induction of immunity against viral infections. Overall, our review aims to summarize innovative research published in the past few years to help clarify questions that have long persisted around RIG-I.


Subject(s)
DEAD Box Protein 58 , Receptors, Immunologic , Humans , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/immunology , DEAD Box Protein 58/genetics , DEAD Box Protein 58/chemistry , Receptors, Immunologic/chemistry , Receptors, Immunologic/metabolism , Animals , Virus Diseases/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Signal Transduction , Protein Multimerization , Immunity, Innate
14.
Front Immunol ; 15: 1397780, 2024.
Article in English | MEDLINE | ID: mdl-39100679

ABSTRACT

Enhancing livestock biosecurity is critical to safeguard the livelihoods of farmers, global and local economies, and food security. Vaccination is fundamental to the control and prevention of exotic and endemic high-priority infectious livestock diseases. Successful implementation of vaccination in a biosecurity plan is underpinned by a strong understanding of correlates of protection-those elements of the immune response that can reliably predict the level of protection from viral challenge. While correlates of protection have been successfully characterized for many human viral vaccines, for many high-priority livestock viral diseases, including African swine fever and foot and mouth disease, they remain largely uncharacterized. Current literature provides insights into potential correlates of protection that should be assessed during vaccine development for these high-priority mammalian livestock viral diseases. Establishment of correlates of protection for biosecurity purposes enables immune surveillance, rationale for vaccine development, and successful implementation of livestock vaccines as part of a biosecurity strategy.


Subject(s)
Livestock , Vaccination , Viral Vaccines , Animals , Livestock/immunology , Livestock/virology , Viral Vaccines/immunology , Vaccination/veterinary , Virus Diseases/prevention & control , Virus Diseases/immunology , Virus Diseases/veterinary , Swine , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease/immunology , African Swine Fever/prevention & control , African Swine Fever/immunology , Humans
15.
Front Public Health ; 12: 1367644, 2024.
Article in English | MEDLINE | ID: mdl-39104887

ABSTRACT

Introduction: Persistent infections caused by certain viruses and parasites have been associated with multiple diseases and substantial mortality. Heavy metals are ubiquitous environmental pollutants with immunosuppressive properties. This study aimed to determine whether heavy metals exposure suppress the immune system, thereby increasing the susceptibility to persistent infections. Methods: Using data from NHANES 1999-2016, we explored the associations between heavy metals exposure and persistent infections: Cytomegalovirus (CMV), Epstein-Barr Virus (EBV), Hepatitis C Virus (HCV), Herpes Simplex Virus Type-1 (HSV-1), Toxoplasma gondii (T. gondii), and Toxocara canis and Toxocara cati (Toxocara spp.) by performing logistic regression, weighted quantile sum (WQS) and Bayesian kernel machine regression (BKMR) models. Mediation analysis was used to determine the mediating role of host immune function in these associations. Results: Logistic regression analysis revealed positive associations between multiple heavy metals and the increased risk of persistent infections. In WQS models, the heavy metals mixture was associated with increased risks of several persistent infections: CMV (OR: 1.58; 95% CI: 1.17, 2.14), HCV (OR: 2.94; 95% CI: 1.68, 5.16), HSV-1 (OR: 1.25; 95% CI: 1.11, 1.42), T. gondii (OR: 1.97; 95% CI: 1.41, 2.76), and Toxocara spp. (OR: 1.76; 95% CI: 1.16, 2.66). BKMR models further confirmed the combined effects of heavy metals mixture and also identified the individual effect of arsenic, cadmium, and lead. On mediation analysis, the systemic immune inflammation index, which reflects the host's immune status, mediated 12.14% of the association of mixed heavy metals exposure with HSV-1 infection. Discussion: The findings of this study revealed that heavy metals exposure may increase susceptibility to persistent infections, with the host's immune status potentially mediating this relationship. Reducing exposure to heavy metals may have preventive implications for persistent infections, and further prospective studies are needed to confirm these findings.


Subject(s)
Environmental Exposure , Metals, Heavy , Humans , Female , Male , Environmental Exposure/adverse effects , Adult , Middle Aged , Logistic Models , Environmental Pollutants/toxicity , Bayes Theorem , Virus Diseases/immunology , Animals
16.
Curr Opin Immunol ; 89: 102445, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39137494

ABSTRACT

All organisms must defend themselves against viral predators. This includes bacteria, which harbor immunity factors such as restriction-modification systems and CRISPR-Cas systems. More recently, a plethora of additional defense systems have been identified, revealing a richer, more sophisticated immune system than previously appreciated. Some of these newly identified defense systems have distant homologs in mammals, suggesting an ancient evolutionary origin of some facets of mammalian immunity. An even broader conservation exists at the level of how these immunity systems operate. Here, we focus at this level, reviewing key principles and high-level attributes of innate immunity in bacteria that are shared with mammalian immunity, while also noting key differences, with a particular emphasis on how cells sense viral infection.


Subject(s)
Bacteria , Immunity, Innate , Viruses , Animals , Humans , Bacteria/immunology , Viruses/immunology , Virus Diseases/immunology , Host-Pathogen Interactions/immunology , Bacterial Infections/immunology , CRISPR-Cas Systems
17.
Nutrients ; 16(15)2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39125326

ABSTRACT

Viral infections pose significant global challenges due to their rapid transmissibility. Therefore, preventing and treating these infections promptly is crucial to curbing their spread. This review focuses on the vital link between nutrition and viral infections, underscoring how dietary factors influence immune system modulation. Malnutrition, characterized by deficiencies in essential nutrients such as vitamins A, C, D, E, and zinc, can impair the immune system, thereby increasing vulnerability to viral infections and potentially leading to more severe health outcomes that complicate recovery. Additionally, emerging evidence highlights the role of commensal microbiota in immune regulation, which can affect hosts' susceptibility to infections. Specific dietary components, including bioactive compounds, vitamins, and probiotics, can beneficially modify gut microbiota, thus enhancing immune response and offering protection against viral infections. This review aims to elucidate the mechanisms by which dietary adjustments and gut microbiota impact the pathogenesis of viral infections, with a particular focus on strengthening the immune system.


Subject(s)
Gastrointestinal Microbiome , Nutritional Status , Probiotics , Virus Diseases , Humans , Virus Diseases/prevention & control , Virus Diseases/immunology , Diet , Vitamins , Malnutrition/prevention & control , Immune System
18.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125870

ABSTRACT

Immune-mediated gastrointestinal (GI) diseases, including achalasia, celiac disease, and inflammatory bowel diseases, pose significant challenges in diagnosis and management due to their complex etiology and diverse clinical manifestations. While genetic predispositions and environmental factors have been extensively studied in the context of these conditions, the role of viral infections and virome dysbiosis remains a subject of growing interest. This review aims to elucidate the involvement of viral infections in the pathogenesis of immune-mediated GI diseases, focusing on achalasia and celiac disease, as well as the virome dysbiosis in IBD. Recent evidence suggests that viral pathogens, ranging from common respiratory viruses to enteroviruses and herpesviruses, may trigger or exacerbate achalasia and celiac disease by disrupting immune homeostasis in the GI tract. Furthermore, alterations in the microbiota and, specifically, in the virome composition and viral-host interactions have been implicated in perpetuating chronic intestinal inflammation in IBD. By synthesizing current knowledge on viral contributions to immune-mediated GI diseases, this review aims to provide insights into the complex interplay between viral infections, host genetics, and virome dysbiosis, shedding light on novel therapeutic strategies aimed at mitigating the burden of these debilitating conditions on patients' health and quality of life.


Subject(s)
Dysbiosis , Virus Diseases , Humans , Dysbiosis/immunology , Virus Diseases/immunology , Virus Diseases/complications , Virus Diseases/virology , Gastrointestinal Diseases/virology , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/etiology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/virology , Animals , Gastrointestinal Microbiome/immunology , Viruses/immunology , Viruses/pathogenicity , Celiac Disease/virology , Celiac Disease/immunology , Virome
19.
Viruses ; 16(8)2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39205259

ABSTRACT

The E3 ubiquitin ligase TRIM7 is known to have dual roles during viral infections. Like other TRIM proteins, TRIM7 can regulate the IFN pathway via the regulation of the cytosolic receptors RIG-I or MDA-5, which promote the production of type I interferons (IFN-I) and antiviral immune responses. Alternatively, under certain infectious conditions, TRIM7 can negatively regulate IFN-I signaling, resulting in increased virus replication. A growing body of evidence has also shown that TRIM7 can, in some cases, ubiquitinate viral proteins to promote viral replication and pathogenesis, while in other cases it can promote degradation of viral proteins through the proteasome, reducing virus infection. TRIM7 can also regulate the host inflammatory response and modulate the production of inflammatory cytokines, which can lead to detrimental inflammation. TRIM7 can also protect the host during infection by reducing cellular apoptosis. Here, we discuss the multiple functions of TRIM7 during viral infections and its potential as a therapeutic target.


Subject(s)
Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Virus Diseases , Virus Replication , Humans , Virus Diseases/immunology , Virus Diseases/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Animals , Ubiquitination , Signal Transduction , Interferon Type I/metabolism , Interferon Type I/immunology , Immunity, Innate , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/immunology , Host-Pathogen Interactions/immunology
20.
Viruses ; 16(8)2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39205302

ABSTRACT

TRIM proteins are a family of innate immune factors that play diverse roles in innate immunity and protect the cell against viral and bacterial aggression. As part of this special issue on TRIM proteins, we will take advantage of our findings on TRIM69, which acts by reorganizing the microtubules (MTs) in a manner that is fundamentally antiviral, to more generally discuss how host-pathogen interactions that take place for the control of the MT network represent a crucial facet of the struggle that opposes viruses to their cell environment. In this context, we will present several other TRIM proteins that are known to interact with microtubules in situations other than viral infection, and we will discuss evidence that may suggest a possible contribution to viral control. Overall, the present review will highlight the importance that the control of the microtubule network bears in host-pathogen interactions.


Subject(s)
Host-Pathogen Interactions , Immunity, Innate , Microtubules , Tripartite Motif Proteins , Microtubules/metabolism , Humans , Host-Pathogen Interactions/immunology , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Animals , Virus Diseases/immunology , Virus Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL