Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.066
Filter
1.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38830771

ABSTRACT

Dengue fever, a neglected tropical arboviral disease, has emerged as a global health concern in the past decade. Necessitating a nuanced comprehension of the intricate dynamics of host-virus interactions influencing disease severity, we analysed transcriptomic patterns using bulk RNA-seq from 112 age- and gender-matched NS1 antigen-confirmed hospital-admitted dengue patients with varying severity. Severe cases exhibited reduced platelet count, increased lymphocytosis, and neutropenia, indicating a dysregulated immune response. Using bulk RNA-seq, our analysis revealed a minimal overlap between the differentially expressed gene and transcript isoform, with a distinct expression pattern across the disease severity. Severe patients showed enrichment in retained intron and nonsense-mediated decay transcript biotypes, suggesting altered splicing efficiency. Furthermore, an up-regulated programmed cell death, a haemolytic response, and an impaired interferon and antiviral response at the transcript level were observed. We also identified the potential involvement of the RBM39 gene among others in the innate immune response during dengue viral pathogenesis, warranting further investigation. These findings provide valuable insights into potential therapeutic targets, underscoring the importance of exploring transcriptomic landscapes between different disease sub-phenotypes in infectious diseases.


Subject(s)
Alternative Splicing , Dengue Virus , Severe Dengue , Humans , Alternative Splicing/genetics , Female , Male , Dengue Virus/genetics , Adult , Severe Dengue/genetics , Severe Dengue/immunology , Severe Dengue/virology , Middle Aged , Transcriptome/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Gene Expression Profiling/methods , Immunity, Innate/genetics , Dengue/genetics , Dengue/immunology , Dengue/virology , Young Adult , Severity of Illness Index , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology
2.
Front Cell Infect Microbiol ; 14: 1398077, 2024.
Article in English | MEDLINE | ID: mdl-38836056

ABSTRACT

Mycobacterium tuberculosis (M.tb), the causative agent of Tuberculosis, is an intracellular bacterium well known for its ability to subvert host energy and metabolic pathways to maintain its intracellular survival. For this purpose, the bacteria utilize various mechanisms of which extracellular vehicles (EVs) related mechanisms attracted more attention. EVs are nanosized particles that are released by almost all cell types containing active biomolecules from the cell of origin and can target bioactive pathways in the recipient cells upon uptake. It is hypothesized that M.tb dictates the processes of host EV biogenesis pathways, selectively incorporating its molecules into the host EV to direct immune responses in its favor. During infection with Mtb, both mycobacteria and host cells release EVs. The composition of these EVs varies over time, influenced by the physiological and nutritional state of the host environment. Additionally, different EV populations contribute differently to the pathogenesis of disease at various stages of illness participating in a complex interplay between host cells and pathogens. These interactions ultimately influence immune responses and disease outcomes. However, the precise mechanisms and roles of EVs in pathogenicity and disease outcomes remain to be fully elucidated. In this review, we explored the properties and function of EVs in the context of M.tb infection within the host microenvironment and discussed their capacity as a novel therapeutic strategy to combat tuberculosis.


Subject(s)
Extracellular Vesicles , Host-Pathogen Interactions , Mycobacterium tuberculosis , Tuberculosis , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Mycobacterium tuberculosis/immunology , Humans , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/metabolism , Host-Pathogen Interactions/immunology , Animals
3.
Front Immunol ; 15: 1401320, 2024.
Article in English | MEDLINE | ID: mdl-38835769

ABSTRACT

Host-microbe interactions are complex and ever-changing, especially during infections, which can significantly impact human physiology in both health and disease by influencing metabolic and immune functions. Infections caused by pathogens such as bacteria, viruses, fungi, and parasites are the leading cause of global mortality. Microbes have evolved various immune evasion strategies to survive within their hosts, which presents a multifaceted challenge for detection. Intracellular microbes, in particular, target specific cell types for survival and replication and are influenced by factors such as functional roles, nutrient availability, immune evasion, and replication opportunities. Identifying intracellular microbes can be difficult because of the limitations of traditional culture-based methods. However, advancements in integrated host microbiome single-cell genomics and transcriptomics provide a promising basis for personalized treatment strategies. Understanding host-microbiota interactions at the cellular level may elucidate disease mechanisms and microbial pathogenesis, leading to targeted therapies. This article focuses on how intracellular microbes reside in specific cell types, modulating functions through persistence strategies to evade host immunity and prolong colonization. An improved understanding of the persistent intracellular microbe-induced differential disease outcomes can enhance diagnostics, therapeutics, and preventive measures.


Subject(s)
Genomics , Single-Cell Analysis , Humans , Genomics/methods , Animals , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Host Microbial Interactions/immunology , Host Microbial Interactions/genetics , Immune Evasion , Microbiota/immunology , Bacteria/genetics , Bacteria/immunology , Severity of Illness Index
5.
Methods Mol Biol ; 2808: 121-127, 2024.
Article in English | MEDLINE | ID: mdl-38743366

ABSTRACT

During the infection of a host cell by an infectious agent, a series of gene expression changes occurs as a consequence of host-pathogen interactions. Unraveling this complex interplay is the key for understanding of microbial virulence and host response pathways, thus providing the basis for new molecular insights into the mechanisms of pathogenesis and the corresponding immune response. Dual RNA sequencing (dual RNA-seq) has been developed to simultaneously determine pathogen and host transcriptomes enabling both differential and coexpression analyses between the two partners as well as genome characterization in the case of RNA viruses. Here, we provide a detailed laboratory protocol and bioinformatics analysis guidelines for dual RNA-seq experiments focusing on - but not restricted to - measles virus (MeV) as a pathogen of interest. The application of dual RNA-seq technologies in MeV-infected patients can potentially provide valuable information on the structure of the viral RNA genome and on cellular innate immune responses and drive the discovery of new targets for antiviral therapy.


Subject(s)
Genome, Viral , Host-Pathogen Interactions , Measles virus , Measles , RNA, Viral , Humans , Measles/virology , Measles/immunology , Measles/genetics , Measles virus/genetics , Measles virus/pathogenicity , RNA, Viral/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Computational Biology/methods , Sequence Analysis, RNA/methods , RNA-Seq/methods , Transcriptome , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods
6.
Int J Mol Sci ; 25(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731848

ABSTRACT

The pathogenesis of chronic wounds (CW) involves a multifaceted interplay of biochemical, immunological, hematological, and microbiological interactions. Biofilm development is a significant virulence trait which enhances microbial survival and pathogenicity and has various implications on the development and management of CW. Biofilms induce a prolonged suboptimal inflammation in the wound microenvironment, associated with delayed healing. The composition of wound fluid (WF) adds more complexity to the subject, with proven pro-inflammatory properties and an intricate crosstalk among cytokines, chemokines, microRNAs, proteases, growth factors, and ECM components. One approach to achieve information on the mechanisms of disease progression and therapeutic response is the use of multiple high-throughput 'OMIC' modalities (genomic, proteomic, lipidomic, metabolomic assays), facilitating the discovery of potential biomarkers for wound healing, which may represent a breakthrough in this field and a major help in addressing delayed wound healing. In this review article, we aim to summarize the current progress achieved in host-microbiome crosstalk in the spectrum of CW healing and highlight future innovative strategies to boost the host immune response against infections, focusing on the interaction between pathogens and their hosts (for instance, by harnessing microorganisms like probiotics), which may serve as the prospective advancement of vaccines and treatments against infections.


Subject(s)
Biofilms , Microbiota , Wound Healing , Humans , Biofilms/growth & development , Animals , Chronic Disease , Host-Pathogen Interactions/immunology
7.
Cell Mol Life Sci ; 81(1): 203, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698289

ABSTRACT

Nitrogen metabolism of M. tuberculosis is critical for its survival in infected host cells. M. tuberculosis has evolved sophisticated strategies to switch between de novo synthesis and uptake of various amino acids from host cells for metabolic demands. Pyridoxal phosphate-dependent histidinol phosphate aminotransferase-HspAT enzyme is critically required for histidine biosynthesis. HspAT is involved in metabolic synthesis of histidine, phenylalanine, tyrosine, tryptophan, and novobiocin. We showed that M. tuberculosis Rv2231c is a conserved enzyme with HspAT activity. Rv2231c is a monomeric globular protein that contains α-helices and ß-sheets. It is a secretory and cell wall-localized protein that regulates critical pathogenic attributes. Rv2231c enhances the survival and virulence of recombinant M. smegmatis in infected RAW264.7 macrophage cells. Rv2231c is recognized by the TLR4 innate immune receptor and modulates the host immune response by suppressing the secretion of the antibacterial pro-inflammatory cytokines TNF, IL-12, and IL-6. It also inhibits the expression of co-stimulatory molecules CD80 and CD86 along with antigen presenting molecule MHC-I on macrophage and suppresses reactive nitrogen species formation, thereby promoting M2 macrophage polarization. Recombinant M. smegmatis expressing Rv2231c inhibited apoptosis in macrophages, promoting efficient bacterial survival and proliferation, thereby increasing virulence. Our results indicate that Rv2231c is a moonlighting protein that regulates multiple functions of M. tuberculosis pathophysiology to increase its virulence. These mechanistic insights can be used to better understand the pathogenesis of M. tuberculosis and to design strategies for tuberculosis mitigation.


Subject(s)
Macrophages , Mycobacterium tuberculosis , Transaminases , Mice , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Animals , RAW 264.7 Cells , Virulence , Macrophages/microbiology , Macrophages/immunology , Macrophages/metabolism , Transaminases/metabolism , Transaminases/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Mycobacterium smegmatis/pathogenicity , Mycobacterium smegmatis/metabolism , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/enzymology , Cytokines/metabolism , Toll-Like Receptor 4/metabolism , Humans , Immunity, Innate , Host-Pathogen Interactions/immunology , Tuberculosis/immunology , Tuberculosis/microbiology
8.
Front Immunol ; 15: 1390468, 2024.
Article in English | MEDLINE | ID: mdl-38726006

ABSTRACT

Introduction: Relapsing fever (RF) remains a neglected human disease that is caused by a number of diverse pathogenic Borrelia (B.) species. Characterized by high cell densities in human blood, relapsing fever spirochetes have developed plentiful strategies to avoid recognition by the host defense mechanisms. In this scenario, spirochetal lipoproteins exhibiting multifunctional binding properties in the interaction with host-derived molecules are known to play a key role in adhesion, fibrinolysis and complement activation. Methods: Binding of CihC/FbpC orthologs to different human proteins and conversion of protein-bound plasminogen to proteolytic active plasmin were examined by ELISA. To analyze the inhibitory capacity of CihC/FbpC orthologs on complement activation, a microtiter-based approach was performed. Finally, AlphaFold predictions were utilized to identified the complement-interacting residues. Results and discussion: Here, we elucidate the binding properties of CihC/FbpC-orthologs from distinct RF spirochetes including B. parkeri, B. hermsii, B. turicatae, and B. recurrentis to human fibronectin, plasminogen, and complement component C1r. All CihC/FbpC-orthologs displayed similar binding properties to fibronectin, plasminogen, and C1r, respectively. Functional studies revealed a dose dependent binding of plasminogen to all borrelial proteins and conversion to active plasmin. The proteolytic activity of plasmin was almost completely abrogated by tranexamic acid, indicating that lysine residues are involved in the interaction with this serine protease. In addition, a strong inactivation capacity toward the classical pathway could be demonstrated for the wild-type CihC/FbpC-orthologs as well as for the C-terminal CihC fragment of B. recurrentis. Pre-incubation of human serum with borrelial molecules except CihC/FbpC variants lacking the C-terminal region protected serum-susceptible Borrelia cells from complement-mediated lysis. Utilizing AlphaFold2 predictions and existing crystal structures, we mapped the putative key residues involved in C1r binding on the CihC/FbpC orthologs attempting to explain the relatively small differences in C1r binding affinity despite the substitutions of key residues. Collectively, our data advance the understanding of the multiple binding properties of structural and functional highly similar molecules of relapsing fever spirochetes proposed to be involved in pathogenesis and virulence.


Subject(s)
Bacterial Proteins , Borrelia , Fibrinolysis , Plasminogen , Protein Binding , Relapsing Fever , Humans , Borrelia/immunology , Borrelia/metabolism , Relapsing Fever/microbiology , Relapsing Fever/immunology , Relapsing Fever/metabolism , Plasminogen/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Complement Activation , Immune Evasion , Bacterial Adhesion , Host-Pathogen Interactions/immunology , Fibronectins/metabolism , Fibrinolysin/metabolism , Complement System Proteins/immunology , Complement System Proteins/metabolism
9.
Front Cell Infect Microbiol ; 14: 1365221, 2024.
Article in English | MEDLINE | ID: mdl-38711929

ABSTRACT

Bunyaviruses are a large group of important viral pathogens that cause significant diseases in humans and animals worldwide. Bunyaviruses are enveloped, single-stranded, negative-sense RNA viruses that infect a wide range of hosts. Upon entry into host cells, the components of viruses are recognized by host innate immune system, leading to the activation of downstream signaling cascades to induce interferons (IFNs) and other proinflammatory cytokines. IFNs bind to their receptors and upregulate the expression of hundreds of interferon-stimulated genes (ISGs). Many ISGs have antiviral activities and confer an antiviral state to host cells. For efficient replication and spread, viruses have evolved different strategies to antagonize IFN-mediated restriction. Here, we discuss recent advances in our understanding of the interactions between bunyaviruses and host innate immune response.


Subject(s)
Bunyaviridae Infections , Immunity, Innate , Orthobunyavirus , Bunyaviridae Infections/immunology , Bunyaviridae Infections/virology , Humans , Animals , Orthobunyavirus/immunology , Host-Pathogen Interactions/immunology , Interferons/immunology , Interferons/metabolism , Signal Transduction , Cytokines/metabolism , Cytokines/immunology , Vector Borne Diseases/immunology , Vector Borne Diseases/virology , Vector Borne Diseases/prevention & control , Virus Replication
10.
Front Immunol ; 15: 1358036, 2024.
Article in English | MEDLINE | ID: mdl-38690262

ABSTRACT

Background: It remains unclear whether BPIV3 infection leads to stress granules formation and whether G3BP1 plays a role in this process and in viral replication. This study aims to clarify the association between BPIV3 and stress granules, explore the effect of G3BP1 on BPIV3 replication, and provide significant insights into the mechanisms by which BPIV3 evades the host's antiviral immunity to support its own survival. Methods: Here, we use Immunofluorescence staining to observe the effect of BPIV3 infection on the assembly of stress granules. Meanwhile, the expression changes of eIF2α and G3BP1 were determined. Overexpression or siRNA silencing of intracellular G3BP1 levels was examined for its regulatory control of BPIV3 replication. Results: We identify that the BPIV3 infection elicited phosphorylation of the eIF2α protein. However, it did not induce the assembly of stress granules; rather, it inhibited the formation of stress granules and downregulated the expression of G3BP1. G3BP1 overexpression facilitated the formation of stress granules within cells and hindered viral replication, while G3BP1 knockdown enhanced BPIV3 expression. Conclusion: This study suggest that G3BP1 plays a crucial role in BPIV3 suppressing stress granule formation and viral replication.


Subject(s)
DNA Helicases , Poly-ADP-Ribose Binding Proteins , RNA Helicases , RNA Recognition Motif Proteins , Stress Granules , Virus Replication , Animals , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , RNA Recognition Motif Proteins/metabolism , DNA Helicases/metabolism , DNA Helicases/genetics , RNA Helicases/metabolism , RNA Helicases/genetics , Stress Granules/metabolism , Cattle , Eukaryotic Initiation Factor-2/metabolism , Respirovirus Infections/immunology , Respirovirus Infections/metabolism , Host-Pathogen Interactions/immunology , Phosphorylation , Cell Line , Cytoplasmic Granules/metabolism
11.
Sci Adv ; 10(22): eadn7848, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38809989

ABSTRACT

Streptococcus agalactiae [group B Streptococcus (GBS)] is a leading cause of neonatal meningitis, with late-onset disease (LOD) occurring after gastrointestinal tract colonization in infants. Bacterial membrane lipids are essential for host-pathogen interactions, and the functions of glycolipids are yet to be fully elucidated. GBS synthesizes three major glycolipids: glucosyl-diacylglycerol (Glc-DAG), diglucosyl-DAG (Glc2-DAG), and lysyl-Glc-DAG (Lys-Glc-DAG). Here, we identify the enzyme, IagB, as responsible for biosynthesis of Glc-DAG, the precursor for the two other glycolipids in GBS. To examine the collective role of glycolipids to GBS virulence, we adapted a murine model of neonatal meningitis to simulate LOD. The GBS∆iagB mutant traversed the gut-epithelial barrier comparable to wild type but was severely attenuated in bloodstream survival, resulting in decreased bacterial loads in the brain. The GBS∆iagB mutant was more susceptible to neutrophil killing and membrane targeting by host antimicrobial peptides. This work reveals an unexplored function of GBS glycolipids with their ability to protect the bacterial cell from host antimicrobial killing.


Subject(s)
Glycolipids , Streptococcal Infections , Streptococcus agalactiae , Streptococcus agalactiae/pathogenicity , Streptococcus agalactiae/immunology , Streptococcus agalactiae/metabolism , Animals , Glycolipids/metabolism , Glycolipids/immunology , Mice , Virulence , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Humans , Disease Models, Animal , Host-Pathogen Interactions/immunology , Neutrophils/immunology , Neutrophils/metabolism , Mutation
12.
Front Immunol ; 15: 1407649, 2024.
Article in English | MEDLINE | ID: mdl-38812510

ABSTRACT

Infection with human papillomavirus (HPV) typically leads to cervical cancer, skin related cancers and many other tumors. HPV is mainly responsible for evading immune tumor monitoring in HPV related cancers. Toll like receptors (TLRs) are particular pattern recognition molecules. When the body is facing immune danger, it can lead to innate and direct adaptive immunity. TLR plays an important role in initiating antiviral immune responses. HPV can affect the expression level of TLR and interfere with TLR related signaling pathways, resulting in sustained viral infection and even carcinogenesis. This paper introduces the HPV virus and HPV related cancers. We discussed the present comprehension of TLR, its expression and signaling, as well as its role in HPV infection. We also provided a detailed introduction to immunotherapy methods for HPV related diseases based on TLR agonists. This will provide insights into methods that support the therapeutic method of HPV related conditions with TLR agonists.


Subject(s)
Papillomaviridae , Papillomavirus Infections , Toll-Like Receptors , Humans , Toll-Like Receptors/metabolism , Toll-Like Receptors/agonists , Toll-Like Receptors/immunology , Papillomavirus Infections/immunology , Papillomavirus Infections/therapy , Papillomavirus Infections/virology , Papillomaviridae/physiology , Papillomaviridae/immunology , Signal Transduction , Neoplasms/therapy , Neoplasms/immunology , Animals , Immunotherapy/methods , Female , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/therapy , Uterine Cervical Neoplasms/immunology , Host-Pathogen Interactions/immunology
13.
Nat Microbiol ; 9(6): 1540-1554, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38806670

ABSTRACT

Epstein-Barr virus (EBV) is an aetiologic risk factor for the development of multiple sclerosis (MS). However, the role of EBV-infected B cells in the immunopathology of MS is not well understood. Here we characterized spontaneous lymphoblastoid cell lines (SLCLs) isolated from MS patients and healthy controls (HC) ex vivo to study EBV and host gene expression in the context of an individual's endogenous EBV. SLCLs derived from MS patient B cells during active disease had higher EBV lytic gene expression than SLCLs from MS patients with stable disease or HCs. Host gene expression analysis revealed activation of pathways associated with hypercytokinemia and interferon signalling in MS SLCLs and upregulation of forkhead box protein 1 (FOXP1), which contributes to EBV lytic gene expression. We demonstrate that antiviral approaches targeting EBV replication decreased cytokine production and autologous CD4+ T cell responses in this ex vivo model. These data suggest that dysregulation of intrinsic B cell control of EBV gene expression drives a pro-inflammatory, pathogenic B cell phenotype that can be attenuated by suppressing EBV lytic gene expression.


Subject(s)
B-Lymphocytes , Epstein-Barr Virus Infections , Herpesvirus 4, Human , Multiple Sclerosis , Humans , Herpesvirus 4, Human/genetics , Multiple Sclerosis/virology , Multiple Sclerosis/immunology , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/complications , Cytokines/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Transcriptome , Virus Replication , Gene Expression Regulation, Viral , Cell Line , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Gene Expression Profiling , Adult , Female , Male
14.
Front Cell Infect Microbiol ; 14: 1384611, 2024.
Article in English | MEDLINE | ID: mdl-38808065

ABSTRACT

Neisseria gonorrhoeae (Ng) is a uniquely adapted human pathogen and the etiological agent of gonorrhea, a sexually transmitted disease. Ng has developed numerous mechanisms to avoid and actively suppress innate and adaptive immune responses. Ng successfully colonizes and establishes topologically distinct colonies in human macrophages and avoids phagocytic killing. During colonization, Ng manipulates the actin cytoskeleton to invade and create an intracellular niche supportive of bacterial replication. The cellular reservoir(s) supporting bacterial replication and persistence in gonorrhea infections are poorly defined. The manner in which gonococci colonize macrophages points to this innate immune phagocyte as a strong candidate for a cellular niche during natural infection. Here we investigate whether nutrients availability and immunological polarization alter macrophage colonization by Ng. Differentiation of macrophages in pro-inflammatory (M1-like) and tolerogenic (M2-like) phenotypes prior to infection reveals that Ng can invade macrophages in all activation states, albeit with lower efficiency in M1-like macrophages. These results suggest that during natural infection, bacteria could invade and grow within macrophages regardless of the nutrients availability and the macrophage immune activation status.


Subject(s)
Macrophages , Neisseria gonorrhoeae , Nutrients , Neisseria gonorrhoeae/immunology , Macrophages/microbiology , Macrophages/immunology , Humans , Gonorrhea/microbiology , Gonorrhea/immunology , Macrophage Activation , Host-Pathogen Interactions/immunology
15.
Proc Natl Acad Sci U S A ; 121(22): e2402764121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38771879

ABSTRACT

Staphylococcus aureus (S. aureus) can evade antibiotics and host immune defenses by persisting within infected cells. Here, we demonstrate that in infected host cells, S. aureus type VII secretion system (T7SS) extracellular protein B (EsxB) interacts with the stimulator of interferon genes (STING) protein and suppresses the inflammatory defense mechanism of macrophages during early infection. The binding of EsxB with STING disrupts the K48-linked ubiquitination of EsxB at lysine 33, thereby preventing EsxB degradation. Furthermore, EsxB-STING binding appears to interrupt the interaction of 2 vital regulatory proteins with STING: aspartate-histidine-histidine-cysteine domain-containing protein 3 (DHHC3) and TNF receptor-associated factor 6. This persistent dual suppression of STING interactions deregulates intracellular proinflammatory pathways in macrophages, inhibiting STING's palmitoylation at cysteine 91 and its K63-linked ubiquitination at lysine 83. These findings uncover an immune-evasion mechanism by S. aureus T7SS during intracellular macrophage infection, which has implications for developing effective immunomodulators to combat S. aureus infections.


Subject(s)
Bacterial Proteins , Macrophages , Membrane Proteins , Staphylococcal Infections , Staphylococcus aureus , Type VII Secretion Systems , Ubiquitination , Staphylococcus aureus/immunology , Membrane Proteins/metabolism , Membrane Proteins/immunology , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Animals , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/metabolism , Type VII Secretion Systems/metabolism , Type VII Secretion Systems/immunology , Type VII Secretion Systems/genetics , Mice , Immune Evasion , Host-Pathogen Interactions/immunology
16.
J Med Virol ; 96(6): e29685, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38783790

ABSTRACT

Human papillomaviruses (HPVs) are double-stranded DNA (dsDNA) tumor viruses causally associated with 5% of human cancers, comprising both anogenital and upper aerodigestive tract carcinomas. Despite the availability of prophylactic vaccines, HPVs continue to pose a significant global health challenge, primarily due to inadequate vaccine access and coverage. These viruses can establish persistent infections by evading both the intrinsic defenses of infected tissues and the extrinsic defenses provided by professional innate immune cells. Crucial for their evasion strategies is their unique intraepithelial life cycle, which effectively shields them from host detection. Thus, strategies aimed at reactivating the innate immune response within infected or transformed epithelial cells, particularly through the production of type I interferons (IFNs) and lymphocyte-recruiting chemokines, are considered viable solutions to counteract the adverse effects of persistent infections by these oncogenic viruses. This review focuses on the complex interplay between the high-risk HPV oncoproteins E6 and E7 and the innate immune response in epithelial cells and HPV-associated cancers. In particular, it details the molecular mechanisms by which E6 and E7 modulate the innate immune response, highlighting significant progress in our comprehension of these processes. It also examines forward-looking strategies that exploit the innate immune system to ameliorate existing anticancer therapies, thereby providing crucial insights into future therapeutic developments.


Subject(s)
Immune Evasion , Immunity, Innate , Oncogene Proteins, Viral , Papillomavirus Infections , Humans , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/immunology , Papillomaviridae/immunology , Papillomaviridae/pathogenicity , Host-Pathogen Interactions/immunology , Epithelial Cells/virology , Epithelial Cells/immunology
17.
Retrovirology ; 21(1): 10, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38778414

ABSTRACT

BACKGROUND: Detection of viruses by host pattern recognition receptors induces the expression of type I interferon (IFN) and IFN-stimulated genes (ISGs), which suppress viral replication. Numerous studies have described HIV-1 as a poor activator of innate immunity in vitro. The exact role that the viral capsid plays in this immune evasion is not fully understood. RESULTS: To better understand the role of the HIV-1 capsid in sensing we tested the effect of making HIV-1 by co-expressing a truncated Gag that encodes the first 107 amino acids of capsid fused with luciferase or GFP, alongside wild type Gag-pol. We found that unlike wild type HIV-1, viral particles produced with a mixture of wild type and truncated Gag fused to luciferase or GFP induced a potent IFN response in THP-1 cells and macrophages. Innate immune activation by Gag-fusion HIV-1 was dependent on reverse transcription and DNA sensor cGAS, suggesting activation of an IFN response by viral DNA. Further investigation revealed incorporation of the Gag-luciferase/GFP fusion proteins into viral particles that correlated with subtle defects in wild type Gag cleavage and a diminished capacity to saturate restriction factor TRIM5α, likely due to aberrant particle formation. We propose that expression of the Gag fusion protein disturbs the correct cleavage and maturation of wild type Gag, yielding viral particles that are unable to effectively shield viral DNA from detection by innate sensors including cGAS. CONCLUSIONS: These data highlight the crucial role of capsid in innate evasion and support growing literature that disruption of Gag cleavage and capsid formation induces a viral DNA- and cGAS-dependent innate immune response. Together these data demonstrate a protective role for capsid and suggest that antiviral activity of capsid-targeting antivirals may benefit from enhanced innate and adaptive immunity in vivo.


Subject(s)
HIV-1 , Immunity, Innate , Nucleotidyltransferases , gag Gene Products, Human Immunodeficiency Virus , HIV-1/immunology , HIV-1/genetics , HIV-1/physiology , Humans , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology , gag Gene Products, Human Immunodeficiency Virus/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Antiviral Restriction Factors , Macrophages/immunology , Macrophages/virology , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , THP-1 Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/immunology , Immune Evasion , Capsid/metabolism , Capsid/immunology , Virus Replication , Virion/metabolism , Virion/genetics , Virion/immunology , Host-Pathogen Interactions/immunology , DNA, Viral/genetics , Cell Line
18.
Adv Exp Med Biol ; 1451: 219-237, 2024.
Article in English | MEDLINE | ID: mdl-38801581

ABSTRACT

The monkeypox virus (MPXV), responsible for human disease, has historically been limited to the African countries, with only a few isolated instances reported elsewhere in the world. Nevertheless, in recent years, there have been occurrences of monkeypox in regions where the disease is typically absent, which has garnered global interest. Within a period of less than four months, the incidence of MPXV infections has surged to over 48,000 cases, resulting in a total of 13 deaths. This chapter has addressed the genetics of the pox virus, specifically the human monkeypox virus, and its interaction with the immune systems of host organisms. The present chapter is skillfully constructed, encompassing diagnostic methodologies that span from traditional to developing molecular techniques. Furthermore, the chapter provides a succinct analysis of the therapeutic methods employed, potential future developments, and the various emerging difficulties encountered in illness management.


Subject(s)
Monkeypox virus , Mpox (monkeypox) , Humans , Monkeypox virus/genetics , Monkeypox virus/immunology , Monkeypox virus/pathogenicity , Mpox (monkeypox)/diagnosis , Mpox (monkeypox)/immunology , Mpox (monkeypox)/epidemiology , Mpox (monkeypox)/virology , Mpox (monkeypox)/therapy , Host-Pathogen Interactions/immunology , Animals
19.
Methods Mol Biol ; 2775: 13-27, 2024.
Article in English | MEDLINE | ID: mdl-38758308

ABSTRACT

Cryptococcal meningitis (CM) is a fungal disease caused by the invasion of Cryptococcus yeast cells into the central nervous system. The organism is thought to enter the body through the lungs and then escape due to dysregulation of the immune response. Multiple animal species have been used to model the infection and characterize CM including mice, rats, dogs, guinea pigs, and rabbits. The rabbit model has over 40 years of data and has been used to study host-pathogen interactions and the efficacy of antifungal therapeutics. The model begins with immune suppression to eliminate the lymphocytic cell population followed by direct infection of the central nervous system via an injection of a suspension of yeast cells into the cisterna magna. The organism remains in the CNS during the course of infection, and cerebrospinal fluid can be repeatedly sampled to quantify the burden of organism, measure drug levels in the CSF, profile the immune response in the CSF, and/or characterize the yeast cells. The rabbit model of infection is a robust experimental model for better understanding CM and Cryptococcus cellular behavior.


Subject(s)
Disease Models, Animal , Meningitis, Cryptococcal , Animals , Meningitis, Cryptococcal/immunology , Meningitis, Cryptococcal/microbiology , Meningitis, Cryptococcal/cerebrospinal fluid , Meningitis, Cryptococcal/pathology , Rabbits , Cryptococcus neoformans , Antifungal Agents/therapeutic use , Antifungal Agents/pharmacology , Host-Pathogen Interactions/immunology , Cryptococcus/immunology
20.
Methods Mol Biol ; 2775: 171-193, 2024.
Article in English | MEDLINE | ID: mdl-38758318

ABSTRACT

The interaction between macrophages and Cryptococcus neoformans is crucial in the pathogenesis of cryptococcosis. These phagocytes are important immune effectors, but also a niche in which facultative intracellular parasites, such as C. neoformans, thrive. Consequently, phagocytosis of cryptococcal cells and its outcomes are very frequently studied. One major issue with several of the tests used for this, however, is that macrophage-C. neoformans interaction does not always result in phagocytosis, as fungi may be attached to the external surface of the phagocyte. The most used methodologies to study phagocytosis of cryptococcal cells have varying degrees of precision in separating fungi that are truly internalized from those that are outside macrophages. Here we describe two assays to measure phagocytosis that can differentiate internal from external C. neoformans cells.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Macrophages , Phagocytosis , Cryptococcus neoformans/immunology , Macrophages/microbiology , Macrophages/immunology , Macrophages/metabolism , Cryptococcosis/microbiology , Cryptococcosis/immunology , Animals , Mice , Humans , Host-Pathogen Interactions/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...