Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.479
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: 41-50, 2025.
Article in English | MEDLINE | ID: mdl-39192117

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems , Gene Knockout Techniques , Herpesvirus 1, Human , Immune Evasion , Immunity, Innate , CRISPR-Cas Systems/genetics , Immunity, Innate/genetics , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/genetics , Immune Evasion/genetics , Humans , Gene Editing/methods , Animals , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Herpes Simplex/immunology , Herpes Simplex/virology , Herpes Simplex/genetics
3.
Methods Mol Biol ; 2854: 51-60, 2025.
Article in English | MEDLINE | ID: mdl-39192118

ABSTRACT

The application of CRISPR-mediated library screening has fundamentally transformed functional genomics by revealing the complexity of virus-host interactions. This protocol describes the use of CRISPR-mediated library screening to identify key functional genes regulating the innate immune response to PEDV infection. We detail a step-by-step process, starting from the design and construction of a customized CRISPR knockout library targeting genes involved in innate immunity to the effective delivery of these constructs into cells using lentiviral vectors. Subsequently, we outline the process of identifying functional genes postviral attack, including the use of next-generation sequencing (NGS), to analyze and identify knockout cells that exhibit altered responses to infection. This integrated approach provides researchers in immunology and virology with a resource and a robust framework for uncovering the genetic basis of host-pathogen interactions and the arsenal of the innate immune system against viral invasions.


Subject(s)
CRISPR-Cas Systems , Gene Knockout Techniques , Gene Library , Immunity, Innate , Immunity, Innate/genetics , CRISPR-Cas Systems/genetics , Humans , High-Throughput Nucleotide Sequencing/methods , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Cell Line , Lentivirus/genetics
4.
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
5.
Methods Mol Biol ; 2854: 127-141, 2025.
Article in English | MEDLINE | ID: mdl-39192125

ABSTRACT

Luciferase reporter systems are commonly used in scientific research to investigate a variety of biological processes, including antiviral innate immunity. These systems employ the use of luciferase enzymes derived from organisms such as fireflies or renilla reniformis, which emit light upon reaction with a substrate. In the context of antiviral innate immunity, the luciferase reporter systems offer a noninvasive and highly sensitive approach for real-time monitoring of immune responses in vitro and in vivo, enabling researchers to delve into the intricate interactions and signaling pathways involved in host-virus dynamic interactions. Here, we describe the methods of the promoter-luciferase reporter and enhancer-luciferase reporter, which provide insights into the transcriptional and post-transcriptional regulation of antiviral innate immunity. Additionally, we outline the split-luciferase complementary reporter method, which was designed to explore protein-protein interactions associated with antiviral immunity. These methodologies offer invaluable knowledge regarding the molecular mechanisms underlying antiviral immune pathways and have the potential to support the development of effective antiviral therapies.


Subject(s)
Genes, Reporter , Immunity, Innate , Luciferases , Humans , Luciferases/metabolism , Luciferases/genetics , Animals , Interferons/metabolism , Interferons/immunology , Promoter Regions, Genetic , Antiviral Agents/pharmacology , HEK293 Cells , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics
6.
Commun Biol ; 7(1): 1287, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39384974

ABSTRACT

Co-localization of spatial transcriptome information of host and pathogen can revolutionize our understanding of microbial pathogenesis. Here, we aimed to demonstrate that customized bacterial probes can be successfully used to identify host-pathogen interactions in formalin-fixed-paraffin-embedded (FFPE) tissues by probe-based spatial transcriptomics technology. We analyzed the spatial gene expression of bacterial transcripts with the host transcriptomic profile in murine lung tissue chronically infected with Mycobacterium abscessus embedded in agar beads. Customized mycobacterial probes were designed for the constitutively expressed rpoB gene (an RNA polymerase ß subunit) and the virulence factor precursor lsr2, modulated by oxidative stress. We found a correlation between the rpoB expression, bacterial abundance in the airways, and an increased expression of lsr2 virulence factor in lung tissue with high oxidative stress. Overall, we demonstrate the potential of dual bacterial and host gene expression assay in FFPE tissues, paving the way for the simultaneous detection of host and bacterial transcriptomes in pathological tissues.


Subject(s)
Host-Pathogen Interactions , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Mycobacterium abscessus/genetics , Animals , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium Infections, Nontuberculous/genetics , Mice , Host-Pathogen Interactions/genetics , Gene Expression Regulation, Bacterial , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Transcriptome , Lung/microbiology , Gene Expression Profiling/methods , Female , Virulence Factors/genetics , Virulence Factors/metabolism , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics
7.
BMC Genomics ; 25(1): 937, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39375608

ABSTRACT

BACKGROUND: Spinach downy mildew, caused by the obligate oomycete pathogen, Peronospora effusa remains a major concern for spinach production. Disease control is predominantly based on development of resistant spinach cultivars. However, new races and novel isolates of the pathogen continue to emerge and overcome cultivar resistance. Currently there are 20 known races of P. effusa. Here we characterized the transcriptomes of spinach, Spinacia oleracea, and P. effusa during disease progression using the spinach cultivar Viroflay, the near isogenic lines NIL1 and NIL3, and P. effusa races, R13 and R19, at 24 h post inoculation and 6 days post inoculation. A total of 54 samples were collected and subjected to sequencing and transcriptomic analysis. RESULTS: Differentially expressed gene (DEG) analysis in resistant spinach interactions of R13-NIL1 and R19-NIL3 revealed spinach DEGs from protein kinase-like and P-loop containing families, which have roles in plant defense. The homologous plant defense genes included but were not limited to, receptor-like protein kinases (Spiol0281C06495, Spiol06Chr21559 and Spiol06Chr24027), a BAK1 homolog (Spiol0223C05961), genes with leucine rich repeat motifs (Spiol04Chr08771, Spiol04Chr01972, Spiol05Chr26812, Spiol04Chr11049, Spiol0084S08137, Spiol03Chr20299) and ABC-transporters (Spiol02Chr28975, Spiol06Chr22112, Spiol06Chr03998 and Spiol04Chr09723). Additionally, analysis of the expression of eight homologous to previously reported downy mildew resistance genes revealed that some are differentially expressed during resistant reactions but not during susceptible reactions. Examination of P. effusa gene expression during infection of susceptible cultivars identified expressed genes present in R19 or R13 including predicted RxLR and Crinkler effector genes that may be responsible for race-specific virulence on NIL1 or NIL3 spinach hosts, respectively. CONCLUSIONS: These findings deliver foundational insight to gene expression in both spinach and P. effusa during susceptible and resistant interactions and provide a library of candidate genes for further exploration and functional analysis. Such resources will be beneficial to spinach breeding efforts for disease resistance in addition to better understanding the virulence mechanisms of this obligate pathogen.


Subject(s)
Disease Resistance , Peronospora , Plant Diseases , Spinacia oleracea , Spinacia oleracea/genetics , Spinacia oleracea/microbiology , Spinacia oleracea/parasitology , Peronospora/physiology , Peronospora/pathogenicity , Disease Resistance/genetics , Plant Diseases/microbiology , Plant Diseases/genetics , Plant Diseases/parasitology , Transcriptome , Gene Expression Profiling , Gene Expression Regulation, Plant , Host-Pathogen Interactions/genetics , Plant Proteins/genetics , Plant Proteins/metabolism
8.
J Med Virol ; 96(10): e29955, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39370864

ABSTRACT

Epstein‒Barr virus (EBV) regulates the expression of host genes involved in functional pathways for viral infection and pathogenicity. Long noncoding RNAs (lncRNAs) have been found to be important regulators of cellular biology. However, how EBV affects host biological processes via lncRNAs remains elusive. Eukaryotic initiation factor 4A3 (EIF4A3) was recently identified as an essential controller of cell fate with an unknown role in EBV infection. Here, the expression of lncRNA brain cytoplasmic 200 (BC200) was shown to be significantly upregulated in EBV-infected cell lines. RNA immunoprecipitation and RNA pulldown assays confirmed that BC200 bound to EIF4A3. Moreover, BC200 promoted EIF4A3 expression at the protein level but not at the mRNA level. Mechanistically, BC200 stabilized the EIF4A3 protein by impeding the K48-linked polyubiquitination of the K195 and K198 residues of EIF4A3. In addition, RNA-seq analysis of EBV-positive cells with knockdown of either BC200 or EIF4A3 revealed that a broad range of cellular genes were differentially regulated, particularly those related to virus infection and immune response pathways. This study is the first to reveal the key residues involved in EIF4A3 polyubiquitination and elucidate the novel regulatory role of EBV in host gene expression via the BC200/EIF4A3 axis. These results have implications for the pathogenesis and treatment of EBV-related diseases.


Subject(s)
Epstein-Barr Virus Infections , Eukaryotic Initiation Factor-4A , Herpesvirus 4, Human , RNA, Long Noncoding , Humans , Cell Line , DEAD-box RNA Helicases , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4A/metabolism , Gene Expression Regulation , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Ubiquitination
9.
BMC Genomics ; 25(1): 917, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39358701

ABSTRACT

BACKGROUND: The fungus Metarhizium brunneum has evolved a remarkable ability to switch between different lifestyles. It develops as a saprophyte, an endophyte establishing mutualistic relationships with plants, or a parasite, enabling its use for the control of insect pests such as the aphid Myzus persicae. We tested our hypothesis that switches between lifestyles must be accompanied by fundamental transcriptional reprogramming, reflecting adaptations to different environmental settings. RESULTS: We combined high throughput RNA sequencing of M. brunneum in vitro and at different stages of pathogenesis to validate the modulation of genes in the fungus and its host during the course of infection. In agreement with our hypothesis, we observed transcriptional reprogramming in M. brunneum following conidial attachment, germination on the cuticle, and early-stage growth within the host. This involved the upregulation of genes encoding degrading enzymes and gene clusters involved in synthesis of secondary metabolites that act as virulence factors. The transcriptional response of the aphid host included the upregulation of genes potentially involved in antifungal activity, but antifungal peptides were not induced. We also observed the induction of a host flightin gene, which may be involved in wing formation and flight muscle development. CONCLUSIONS: The switch from saprophytic to parasitic development in M. brunneum is accompanied by fundamental transcriptional reprogramming during the course of the infection. The aphid host responds to fungal infection with its own transcriptional reprogramming, reflecting its inability to express antifungal peptides but featuring the induction of genes involved in winged morphs that may enable offspring to avoid the contaminated environment.


Subject(s)
Aphids , Metarhizium , Animals , Aphids/microbiology , Aphids/physiology , Metarhizium/physiology , Metarhizium/genetics , Metarhizium/pathogenicity , Gene Expression Regulation, Fungal , Host-Pathogen Interactions/genetics , Gene Expression Profiling , Transcription, Genetic
10.
Am Nat ; 204(4): 370-380, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39326061

ABSTRACT

AbstractHost organisms can harbor microbial symbionts that defend them from pathogen infection in addition to the resistance encoded by the host genome. Here, we investigated how variation in defenses, generated from host genetic background and symbiont presence, affects the emergence of pathogen genetic diversity across evolutionary time. We passaged the opportunistic pathogen Pseudomonas aeruginosa through populations of the nematode Caenorhabditis elegans varying in genetic-based defenses and prevalence of a protective symbiont. After 14 passages, we assessed the amount of genetic variation accumulated in evolved pathogen lineages. We found that diversity begets diversity. An overall greater level of pathogen whole-genome and per-gene genetic diversity was measured in pathogens evolved in mixed host populations compared with those evolved in host populations composed of one type of defense. Our findings directly demonstrate that symbiont-generated heterogeneity in host defense can be a significant contributor to pathogen genetic variation.


Subject(s)
Caenorhabditis elegans , Genetic Variation , Pseudomonas aeruginosa , Symbiosis , Animals , Pseudomonas aeruginosa/genetics , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/genetics , Host-Pathogen Interactions/genetics , Biological Evolution
11.
Sci Adv ; 10(39): eadn5945, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39331715

ABSTRACT

Orsay virus (OrV) is the only known natural virus affecting Caenorhabditis elegans, with minimal impact on the animal's fitness due to its robust innate immune response. This study aimed to understand the interactions between C. elegans and OrV by tracking the infection's progression during larval development. Four distinct stages of infection were identified on the basis of viral load, with a peak in capsid-encoding RNA2 coinciding with the first signs of viral egression. Transcriptomic analysis revealed temporal changes in gene expression and functions induced by the infection. A specific set of up-regulated genes remained active throughout the infection, and genes correlated and anticorrelated with virus accumulation were identified. Responses to OrV mirrored reactions to other biotic stressors, distinguishing between virus-specific responses and broader immune responses. Moreover, mutants of early response genes and defense-related processes showed altered viral load progression, uncovering additional players in the antiviral defense response.


Subject(s)
Caenorhabditis elegans , Host-Pathogen Interactions , Viral Load , Animals , Caenorhabditis elegans/virology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/immunology , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Immunity, Innate , Nodaviridae/physiology , Gene Expression Profiling , Transcriptome , Gene Expression Regulation
12.
BMC Genomics ; 25(1): 876, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294575

ABSTRACT

BACKGROUND: Begomoviruses are major constraint in the production of many crops. Upon infection, begomoviruses may substantially modulate plant biological processes. While how monopartite begomoviruses interact with their plant hosts has been investigated extensively, bipartite begomoviruses-plant interactions are understudied. Moreover, as one of the major groups of hosts, cucurbitaceous plants have been seldom examined in the interaction with begomoviruses. RESULTS: We profiled the zucchini transcriptomic changes induced by a bipartite begomovirus squash leaf curl China virus (SLCCNV). We identified 2275 differentially-expressed genes (DEGs), of which 1310 were upregulated and 965 were downregulated. KEGG enrichment analysis of the DEGs revealed that many pathways related to primary and secondary metabolisms were enriched. qRT-PCR verified the transcriptional changes of twelve selected DEGs induced by SLCCNV infection. Close examination revealed that the expression levels of all the DEGs of the pathway Photosynthesis were downregulated upon SLCCNV infection. Most DEGs in the pathway Plant-pathogen interaction were upregulated, including some positive regulators of plant defenses. Moreover, the majority of DEGs in the MAPK signaling pathway-plant were upregulated. CONCLUSION: Our findings indicates that SLCCNV actively interact with its cucurbitaceous plant host by suppressing the conversion of light energy to chemical energy and inducing immune responses. Our study not only provides new insights into the interactions between begomoviruses and host plants, but also adds to our knowledge on virus-plant interactions in general.


Subject(s)
Begomovirus , Gene Expression Profiling , Host-Pathogen Interactions , Plant Diseases , Begomovirus/genetics , Host-Pathogen Interactions/genetics , Plant Diseases/virology , Plant Diseases/genetics , Transcriptome , Gene Expression Regulation, Plant , Cucurbita/virology , Cucurbita/genetics
13.
Front Cell Infect Microbiol ; 14: 1368887, 2024.
Article in English | MEDLINE | ID: mdl-39290979

ABSTRACT

Introduction: Newcastle disease is one of the significant issues in the poultry industry, having catastrophic effects worldwide. The lung is one of the essential organs which harbours Bronchus-associated lymphoid tissue and plays a vital role in the immune response. Leghorn and Fayoumi breeds are known to have differences in resistance to Newcastle disease. Along with genes and long non-coding RNAs (lncRNAs) are also known to regulate various biological pathways through gene regulation. Methods: This study analysed the lung transcriptome data and identified the role of genes and long non-coding RNAs in differential immune resistance. The computational pipeline, FHSpipe, as used in our previous studies on analysis of harderian gland and trachea transcriptome was used to identify genes and lncRNAs. This was followed by differential expression analysis, functional annotation of genes and lncRNAs, identification of transcription factors, microRNAs and finally validation using qRT-PCR. Results and discussion: A total of 8219 novel lncRNAs were identified. Of them, 1263 lncRNAs and 281 genes were differentially expressed. About 66 genes were annotated with either an immune-related GO term or pathway, and 12 were annotated with both. In challenge and breed-based analysis, most of these genes were upregulated in Fayoumi compared to Leghorn, and in timepoint-based analysis, Leghorn challenge chicken showed downregulation between time points. A similar trend was observed in the expression of lncRNAs. Co-expression analysis has revealed several lncRNAs co-expressing with immune genes with a positive correlation. Several genes annotated with non-immune pathways, including metabolism, signal transduction, transport of small molecules, extracellular matrix organization, developmental biology and cellular processes, were also impacted. With this, we can understand that Fayoumi chicken showed upregulated immune genes and positive cis-lncRNAs during both the non-challenged and NDV-challenge conditions, even without viral transcripts in the tissue. This finding shows that these immune-annotated genes and coexpressing cis-lncRNAs play a significant role in Fayoumi being comparatively resistant to NDV compared to Leghorn. Our study affirms and expands upon the outcomes of previous studies and highlights the crucial role of lncRNAs during the immune response to NDV. Conclusion: This analysis clearly shows the differences in the gene expression patterns and lncRNA co-expression with the genes between Leghorn and Fayoumi, indicating that the lncRNAs and co-expressing genes might potentially have a role in differentiating these breeds. We hypothesise that these genes and lncRNAs play a vital role in the higher resistance of Fayoumi to NDV than Leghorn. This study can pave the way for future studies to unravel the biological mechanism behind the regulation of immune-related genes.


Subject(s)
Chickens , Gene Expression Profiling , Lung , MicroRNAs , Newcastle Disease , Newcastle disease virus , Poultry Diseases , RNA, Long Noncoding , Transcriptome , Animals , Chickens/genetics , Chickens/immunology , Newcastle Disease/immunology , Newcastle Disease/virology , Newcastle Disease/genetics , Lung/immunology , Lung/virology , Newcastle disease virus/immunology , Newcastle disease virus/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Poultry Diseases/virology , Poultry Diseases/immunology , Poultry Diseases/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Gene Expression Regulation , Disease Resistance/genetics , Computational Biology/methods , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology
14.
J Gen Virol ; 105(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-39292505

ABSTRACT

Arabidopsis thaliana is more susceptible to certain viruses during its later developmental stages. The differential responses and the mechanisms behind this development-dependent susceptibility to infection are still not fully understood. Here we explored the outcome of a viral infection at different host developmental stages by studying the response of A. thaliana to infection with turnip mosaic virus at three developmental stages: juvenile vegetative, bolting, and mature flowering plants. We found that infected plants at later stages downregulate cell wall biosynthetic genes and that this downregulation may be one factor facilitating viral spread and systemic infection. We also found that, despite being more susceptible to infection, infected mature flowering plants were more fertile (i.e. produce more viable seeds) than juvenile vegetative and bolting infected plants; that is, plants infected at the reproductive stage have greater fitness than plants infected at earlier developmental stages. Moreover, treatment of mature plants with salicylic acid increased resistance to infection at the cost of significantly reducing fertility. Together, these observations support a negative trade-off between viral susceptibility and plant fertility. Our findings point towards a development-dependent tolerance to infection.


Subject(s)
Arabidopsis , Gene Expression Regulation, Plant , Plant Diseases , Potyvirus , Plant Diseases/virology , Arabidopsis/virology , Arabidopsis/genetics , Arabidopsis/growth & development , Potyvirus/physiology , Salicylic Acid/metabolism , Host-Pathogen Interactions/genetics , Plant Growth Regulators/metabolism , Gene Expression Profiling
15.
Front Immunol ; 15: 1419782, 2024.
Article in English | MEDLINE | ID: mdl-39295861

ABSTRACT

Bacterial infections remain a significant global health concern, necessitating a comprehensive understanding of the intricate host-pathogen interactions that play a critical role in the outcome of infectious diseases. Recent investigations have revealed that noncoding RNAs (ncRNAs) are key regulators of these complex interactions. Among them, long noncoding RNAs (lncRNAs) have gained significant attention because of their diverse regulatory roles in gene expression, cellular processes and the production of cytokines and chemokines in response to bacterial infections. The host utilizes lncRNAs as a defense mechanism to limit microbial pathogen invasion and replication. On the other hand, some host lncRNAs contribute to the establishment and maintenance of bacterial pathogen reservoirs within the host by promoting bacterial pathogen survival, replication, and dissemination. However, our understanding of host lncRNAs in the context of bacterial infections remains limited. This review focuses on the impact of host lncRNAs in shaping host-pathogen interactions, shedding light on their multifaceted functions in both host defense and bacterial survival, and paving the way for future research aimed at harnessing their regulatory potential for clinical applications.


Subject(s)
Bacterial Infections , Host-Pathogen Interactions , RNA, Long Noncoding , RNA, Long Noncoding/genetics , Humans , Bacterial Infections/immunology , Bacterial Infections/genetics , Bacterial Infections/microbiology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Animals , Bacteria/genetics , Bacteria/immunology , Gene Expression Regulation
16.
Front Immunol ; 15: 1431207, 2024.
Article in English | MEDLINE | ID: mdl-39308873

ABSTRACT

The Mycobacterium tuberculosis variant bovis (M. bovis) is a highly pathogenic environmental microorganism that causes bovine tuberculosis (bTB), a significant zoonotic disease. Currently, "test and culling" is the primary measure for controlling bTB, but it has been proven to be inadequate in animals due to their high susceptibility to the pathogen. Selective breeding for increased host resistance to bTB to reduce its prevalence is feasible. In this study, we found a vital host-dependent factor, RBMX2, that can potentially promote M. bovis infection. By knocking RBMX2 out, we investigated its function during M. bovis infection. Through transcriptome sequencing and alternative splicing transcriptome sequencing, we concluded that after M. bovis infection, embryo bovine lung (EBL) cells were significantly enriched in RNA splicing associated with apoptosis compared with wild-type EBL cells. Through protein/molecular docking, molecular dynamics simulations, and real-time quantitative PCR, we demonstrated that RBMX2 promotes the apoptosis of epithelial cells by upregulating and binding to apoptotic peptidase activating factor 1 (APAF-1), resulting in the alternative splicing of APAF-1 as a retention intron. To our knowledge, this is the first report of M. bovis affecting host epithelial cell apoptosis by hijacking RBMX2 to promote the intron splicing of downstream APAF-1. These findings may represent a significant contribution to the development of novel TB prevention and control strategies.


Subject(s)
Apoptosis , Apoptotic Protease-Activating Factor 1 , Epithelial Cells , Introns , Mycobacterium bovis , Tuberculosis, Bovine , Animals , Apoptotic Protease-Activating Factor 1/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Apoptosis/genetics , Mycobacterium bovis/physiology , Cattle , Tuberculosis, Bovine/microbiology , Tuberculosis, Bovine/genetics , Introns/genetics , Epithelial Cells/microbiology , Epithelial Cells/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Alternative Splicing , Down-Regulation , Host-Pathogen Interactions/genetics
18.
BMC Genomics ; 25(1): 828, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227786

ABSTRACT

Disease can act as a driving force in shaping genetic makeup across populations, even species, if the impacts influence a particularly sensitive part of their life cycles. White-nose disease is caused by a fungal pathogen infecting bats during hibernation. The mycosis has caused massive population declines of susceptible species in North America, particularly in the genus Myotis. However, Myotis bats appear to tolerate infection in Eurasia, where the fungal pathogen has co-evolved with its bat hosts for an extended period of time. Therefore, with susceptible and tolerant populations, the fungal disease provides a unique opportunity to tease apart factors contributing to tolerance at a genomic level to and gain an understanding of the evolution of non-harmful in host-parasite interactions. To investigate if the fungal disease has caused adaptation on a genomic level in Eurasian bat species, we adopted both whole-genome sequencing approaches and a literature search to compile a set of 300 genes from which to investigate signals of positive selection in genomes of 11 Eurasian bats at the codon-level. Our results indicate significant positive selection in 38 genes, many of which have a marked role in responses to infection. Our findings suggest that white-nose syndrome may have applied a significant selective pressure on Eurasian Myotis-bats in the past, which can contribute their survival in co-existence with the pathogen. Our findings provide an insight on the selective pressure pathogens afflict on their hosts using methodology that can be adapted to other host-pathogen study systems.


Subject(s)
Chiroptera , Selection, Genetic , Chiroptera/microbiology , Chiroptera/genetics , Animals , Host-Pathogen Interactions/genetics , Genome , Mycoses/microbiology , Mycoses/veterinary , Evolution, Molecular , Genomics/methods , Whole Genome Sequencing
19.
Nat Commun ; 15(1): 7640, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223139

ABSTRACT

Genetic parasites, including viruses and transposons, exploit components from the host for their own replication. However, little is known about virus-transposon interactions within host cells. Here, we discover a strategy where human cytomegalovirus (HCMV) hijacks L1 retrotransposon encoded protein during its replication cycle. HCMV infection upregulates L1 expression by enhancing both the expression of L1-activating transcription factors, YY1 and RUNX3, and the chromatin accessibility of L1 promoter regions. Increased L1 expression, in turn, promotes HCMV replicative fitness. Affinity proteomics reveals UL44, HCMV DNA polymerase subunit, as the most abundant viral binding protein of the L1 ribonucleoprotein (RNP) complex. UL44 directly interacts with L1 ORF2p, inducing DNA damage responses in replicating HCMV compartments. While increased L1-induced mutagenesis is not observed in HCMV for genetic adaptation, the interplay between UL44 and ORF2p accelerates viral DNA replication by alleviating replication stress. Our findings shed light on how HCMV exploits host retrotransposons for enhanced viral fitness.


Subject(s)
Cytomegalovirus , DNA Replication , Long Interspersed Nucleotide Elements , Viral Proteins , Virus Replication , Humans , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Virus Replication/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , DNA Replication/genetics , Long Interspersed Nucleotide Elements/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/genetics , Host-Pathogen Interactions/genetics , Retroelements/genetics , DNA-Binding Proteins
20.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273397

ABSTRACT

Fusarium head blight (FHB), caused by the Fusarium graminearum species complex, is a destructive disease in wheat worldwide. The lack of FHB-resistant germplasm is a barrier in wheat breeding for resistance to FHB. Thinopyrum elongatum is an important relative that has been successfully used for the genetic improvement of wheat. In this study, a translocation line, YNM158, with the YM158 genetic background carrying a fragment of diploid Th. elongatum 7EL chromosome created using 60Co-γ radiation, showed high resistance to FHB under both field and greenhouse conditions. Transcriptome analysis confirmed that the horizontal transfer gene, encoding glutathione S-transferase (GST), is an important contributor to FHB resistance in the pathogen infection stage, whereas the 7EL chromosome fragment carries other genes regulated by F. graminearum during the colonization stage. Introgression of the 7EL fragment affected the expression of wheat genes that were enriched in resistance pathways, including the phosphatidylinositol signaling system, protein processing in the endoplasmic reticulum, plant-pathogen interaction, and the mitogen-activated protein kinase (MAPK) signaling pathway at different stages after F. graminearium infection. This study provides a novel germplasm for wheat resistance to FHB and new insights into the molecular mechanisms of wheat resistance to FHB.


Subject(s)
Disease Resistance , Fusarium , Plant Diseases , Triticum , Fusarium/pathogenicity , Triticum/microbiology , Triticum/genetics , Disease Resistance/genetics , Plant Diseases/microbiology , Plant Diseases/genetics , Transcriptome/genetics , Translocation, Genetic , Gene Expression Regulation, Plant , Gene Expression Profiling/methods , Plant Proteins/genetics , Plant Proteins/metabolism , Poaceae/genetics , Poaceae/microbiology , Host-Pathogen Interactions/genetics
SELECTION OF CITATIONS
SEARCH DETAIL