Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.627
Filter
1.
Front Cell Infect Microbiol ; 14: 1410015, 2024.
Article in English | MEDLINE | ID: mdl-38957797

ABSTRACT

Background: Tuberculosis (TB) persists as a global health challenge, with its treatment hampered by the side effects of long-term combination drug therapies and the growing issue of drug resistance. Therefore, the development of novel therapeutic strategies is critical. This study focuses on the role of immune checkpoint molecules (ICs) and functions of CD8+ T cells in the search for new potential targets against TB. Methods: We conducted differential expression genes analysis and CD8+ T cell functional gene analysis on 92 TB samples and 61 healthy individual (HI) samples from TB database GSE83456, which contains data on 34,603 genes. The GSE54992 dataset was used to validated the findings. Additionally, a cluster analysis on single-cell data from primates infected with mycobacterium tuberculosis and those vaccinated with BCG was performed. Results: The overexpression of LAG-3 gene was found as a potentially important characteristic of both pulmonary TB (PTB) and extrapulmonary TB (EPTB). Further correlation analysis showed that LAG-3 gene was correlated with GZMB, perforin, IL-2 and IL-12. A significant temporal and spatial variation in LAG-3 expression was observed in T cells and macrophages during TB infection and after BCG vaccination. Conclusion: LAG-3 was overexpressed in TB samples. Targeting LAG-3 may represent a potential therapeutic target for tuberculosis.


Subject(s)
Antigens, CD , CD8-Positive T-Lymphocytes , Lymphocyte Activation Gene 3 Protein , Mycobacterium tuberculosis , Tuberculosis , Humans , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/genetics , CD8-Positive T-Lymphocytes/immunology , Tuberculosis/immunology , Tuberculosis/microbiology , Animals , Antigens, CD/genetics , BCG Vaccine/immunology , Macrophages/immunology , Macrophages/microbiology , Interleukin-2/metabolism , Interleukin-2/genetics , Gene Expression Profiling , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Interleukin-12/genetics , Interleukin-12/metabolism , Perforin/genetics , Perforin/metabolism , Male
2.
Science ; 385(6704): eadi0908, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38963857

ABSTRACT

The major human bacterial pathogen Pseudomonas aeruginosa causes multidrug-resistant infections in people with underlying immunodeficiencies or structural lung diseases such as cystic fibrosis (CF). We show that a few environmental isolates, driven by horizontal gene acquisition, have become dominant epidemic clones that have sequentially emerged and spread through global transmission networks over the past 200 years. These clones demonstrate varying intrinsic propensities for infecting CF or non-CF individuals (linked to specific transcriptional changes enabling survival within macrophages); have undergone multiple rounds of convergent, host-specific adaptation; and have eventually lost their ability to transmit between different patient groups. Our findings thus explain the pathogenic evolution of P. aeruginosa and highlight the importance of global surveillance and cross-infection prevention in averting the emergence of future epidemic clones.


Subject(s)
Cystic Fibrosis , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/physiology , Pseudomonas aeruginosa/pathogenicity , Pseudomonas Infections/microbiology , Humans , Cystic Fibrosis/microbiology , Evolution, Molecular , Adaptation, Physiological , Gene Transfer, Horizontal , Host Specificity , Host Adaptation , Macrophages/microbiology , Macrophages/immunology
3.
Virulence ; 15(1): 2367659, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38951957

ABSTRACT

Vancomycin-resistant Enterococcus faecium (E. faecium) infection is associated with higher mortality rates. Previous studies have emphasized the importance of innate immune cells and signalling pathways in clearing E. faecium, but a comprehensive analysis of host-pathogen interactions is lacking. Here, we investigated the interplay of host and E. faecium in a murine model of septic peritonitis. Following injection with a sublethal dose, we observed significantly increased murine sepsis score and histological score, decreased weight and bacterial burden, neutrophils and macrophages infiltration, and comprehensive activation of cytokine-mediated signalling pathway. In mice receiving a lethal dose, hypothermia significantly improved survival, reduced bacterial burden, cytokines, and CD86 expression of MHC-II+ recruited macrophages compared to the normothermia group. A mathematical model constructed by observational data from 80 animals, recapitulated the host-pathogen interplay, and further verified the benefits of hypothermia. These findings indicate that E. faecium triggers a severe activation of cytokine-mediated signalling pathway, and hypothermia can improve outcomes by reducing bacterial burden and inflammation.


Subject(s)
Cytokines , Disease Models, Animal , Enterococcus faecium , Gram-Positive Bacterial Infections , Host-Pathogen Interactions , Peritonitis , Sepsis , Vancomycin-Resistant Enterococci , Animals , Peritonitis/microbiology , Peritonitis/immunology , Mice , Gram-Positive Bacterial Infections/immunology , Gram-Positive Bacterial Infections/microbiology , Vancomycin-Resistant Enterococci/pathogenicity , Sepsis/microbiology , Sepsis/immunology , Cytokines/metabolism , Mice, Inbred C57BL , Macrophages/immunology , Macrophages/microbiology , Signal Transduction
4.
Med Microbiol Immunol ; 213(1): 13, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38967888

ABSTRACT

Candida auris is an emerging pathogenic yeast that has been categorized as a global public health threat and a critical priority among fungal pathogens. Despite this, the immune response against C. auris infection is still not well understood. Hosts fight Candida infections through the immune system that recognizes pathogen-associated molecular patterns such as ß-glucan, mannan, and chitin on the fungal cell wall. In this study, levels of ß-glucan and mannan exposures in C. auris grown under different physiologically relevant stimuli were quantified by flow cytometry-based analysis. Lactate, hypoxia, and sublethal concentration of fluconazole trigger a decrease in surface ß-glucan while low pH triggers an increase in ß-glucan. There is no inverse pattern between exposure levels of ß-glucan and mannan in the cell wall architecture among the three clades. To determine the effect of cell wall remodeling on the immune response, a phagocytosis assay was performed, followed by quantification of released cytokines by ELISA. Lactate-induced decrease in ß-glucan leads to reduced uptake of C. auris by PMA-differentiated THP-1 and RAW 264.7 macrophages. Furthermore, reduced production of CCL3/MIP-1⍺ but not TNF-⍺ and IL-10 were observed. An in vivo infection analysis using silkworms reveals that a reduction in ß-glucan triggers an increase in the virulence of C. auris. This study demonstrates that ß-glucan alteration occurs in C. auris and serves as an escape mechanism from immune cells leading to increased virulence.


Subject(s)
Candida auris , Cell Wall , Immune Evasion , beta-Glucans , beta-Glucans/metabolism , Animals , Virulence , Mice , Cell Wall/immunology , Cell Wall/chemistry , Cell Wall/metabolism , Humans , Candida auris/pathogenicity , RAW 264.7 Cells , Candidiasis/microbiology , Candidiasis/immunology , Cytokines/metabolism , Phagocytosis , Macrophages/immunology , Macrophages/microbiology , Mannans/pharmacology , Lactic Acid/metabolism , Disease Models, Animal , THP-1 Cells
5.
Elife ; 132024 Jun 18.
Article in English | MEDLINE | ID: mdl-38896446

ABSTRACT

Tuberculosis is a major global health problem and is one of the top 10 causes of death worldwide. There is a pressing need for new treatments that circumvent emerging antibiotic resistance. Mycobacterium tuberculosis parasitises macrophages, reprogramming them to establish a niche in which to proliferate, therefore macrophage manipulation is a potential host-directed therapy if druggable molecular targets could be identified. The pseudokinase Tribbles1 (Trib1) regulates multiple innate immune processes and inflammatory profiles making it a potential drug target in infections. Trib1 controls macrophage function, cytokine production, and macrophage polarisation. Despite wide-ranging effects on leukocyte biology, data exploring the roles of Tribbles in infection in vivo are limited. Here, we identify that human Tribbles1 is expressed in monocytes and is upregulated at the transcript level after stimulation with mycobacterial antigen. To investigate the mechanistic roles of Tribbles in the host response to mycobacteria in vivo, we used a zebrafish Mycobacterium marinum (Mm) infection tuberculosis model. Zebrafish Tribbles family members were characterised and shown to have substantial mRNA and protein sequence homology to their human orthologues. trib1 overexpression was host-protective against Mm infection, reducing burden by approximately 50%. Conversely, trib1 knockdown/knockout exhibited increased infection. Mechanistically, trib1 overexpression significantly increased the levels of proinflammatory factors il-1ß and nitric oxide. The host-protective effect of trib1 was found to be dependent on the E3 ubiquitin kinase Cop1. These findings highlight the importance of Trib1 and Cop1 as immune regulators during infection in vivo and suggest that enhancing macrophage TRIB1 levels may provide a tractable therapeutic intervention to improve bacterial infection outcomes in tuberculosis.


Subject(s)
Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases , Zebrafish , Animals , Humans , Disease Models, Animal , Host-Pathogen Interactions , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/microbiology , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium Infections, Nontuberculous/genetics , Mycobacterium marinum , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Zebrafish/microbiology , Male , Female
6.
Can J Microbiol ; 70(7): 252-261, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38855942

ABSTRACT

Non-tuberculosis infections in immunocompromised patients represent a cause for concern, given the increased risks of infection, and limited treatments available. Herein, we report that molecules for binding to the catalytic site of histone deacetylase (HDAC) inhibit its activity, thus increasing the innate immune response against environmental mycobacteria. The action of HDAC inhibitors (iHDACs) was explored in a model of type II pneumocytes and macrophages infection by Mycobacterium aurum. The results show that the use of 1,3-diphenylurea increases the expression of the TLR-4 in M. aurum infected MDMs, as well as the production of defb4, IL-1ß, IL-12, and IL-6. Moreover, we observed that aminoacetanilide upregulates the expression of TLR-4 together with TLR-9, defb4, CAMP, RNase 6, RNase 7, IL-1ß, IL-12, and IL-6 in T2P. Results conclude that the tested iHDACs selectively modulate the expression of cytokines and antimicrobial peptides that are associated with reduction of non-tuberculous mycobacteria infection.


Subject(s)
Cytokines , Drug Repositioning , Histone Deacetylase Inhibitors , Immunity, Innate , Mycobacterium Infections, Nontuberculous , Immunity, Innate/drug effects , Humans , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium Infections, Nontuberculous/microbiology , Histone Deacetylase Inhibitors/pharmacology , Cytokines/metabolism , Macrophages/immunology , Macrophages/drug effects , Macrophages/microbiology , Nontuberculous Mycobacteria/drug effects , Nontuberculous Mycobacteria/immunology , Mycobacterium/immunology , Mycobacterium/drug effects
8.
Microbiologyopen ; 13(3): e23, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867416

ABSTRACT

The G protein-coupled estrogen receptor, also known as GPER1 or originally GPR30, is found in various tissues, indicating its diverse functions. It is typically present in immune cells, suggesting its role in regulating immune responses to infectious diseases. Our previous studies have shown that G-1, a selective GPER agonist, can limit the pathogenesis mediated by Staphylococcus aureus alpha-hemolysin (Hla). It aids in clearing bacteria in a mouse skin infection model and restricts the surface display of the Hla receptor, ADAM10 (a disintegrin and metalloprotease 10) in HaCaT keratinocytes. In this report, we delve into the modulation of GPER in human immune cells in relation to the NLRP3 inflammasome. We used macrophage-like differentiated THP-1 cells for our study. We found that treating these cells with G-1 reduces ATP release, decreases the activity of the caspase-1 enzyme, and lessens cell death following Hla intoxication. This is likely due to the reduced levels of ADAM10 and NLRP3 proteins, as well as the decreased display of the ADAM10 receptor in the G-1-treated THP-1 cells. Our studies, along with our previous work, suggest the potential therapeutic use of G-1 in reducing Hla susceptibility in humans. This highlights the importance of GPER in immune regulation and its potential as a therapeutic target.


Subject(s)
ADAM10 Protein , Amyloid Precursor Protein Secretases , Bacterial Toxins , Hemolysin Proteins , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Estrogen , Receptors, G-Protein-Coupled , Staphylococcus aureus , ADAM10 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Humans , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Hemolysin Proteins/metabolism , Inflammasomes/metabolism , Bacterial Toxins/metabolism , THP-1 Cells , Receptors, Estrogen/metabolism , Amyloid Precursor Protein Secretases/metabolism , Staphylococcus aureus/drug effects , Membrane Proteins/metabolism , Membrane Proteins/agonists , Caspase 1/metabolism , Adenosine Triphosphate/metabolism , Macrophages/immunology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/microbiology , Dipeptides , Hydroxamic Acids
9.
PLoS Genet ; 20(6): e1011313, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38870230

ABSTRACT

A quarter of humanity is estimated to have been exposed to Mycobacterium tuberculosis (Mtb) with a 5-10% risk of developing tuberculosis (TB) disease. Variability in responses to Mtb infection could be due to host or pathogen heterogeneity. Here, we focused on host genetic variation in a Peruvian population and its associations with gene regulation in monocyte-derived macrophages and dendritic cells (DCs). We recruited former household contacts of TB patients who previously progressed to TB (cases, n = 63) or did not progress to TB (controls, n = 63). Transcriptomic profiling of monocyte-derived DCs and macrophages measured the impact of genetic variants on gene expression by identifying expression quantitative trait loci (eQTL). We identified 330 and 257 eQTL genes in DCs and macrophages (False Discovery Rate (FDR) < 0.05), respectively. Four genes in DCs showed interaction between eQTL variants and TB progression status. The top eQTL interaction for a protein-coding gene was with FAH, the gene encoding fumarylacetoacetate hydrolase, which mediates the last step in mammalian tyrosine catabolism. FAH expression was associated with genetic regulatory variation in cases but not controls. Using public transcriptomic and epigenomic data of Mtb-infected monocyte-derived dendritic cells, we found that Mtb infection results in FAH downregulation and DNA methylation changes in the locus. Overall, this study demonstrates effects of genetic variation on gene expression levels that are dependent on history of infectious disease and highlights a candidate pathogenic mechanism through pathogen-response genes. Furthermore, our results point to tyrosine metabolism and related candidate TB progression pathways for further investigation.


Subject(s)
Dendritic Cells , Macrophages , Mycobacterium tuberculosis , Quantitative Trait Loci , Tuberculosis , Humans , Peru , Tuberculosis/genetics , Tuberculosis/microbiology , Macrophages/metabolism , Macrophages/microbiology , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/genetics , Female , Dendritic Cells/metabolism , Male , Adult , Genetic Predisposition to Disease , Genetic Variation , Gene Expression Regulation , Middle Aged , Polymorphism, Single Nucleotide , Gene Expression Profiling
10.
Microbiology (Reading) ; 170(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38916198

ABSTRACT

Bacterial infection is a dynamic process resulting in a heterogenous population of infected and uninfected cells. These cells respond differently based on their bacterial load and duration of infection. In the case of infection of macrophages with Crohn's disease (CD) associated adherent-invasive Escherichia coli (AIEC), understanding the drivers of pathogen success may allow targeting of cells where AIEC replicate to high levels. Here we show that stratifying immune cells based on their bacterial load identifies novel pathways and therapeutic targets not previously associated with AIEC when using a traditional homogeneous infected population approach. Using flow cytometry-based cell sorting we stratified cells into those with low or high intracellular pathogen loads, or those which were bystanders to infection. Immune cells transcriptomics revealed a diverse response to the varying levels of infection while pathway analysis identified novel intervention targets that were directly related to increasing intracellular AIEC numbers. Chemical inhibition of identified targets reduced AIEC intracellular replication or inhibited secretion of tumour necrosis factor alpha (TNFα), a key cytokine associated with AIEC infection. Our results have identified new avenues of intervention in AIEC infection that may also be applicable to CD through the repurposing of already available inhibitors. Additionally, they highlight the applicability of immune cell stratification post-infection as an effective approach for the study of microbial pathogens.


Subject(s)
Crohn Disease , Escherichia coli Infections , Escherichia coli , Macrophages , Tumor Necrosis Factor-alpha , Crohn Disease/microbiology , Crohn Disease/immunology , Macrophages/microbiology , Macrophages/immunology , Humans , Escherichia coli Infections/microbiology , Escherichia coli Infections/immunology , Escherichia coli/genetics , Tumor Necrosis Factor-alpha/metabolism , Bacterial Load , Bacterial Adhesion , Host-Pathogen Interactions
11.
Arch Microbiol ; 206(7): 287, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38833010

ABSTRACT

Hepcidin is a crucial regulator of iron homeostasis with protective effects on liver fibrosis. Additionally, gut microbiota can also affect liver fibrosis and iron metabolism. Although the hepatoprotective potential of Akkermansia muciniphila and Faecalibacterium duncaniae, formerly known as F. prausnitzii, has been reported, however, their effects on hepcidin expression remain unknown. We investigated the direct and macrophage stimulation-mediated effects of active, heat-inactivated, and cell-free supernatant (CFS) forms of A. muciniphila and F. duncaniae on hepcidin expression in HepG2 cells by RT-qPCR analysis. Following stimulation of phorbol-12-myristate-13-acetate (PMA) -differentiated THP-1 cells with A. muciniphila and F. duncaniae, IL-6 concentration was assessed via ELISA. Additionally, the resulting supernatant was treated with HepG2 cells to evaluate the effect of macrophage stimulation on hepcidin gene expression. The expression of genes mediating iron absorption and export was also examined in HepG2 and Caco-2 cells via RT-qPCR. All forms of F. duncaniae increased hepcidin expression while active and heat-inactivated/CFS forms of A. muciniphila upregulated and downregulated its expression, respectively. Active, heat-inactivated, and CFS forms of A. muciniphila and F. duncaniae upregulated hepcidin expression, consistent with the elevation of IL-6 released from THP-1-stimulated cells as a macrophage stimulation effect in HepG2 cells. A. muciniphila and F. duncaniae in active, inactive, and CFS forms altered the expression of hepatocyte and intestinal iron-mediated absorption /exporter genes, namely dcytb and dmt1, and fpn in HepG2 and Caco-2 cells, respectively. In conclusion, A. muciniphila and F. duncaniae affect not only directly but also through macrophage stimulation the expression of hepcidin gene in HepG2 cells. These findings underscore the potential of A. muciniphila and F. duncaniae as a potential therapeutic target for liver fibrosis by modulating hepcidin and intestinal and hepatocyte iron metabolism mediated gene expression.


Subject(s)
Akkermansia , Hepcidins , Macrophages , Humans , Hepcidins/genetics , Hepcidins/metabolism , Hep G2 Cells , Caco-2 Cells , Macrophages/immunology , Macrophages/microbiology , Macrophages/metabolism , THP-1 Cells , Iron/metabolism , Interleukin-6/metabolism , Interleukin-6/genetics , Macrophage Activation , Gastrointestinal Microbiome
12.
Front Immunol ; 15: 1402024, 2024.
Article in English | MEDLINE | ID: mdl-38873598

ABSTRACT

Mycobacterium tuberculosis (Mtb) is an intracellular pathogen capable of adapting and surviving within macrophages, utilizing host nutrients for its growth and replication. Cholesterol is the main carbon source during the infection process of Mtb. Cholesterol metabolism in macrophages is tightly associated with cell functions such as phagocytosis of pathogens, antigen presentation, inflammatory responses, and tissue repair. Research has shown that Mtb infection increases the uptake of low-density lipoprotein (LDL) and cholesterol by macrophages, and enhances de novo cholesterol synthesis in macrophages. Excessive cholesterol is converted into cholesterol esters, while the degradation of cholesterol esters in macrophages is inhibited by Mtb. Furthermore, Mtb infection suppresses the expression of ATP-binding cassette (ABC) transporters in macrophages, impeding cholesterol efflux. These alterations result in the massive accumulation of cholesterol in macrophages, promoting the formation of lipid droplets and foam cells, which ultimately facilitates the persistent survival of Mtb and the progression of tuberculosis (TB), including granuloma formation, tissue cavitation, and systemic dissemination. Mtb infection may also promote the conversion of cholesterol into oxidized cholesterol within macrophages, with the oxidized cholesterol exhibiting anti-Mtb activity. Recent drug development has discovered that reducing cholesterol levels in macrophages can inhibit the invasion of Mtb into macrophages and increase the permeability of anti-tuberculosis drugs. The development of drugs targeting cholesterol metabolic pathways in macrophages, as well as the modification of existing drugs, holds promise for the development of more efficient anti-tuberculosis medications.


Subject(s)
Cholesterol , Macrophages , Mycobacterium tuberculosis , Tuberculosis , Mycobacterium tuberculosis/immunology , Cholesterol/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology , Animals , Host-Pathogen Interactions/immunology , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Lipid Metabolism
13.
Front Cell Infect Microbiol ; 14: 1411196, 2024.
Article in English | MEDLINE | ID: mdl-38873097

ABSTRACT

Bacterial extracellular vesicles (EVs) are crucial mediators of information transfer between bacteria and host cells. Macrophages, as key effector cells in the innate immune system, have garnered widespread attention for their interactions with bacterial EVs. Increasing evidence indicates that bacterial EVs can be internalized by macrophages through multiple pathways, thereby influencing their immune functions. These functions include inflammatory responses, antimicrobial activity, antigen presentation, and programmed cell death. Therefore, this review summarizes current research on the interactions between bacterial EVs and macrophages. This will aid in the deeper understanding of immune modulation mediated by pathogenic microorganisms and provide a basis for developing novel antibacterial therapeutic strategies.


Subject(s)
Bacteria , Extracellular Vesicles , Immunity, Innate , Macrophages , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Macrophages/immunology , Macrophages/microbiology , Humans , Animals , Bacteria/immunology , Host-Pathogen Interactions/immunology
14.
Methods Mol Biol ; 2813: 107-115, 2024.
Article in English | MEDLINE | ID: mdl-38888773

ABSTRACT

Mass spectrometry-based proteomics provides a wealth of information about changes in protein production and abundance under diverse conditions, as well as mechanisms of regulation, signaling cascades, interaction partners, and communication patterns across biological systems. For profiling of intracellular pathogens, proteomic profiling can be performed in the absence of a host to singularly define the pathogenic proteome or during an infection-like setting to identify dual perspectives of infection. In this chapter, we present techniques to extract proteins from the human bacterial intracellular pathogen, Salmonella enterica serovar Typhimurium, in the presence of macrophages, an important innate immune cell in host defense. We outline sample preparation, including protein extraction, digestion, and purification, as well as mass spectrometry measurements and bioinformatics analysis. The data generated from our dual perspective profiling approach provides new insight into pathogen and host protein modulation under infection-like conditions.


Subject(s)
Bacterial Proteins , Macrophages , Proteomics , Salmonella typhimurium , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Proteomics/methods , Humans , Bacterial Proteins/metabolism , Macrophages/microbiology , Macrophages/metabolism , Host-Pathogen Interactions , Proteome/metabolism , Salmonella Infections/microbiology , Salmonella Infections/metabolism , Salmonella Infections/immunology , Computational Biology/methods , Mass Spectrometry/methods
15.
Commun Biol ; 7(1): 731, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879692

ABSTRACT

Ulcerative colitis (UC) is a significant inflammatory bowel disease caused by an abnormal immune response to gut microbes. However, there are still gaps in our understanding of how immune and metabolic changes specifically contribute to this disease. Our research aims to address this gap by examining mouse colons after inducing ulcerative colitis-like symptoms. Employing single-cell RNA-seq and 16 s rRNA amplicon sequencing to analyze distinct cell clusters and microbiomes in the mouse colon at different time points after induction with dextran sodium sulfate. We observe a significant reduction in epithelial populations during acute colitis, indicating tissue damage, with a partial recovery observed in chronic inflammation. Analyses of cell-cell interactions demonstrate shifts in networking patterns among different cell types during disease progression. Notably, macrophage phenotypes exhibit diversity, with a pronounced polarization towards the pro-inflammatory M1 phenotype in chronic conditions, suggesting the role of macrophage heterogeneity in disease severity. Increased expression of Nampt and NOX2 complex subunits in chronic UC macrophages contributes to the inflammatory processes. The chronic UC microbiome exhibits reduced taxonomic diversity compared to healthy conditions and acute UC. The study also highlights the role of T cell differentiation in the context of dysbiosis and its implications in colitis progression, emphasizing the need for targeted interventions to modulate the inflammatory response and immune balance in colitis.


Subject(s)
Colitis, Ulcerative , Dextran Sulfate , Gastrointestinal Microbiome , Macrophages , Single-Cell Analysis , Animals , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/immunology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Macrophages/immunology , Macrophages/microbiology , Macrophages/metabolism , Dextran Sulfate/toxicity , Dextran Sulfate/adverse effects , Mice , RNA-Seq , Mice, Inbred C57BL , Disease Models, Animal , DNA Barcoding, Taxonomic , RNA, Ribosomal, 16S/genetics , Male , Single-Cell Gene Expression Analysis
16.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38892443

ABSTRACT

Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis (TB), a prevalent infectious disease affecting populations worldwide. A classic trait of TB pathology is the formation of granulomas, which wall off the pathogen, via the innate and adaptive immune systems. Some key players involved include tumor necrosis factor-alpha (TNF-α), foamy macrophages, type I interferons (IFNs), and reactive oxygen species, which may also show overlap with cell death pathways. Additionally, host cell death is a primary method for combating and controlling Mtb within the body, a process which is influenced by both host and bacterial factors. These cell death modalities have distinct molecular mechanisms and pathways. Programmed cell death (PCD), encompassing apoptosis and autophagy, typically confers a protective response against Mtb by containing the bacteria within dead macrophages, facilitating their phagocytosis by uninfected or neighboring cells, whereas necrotic cell death benefits the pathogen, leading to the release of bacteria extracellularly. Apoptosis is triggered via intrinsic and extrinsic caspase-dependent pathways as well as caspase-independent pathways. Necrosis is induced via various pathways, including necroptosis, pyroptosis, and ferroptosis. Given the pivotal role of host cell death pathways in host defense against Mtb, therapeutic agents targeting cell death signaling have been investigated for TB treatment. This review provides an overview of the diverse mechanisms underlying Mtb-induced host cell death, examining their implications for host immunity. Furthermore, it discusses the potential of targeting host cell death pathways as therapeutic and preventive strategies against Mtb infection.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/pathology , Animals , Cell Death/immunology , Host-Pathogen Interactions/immunology , Apoptosis , Immunity, Innate , Autophagy/immunology , Signal Transduction , Macrophages/immunology , Macrophages/microbiology
17.
Sci Rep ; 14(1): 12811, 2024 06 04.
Article in English | MEDLINE | ID: mdl-38834738

ABSTRACT

Macrophages provide a crucial environment for Salmonella enterica serovar Typhi (S. Typhi) to multiply during typhoid fever, yet our understanding of how human macrophages and S. Typhi interact remains limited. In this study, we delve into the dynamics of S. Typhi replication within human macrophages and the resulting heterogeneous transcriptomic responses of macrophages during infection. Our study reveals key factors that influence macrophage diversity, uncovering distinct immune and metabolic pathways associated with different stages of S. Typhi intracellular replication in macrophages. Of note, we found that macrophages harboring replicating S. Typhi are skewed towards an M1 pro-inflammatory state, whereas macrophages containing non-replicating S. Typhi exhibit neither a distinct M1 pro-inflammatory nor M2 anti-inflammatory state. Additionally, macrophages with replicating S. Typhi were characterized by the increased expression of genes associated with STAT3 phosphorylation and the activation of the STAT3 transcription factor. Our results shed light on transcriptomic pathways involved in the susceptibility of human macrophages to intracellular S. Typhi replication, thereby providing crucial insight into host phenotypes that restrict and support S. Typhi infection.


Subject(s)
Macrophages , STAT3 Transcription Factor , Salmonella typhi , Typhoid Fever , Humans , Macrophages/metabolism , Macrophages/microbiology , Salmonella typhi/genetics , Typhoid Fever/microbiology , Typhoid Fever/immunology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Gene Expression Profiling , Phenotype , Transcriptome , Phosphorylation
18.
PLoS One ; 19(6): e0304876, 2024.
Article in English | MEDLINE | ID: mdl-38848336

ABSTRACT

We have identified an acyl-carrier protein, Rv0100, that is up-regulated in a dormancy model. This protein plays a critical role in the fatty acid biosynthesis pathway, which is important for energy storage and cell wall synthesis in Mycobacterium tuberculosis (MTB). Knocking out the Rv0100 gene resulted in a significant reduction of growth compared to wild-type MTB in the Wayne model of non-replicating persistence. We have also shown that Rv0100 is essential for the growth and survival of this pathogen during infection in mice and a macrophage model. Furthermore, knocking out Rv0100 disrupted the synthesis of phthiocerol dimycocerosates, the virulence-enhancing lipids produced by MTB and Mycobacterium bovis. We hypothesize that this essential gene contributes to MTB virulence in the state of latent infection. Therefore, inhibitors targeting this gene could prove to be potent antibacterial agents against this pathogen.


Subject(s)
Acyl Carrier Protein , Bacterial Proteins , Mycobacterium tuberculosis , Animals , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Mice , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Acyl Carrier Protein/metabolism , Acyl Carrier Protein/genetics , Macrophages/microbiology , Macrophages/metabolism , Virulence , Gene Expression Regulation, Bacterial , Tuberculosis/microbiology , Lipids/chemistry
19.
Med Microbiol Immunol ; 213(1): 9, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38900248

ABSTRACT

Endogenous antimicrobial peptides (AMPs) play a key role in the host defense against pathogens. AMPs attack pathogens preferentially at the site of entry to prevent invasive infection. Mycobacterium tuberculosis (Mtb) enters its host via the airways. AMPs released into the airways are therefore likely candidates to contribute to the clearance of Mtb immediately after infection. Since lysozyme is detectable in airway secretions, we evaluated its antimicrobial activity against Mtb. We demonstrate that lysozyme inhibits the growth of extracellular Mtb, including isoniazid-resistant strains. Lysozyme also inhibited the growth of non-tuberculous mycobacteria. Even though lysozyme entered Mtb-infected human macrophages and co-localized with the pathogen we did not observe antimicrobial activity. This observation was unlikely related to the large size of lysozyme (14.74 kDa) because a smaller lysozyme-derived peptide also co-localized with Mtb without affecting the viability. To evaluate whether the activity of lysozyme against extracellular Mtb could be relevant in vivo, we incubated Mtb with fractions of human serum and screened for antimicrobial activity. After several rounds of sub-fractionation, we identified a highly active fraction-component as lysozyme by mass spectrometry. In summary, our results identify lysozyme as an antimycobacterial protein that is detectable as an active compound in human serum. Our results demonstrate that the activity of AMPs against extracellular bacilli does not predict efficacy against intracellular pathogens despite co-localization within the macrophage. Ongoing experiments are designed to unravel peptide modifications that occur in the intracellular space and interfere with the deleterious activity of lysozyme in the extracellular environment.


Subject(s)
Macrophages , Muramidase , Mycobacterium tuberculosis , Muramidase/pharmacology , Muramidase/metabolism , Humans , Macrophages/metabolism , Macrophages/microbiology , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/metabolism , Microbial Sensitivity Tests , Microbial Viability/drug effects
20.
Pathog Dis ; 822024 Feb 07.
Article in English | MEDLINE | ID: mdl-38845379

ABSTRACT

Tuberculosis (TB) continues to pose a significant global health challenge, emphasizing the critical need for effective preventive measures. Although many studies have tried to develop new attenuated vaccines, there is no effective TB vaccine. In this study, we report a novel attenuated Mycobacterium tuberculosis (M. tb) strain, CHVAC-25, cultured continuously for 25 years in the laboratory. CHVAC-25 exhibited significantly reduced virulence compared to both the virulent H37Rv strain in C57BL/6J and severe combined immunodeficiency disease mice. The comparative genomic analysis identified 93 potential absent genomic segments and 65 single nucleotide polymorphic sites across 47 coding genes. Notably, the deletion mutation of ppsC (Rv2933) involved in phthiocerol dimycocerosate synthesis likely contributes to CHVAC-25 virulence attenuation. Furthermore, the comparative analysis of immune responses between H37Rv- and CHVAC-25-infected macrophages showed that CHVAC-25 triggered a robust upregulation of 173 genes, particularly cytokines crucial for combating M. tb infection. Additionally, the survival of CHVAC-25 was significantly reduced compared to H37Rv in macrophages. These findings reiterate the possibility of obtaining attenuated M. tb strains through prolonged laboratory cultivation, echoing the initial conception of H37Ra nearly a century ago. Additionally, the similarity of CHVAC-25 to genotypes associated with attenuated M. tb vaccine positions it as a promising candidate for TB vaccine development.


Subject(s)
Macrophages , Mycobacterium tuberculosis , Tuberculosis Vaccines , Vaccines, Attenuated , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Animals , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/genetics , Mice , Macrophages/immunology , Macrophages/microbiology , Virulence/genetics , Vaccines, Attenuated/immunology , Vaccines, Attenuated/genetics , Genome, Bacterial , Genomics/methods , Mice, Inbred C57BL , Cytokines/metabolism , Tuberculosis/microbiology , Tuberculosis/immunology , Tuberculosis/prevention & control , Polymorphism, Single Nucleotide , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL
...