Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.163
Filter
1.
Arch Microbiol ; 206(8): 361, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39066807

ABSTRACT

In the complex realm of bacterial infections, particularly those caused by Staphylococcus aureus (S. aureus), macrophages play a pivotal role in orchestrating the immune response. During the initial stages of infection, the monocytes give rise to macrophages with a pro-inflammatory (M1 type) behaviour, engulfing and neutralizing the invading pathogens. However, under the sustained influence of S. aureus infection, monocytes can undergo a transition into an anti-inflammatory M2 state (pro-infection) rather than the M1 state (anti-infection), thereby compromising effective infection control. Therefore, it is necessary to develop a strategy that would preserve the pro-inflammatory functions of macrophages, in a safe and controlled manner. For this, we focused on harnessing the potential of S. aureus-derived ghost cells (GCs) which are non-live empty envelopes of bacterial cells, but with the antigenic determinants intact. Through a unique Lugol's-iodine treatment, we generated GCs and characterization of these GCs using gel electrophoresis, FTIR, flow cytometry, TEM, and SEM confirmed their structural integrity. Following this, we assessed the extend of cellular association of the GCs with RAW267.4 macrophages, and observed an immediate interaction between the two, as evident from the flowcytometry and microscopy studies. We then performed macrophage polarisation on a human monocyte-macrophage model cell line, THP-1. Our findings revealed that GCs effectively activated macrophages, and promoted a pro-inflammatory polarisation with the expression of M1 differentiation markers (CD86, TNFα, IL-1ß, IL-6, IL-12) evaluated through both qPCR and ELISA. Interestingly an intermediary expression of M2 markers viz., CD206 and IL-10 was also observed, but was overruled by the enhanced expression of M1 markers at a later time point. Overall, our study introduces a novel approach utilizing GCs to guide naïve macrophages towards M1 subtypes, thereby potentiating immune responses during microbial infections. This innovative strategy can modulate macrophage function, ultimately improving outcomes in S. aureus infections and beyond.


Subject(s)
Cell Differentiation , Macrophages , Staphylococcal Infections , Staphylococcus aureus , Macrophages/immunology , Macrophages/microbiology , Staphylococcus aureus/immunology , Humans , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Animals , Mice , Monocytes/immunology , Monocytes/microbiology , Cytokines/metabolism , Cytokines/immunology , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/metabolism , Inflammation/immunology
2.
Int J Mol Sci ; 25(14)2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39063137

ABSTRACT

Ehrlichia chaffeensis infects and proliferates inside monocytes or macrophages and causes human monocytic ehrlichiosis (HME), an emerging life-threatening tick-borne zoonosis. After internalization, E. chaffeensis resides in specialized membrane-bound inclusions, E. chaffeensis-containing vesicles (ECVs), to evade from host cell innate immune responses and obtain nutrients. However, mechanisms exploited by host cells to inhibit E. chaffeensis growth in ECVs are still largely unknown. Here we demonstrate that host cells recognize E. chaffeensis Ech_1067, a penicillin-binding protein, and then upregulate the expression of PIT1, which is a phosphate transporter and transports phosphate from ECVs to the cytosol to inhibit bacterial growth. We found that host cells upregulate the PIT1 expression upon E. chaffeensis infection using transcriptome sequencing, qRT-PCR and Western blotting, and PIT1 is localized on the ECV membrane in infected THP-1 cells using confocal microscopy. Silence of PIT1 using shRNA enhances E. chaffeensis intracellular growth. Finally, we found that E. chaffeensis Ech_1067 induces the upregulation of PIT1 expression through the MyD88-NF-κB pathway using recombinant protein for stimulation and siRNA for silence. Our findings deepen the understanding of the innate immune responses of host cells to inhibit bacterial intracellular growth and facilitate the development of new therapeutics for HME.


Subject(s)
Ehrlichia chaffeensis , Humans , Ehrlichia chaffeensis/metabolism , Ehrlichia chaffeensis/genetics , THP-1 Cells , Up-Regulation , Ehrlichiosis/microbiology , Ehrlichiosis/metabolism , Host-Pathogen Interactions , Macrophages/metabolism , Macrophages/microbiology , Macrophages/immunology , Phosphates/metabolism , NF-kappa B/metabolism , Monocytes/metabolism , Monocytes/microbiology
3.
Lett Appl Microbiol ; 77(5)2024 May 03.
Article in English | MEDLINE | ID: mdl-38755018

ABSTRACT

Our study aimed to identify markers of enterococci's virulence potential by evaluating the properties of strains of different sites of isolation. Enterococcal strains were isolated as commensals from faeces and as invasive strains from the urine and blood of patients from the University Clinical Centre, Gdansk, Poland. Changes in monocytes' susceptibility to the cytotoxic activity of isolates of different origins and their adherence to biofilm were evaluated using a flow cytometer. The bacterial protein profile was estimated by matrix assisted laser desorption ionization-time of flight mass spectrometer. The cytotoxicity of biofilm and monocytes' adherence to it were the most accurate factors in predicting the prevalence of the strain in the specific niche. Additionally, a bacterial protein with mass-to-charge ratio (m/z) 5000 was found to be responsible for the increased bacterial cytotoxicity, while monocytes' decreased adherence to biofilm was linked with the presence of proteins either with m/z 3330 or 2435. The results illustrate that monocytes' reaction when exposed to the bacterial biofilm can be used as an estimator of pathogens' virulence potential. The observed differences in monocytes' response are explainable by the bacterial proteins' profile. Additionally, the results indicate that the features of both bacteria and monocytes impact the outcome of the infection.


Subject(s)
Biofilms , Monocytes , Biofilms/growth & development , Monocytes/microbiology , Humans , Virulence , Bacterial Adhesion , Gram-Positive Bacterial Infections/microbiology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Enterococcus/pathogenicity , Poland , Feces/microbiology
4.
PLoS Pathog ; 20(5): e1012205, 2024 May.
Article in English | MEDLINE | ID: mdl-38701094

ABSTRACT

Mycobacterium tuberculosis (Mtb) infects lung myeloid cells, but the specific Mtb-permissive cells and host mechanisms supporting Mtb persistence during chronic infection are incompletely characterized. We report that after the development of T cell responses, CD11clo monocyte-derived cells harbor more live Mtb than alveolar macrophages (AM), neutrophils, and CD11chi monocyte-derived cells. Transcriptomic and functional studies revealed that the lysosome pathway is underexpressed in this highly permissive subset, characterized by less lysosome content, acidification, and proteolytic activity than AM, along with less nuclear TFEB, a regulator of lysosome biogenesis. Mtb infection does not drive lysosome deficiency in CD11clo monocyte-derived cells but promotes recruitment of monocytes that develop into permissive lung cells, mediated by the Mtb ESX-1 secretion system. The c-Abl tyrosine kinase inhibitor nilotinib activates TFEB and enhances lysosome functions of macrophages in vitro and in vivo, improving control of Mtb infection. Our results suggest that Mtb exploits lysosome-poor lung cells for persistence and targeting lysosome biogenesis is a potential host-directed therapy for tuberculosis.


Subject(s)
Lysosomes , Macrophages, Alveolar , Monocytes , Mycobacterium tuberculosis , Lysosomes/metabolism , Lysosomes/microbiology , Animals , Monocytes/metabolism , Monocytes/microbiology , Mice , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/metabolism , Lung/microbiology , Lung/metabolism , Mice, Inbred C57BL , Chronic Disease , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology , Humans , Tuberculosis/microbiology , Tuberculosis/immunology , Tuberculosis/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
5.
Microbiol Res ; 285: 127749, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38761490

ABSTRACT

Among 5 types of the Christie-Atkins-Munch-Petersen factor (CAMP) of Cutibacterium acnes, CAMP1 is highly expressed in phylotype II as well as IB, and thought to be a virulence factor of opportunistic but fatal blood, soft tissue, and implant-related infections. The target of a human single-chain variable antibody fragment (scFv), recently isolated from a phage display library, has been identified as CAMP1 of phylotype II, using immunoprecipitation followed by mass spectrometry, phage display peptide biopanning, 3D-modelling, and ELISA. The IgG1 format of the antibody could enhance phagocytosis of C. acnes DMST 14916 by THP-1 human monocytes. Our results suggest that the antibody-dependent phagocytosis process is mediated by the caveolae membrane system and involves the induction of IL-1ß. This is the first report on the study of a human antibody against CAMP1 of C. acnes phylotype II, of which a potential use as therapeutic antibody against virulence C. acnes infection is postulated.


Subject(s)
Immunoglobulin G , Macrophages , Phagocytosis , Humans , Macrophages/immunology , Macrophages/microbiology , Immunoglobulin G/immunology , Interleukin-1beta/metabolism , Interleukin-1beta/immunology , THP-1 Cells , Virulence Factors/immunology , Antibodies, Bacterial/immunology , Monocytes/immunology , Monocytes/microbiology , Single-Chain Antibodies/immunology , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Propionibacteriaceae/immunology
6.
Virulence ; 15(1): 2333367, 2024 12.
Article in English | MEDLINE | ID: mdl-38515333

ABSTRACT

Our immune system possesses sophisticated mechanisms to cope with invading microorganisms, while pathogens evolve strategies to deal with threats imposed by host immunity. Human plasma protein α1-antitrypsin (AAT) exhibits pleiotropic immune-modulating properties by both preventing immunopathology and improving antimicrobial host defence. Genetic associations suggested a role for AAT in candidemia, the most frequent fungal blood stream infection in intensive care units, yet little is known about how AAT influences interactions between Candida albicans and the immune system. Here, we show that AAT differentially impacts fungal killing by innate phagocytes. We observed that AAT induces fungal transcriptional reprogramming, associated with cell wall remodelling and downregulation of filamentation repressors. At low concentrations, the cell-wall remodelling induced by AAT increased immunogenic ß-glucan exposure and consequently improved fungal clearance by monocytes. Contrastingly, higher AAT concentrations led to excessive C. albicans filamentation and thus promoted fungal immune escape from monocytes and macrophages. This underscores that fungal adaptations to the host protein AAT can differentially define the outcome of encounters with innate immune cells, either contributing to improved immune recognition or fungal immune escape.


Subject(s)
Candida albicans , beta-Glucans , Humans , Candida albicans/metabolism , Host-Pathogen Interactions , Macrophages/microbiology , Monocytes/microbiology , beta-Glucans/metabolism
7.
J Immunol ; 211(5): 804-815, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37436030

ABSTRACT

Because of the growing numbers of immunocompromised patients, the incidence of life-threatening fungal infections caused by Candida albicans and Aspergillus fumigatus is increasing. We have recently identified enolase 1 (Eno1) from A. fumigatus as an immune evasion protein. Eno1 is a fungal moonlighting protein that mediates adhesion and invasion of human cells and also immune evasion through complement inactivation. We now show that soluble Eno1 has immunostimulatory activity. We observed that Eno1 from both C. albicans and A. fumigatus directly binds to the surface of lymphocytes, preferentially human and mouse B cells. Functionally, Eno1 upregulated CD86 expression on B cells and induced proliferation. Although the receptor for fungal Eno1 on B lymphocytes is still unknown, the comparison of B cells from wild-type and MyD88-deficient mice showed that B cell activation by Eno1 required MyD88 signaling. With respect to infection biology, we noted that mouse B cells stimulated by Eno1 secreted IgM and IgG2b. These Igs bound C. albicans hyphae in vitro, suggesting that Eno1-induced Ab secretion might contribute to protection from invasive fungal disease in vivo. Eno1 also triggered the release of proinflammatory cytokines from monocytes, particularly IL-6, which is a potent activator of B cells. Together, our data shed new light on the role of secreted Eno1 in infections with C. albicans and A. fumigatus. Eno1 secretion by these pathogenic microbes appears to be a double-edged sword by supporting fungal pathogenicity while triggering (antifungal) immunity.


Subject(s)
Aspergillus fumigatus , Candida albicans , Phosphopyruvate Hydratase , Animals , Humans , Mice , Aspergillus fumigatus/enzymology , Aspergillus fumigatus/metabolism , Candida albicans/enzymology , Candida albicans/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Monocytes/metabolism , Monocytes/microbiology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Phosphopyruvate Hydratase/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/microbiology
8.
Nat Microbiol ; 7(2): 312-326, 2022 02.
Article in English | MEDLINE | ID: mdl-35102304

ABSTRACT

Host cell chromatin changes are thought to play an important role in the pathogenesis of infectious diseases. Here we describe a histone acetylome-wide association study (HAWAS) of an infectious disease, on the basis of genome-wide H3K27 acetylation profiling of peripheral blood granulocytes and monocytes from persons with active Mycobacterium tuberculosis (Mtb) infection and healthy controls. We detected >2,000 differentially acetylated loci in either cell type in a Singapore Chinese discovery cohort (n = 46), which were validated in a subsequent multi-ethnic Singapore cohort (n = 29), as well as a longitudinal cohort from South Africa (n = 26), thus demonstrating that HAWAS can be independently corroborated. Acetylation changes were correlated with differential gene expression. Differential acetylation was enriched near potassium channel genes, including KCNJ15, which modulates apoptosis and promotes Mtb clearance in vitro. We performed histone acetylation quantitative trait locus (haQTL) analysis on the dataset and identified 69 candidate causal variants for immune phenotypes among granulocyte haQTLs and 83 among monocyte haQTLs. Our study provides proof-of-principle for HAWAS to infer mechanisms of host response to pathogens.


Subject(s)
Genetic Association Studies , Histones/genetics , Mycobacterium tuberculosis/immunology , Tuberculosis/genetics , Tuberculosis/immunology , Acetylation , Adult , Chromatin , Cohort Studies , Female , Granulocytes/immunology , Histones/immunology , Humans , Longitudinal Studies , Male , Monocytes/immunology , Monocytes/microbiology , Proof of Concept Study , Quantitative Trait Loci , Singapore , South Africa , THP-1 Cells , Tuberculosis/microbiology , Young Adult
9.
J Immunol ; 208(5): 1146-1154, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35110422

ABSTRACT

Porphyromonas gingivalis is commonly known as one of the major pathogens contributing to periodontitis, and its persistent infection may increase the risk for the disease. The proinflammatory mediators, including IL-6, TNF-α, and cyclooxygenase-2 (COX-2)/PGE2, are closely associated with progression of periodontitis. In this study, we focused on the cysteine protease "gingipains," lysine-specific gingipain, arginine-specific gingipain (Rgp) A, and RgpB, produced by P. gingivalis, and used the wild-type strain and several gene-deletion mutants (rgpA, rgpB, kgp, and fimA) to elucidate the involvement of gingipains in COX-2 expression and PGE2 production. We infected human monocytes, which are THP-1 cells and primary monocytes, with these bacterial strains and found that gingipains were involved in induction of COX-2 expression and PGE2 production. We have shown that the protease activity of gingipains was crucial for these events by using gingipain inhibitors. Furthermore, activation of ERK1/2 and IκB kinase was required for gingipain-induced COX-2 expression/PGE2 production, and these kinases activated two transcription factors, c-Jun/c-Fos (AP-1) and NF-κB p65, respectively. In particular, these data suggest that gingipain-induced c-Fos expression via ERK is essential for AP-1 formation with c-Jun, and activation of AP-1 and NF-κB p65 plays a central role in COX-2 expression/PGE2 production. Thus, we show the (to our knowledge) novel finding that gingipains with the protease activity from P. gingivalis induce COX-2 expression and PGE2 production via activation of MEK/ERK/AP-1 and IκB kinase/NF-κB p65 in human monocytes. Hence it is likely that gingipains closely contribute to the inflammation of periodontal tissues.


Subject(s)
Cyclooxygenase 2/biosynthesis , Dinoprostone/biosynthesis , Gingipain Cysteine Endopeptidases/metabolism , MAP Kinase Signaling System/physiology , Periodontitis/pathology , Porphyromonas gingivalis/metabolism , Bacterial Proteins/genetics , Cell Line , Cysteine Endopeptidases/genetics , Fimbriae Proteins/genetics , Gingipain Cysteine Endopeptidases/genetics , Humans , I-kappa B Kinase/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/microbiology , Periodontitis/microbiology , THP-1 Cells , Transcription Factor AP-1/metabolism , Transcription Factor RelA/metabolism
10.
PLoS Pathog ; 18(1): e1010176, 2022 01.
Article in English | MEDLINE | ID: mdl-35007290

ABSTRACT

COVID-19 displays diverse disease severities and symptoms including acute systemic inflammation and hypercytokinemia, with subsequent dysregulation of immune cells. Bacterial superinfections in COVID-19 can further complicate the disease course and are associated with increased mortality. However, there is limited understanding of how SARS-CoV-2 pathogenesis and hypercytokinemia impede the innate immune function against bacterial superinfections. We assessed the influence of COVID-19 plasma hypercytokinemia on the functional responses of myeloid immune cells upon bacterial challenges from acute-phase COVID-19 patients and their corresponding recovery-phase. We show that a severe hypercytokinemia status in COVID-19 patients correlates with the development of bacterial superinfections. Neutrophils and monocytes derived from COVID-19 patients in their acute-phase showed an impaired intracellular microbicidal capacity upon bacterial challenges. The impaired microbicidal capacity was reflected by abrogated MPO and reduced NETs production in neutrophils along with reduced ROS production in both neutrophils and monocytes. Moreover, we observed a distinct pattern of cell surface receptor expression on both neutrophils and monocytes, in line with suppressed autocrine and paracrine cytokine signaling. This phenotype was characterized by a high expression of CD66b, CXCR4 and low expression of CXCR1, CXCR2 and CD15 in neutrophils and low expression of HLA-DR, CD86 and high expression of CD163 and CD11b in monocytes. Furthermore, the impaired antibacterial effector function was mediated by synergistic effect of the cytokines TNF-α, IFN-γ and IL-4. COVID-19 patients receiving dexamethasone showed a significant reduction of overall inflammatory markers in the plasma as well as exhibited an enhanced immune response towards bacterial challenge ex vivo. Finally, broad anti-inflammatory treatment was associated with a reduction in CRP, IL-6 levels as well as length of ICU stay and ventilation-days in critically ill COVID-19 patients. Our data provides insights into the transient functional dysregulation of myeloid immune cells against subsequent bacterial infections in COVID-19 patients and describe a beneficial role for the use of dexamethasone in these patients.


Subject(s)
COVID-19/microbiology , Cytokine Release Syndrome/complications , Cytokines/metabolism , Monocytes/virology , Neutrophils/virology , COVID-19/virology , Cytokine Release Syndrome/microbiology , Cytokine Release Syndrome/virology , Humans , Lymphocytes/immunology , Lymphocytes/microbiology , Lymphocytes/virology , Monocytes/immunology , Monocytes/microbiology , Neutrophils/immunology , Neutrophils/microbiology , SARS-CoV-2/pathogenicity
11.
Gen Comp Endocrinol ; 315: 113796, 2022 01 01.
Article in English | MEDLINE | ID: mdl-33901496

ABSTRACT

Adrenocorticotropic hormone (ACTH), a bioactive peptide of the family of melanocortins, is generated from pro-opiomelanocortin (POMC). So far, the research on the specific functions of ACTH in the immune system of teleosts is limited. We determined two complementary DNA (cDNA) sequences of POMC in ayu (Plecoglossus altivelis), termed PaPOMC-A and PaPOMC-B. PaPOMCs transcripts occurred in all examined tissues, and their expression in immune tissues changed following experimental infection with Vibrio anguillarum. PaACTH-B, but not PaACTH-A, suppressed the phagocytosis of monocytes/macrophages (MO/MФ). Two isoforms of PaACTH increased the bactericidal capacity of MO/MФ. PaACTH-A increased anti-inflammatory cytokine expression, while PaACTH-B decreased pro-inflammatory cytokine expression in MO/MФ. Compared with PaACTH-B treatment, the PaACTH-A treatment improved survival rate and reduced the bacterial load in V. anguillarum-infected ayu through interleukin (IL)-10. Our results indicate that the two PaACTH isoforms exert different effects in the host defense against bacterial infection.


Subject(s)
Fish Diseases , Osmeriformes , Vibrio Infections , Vibrio , Adrenocorticotropic Hormone/metabolism , Adrenocorticotropic Hormone/pharmacology , Animals , Fish Diseases/genetics , Fish Proteins/genetics , Fish Proteins/metabolism , Macrophages/metabolism , Macrophages/microbiology , Monocytes/metabolism , Monocytes/microbiology , Osmeriformes/genetics , Osmeriformes/metabolism , Vibrio Infections/genetics , Vibrio Infections/microbiology
12.
Front Immunol ; 12: 734652, 2021.
Article in English | MEDLINE | ID: mdl-34867954

ABSTRACT

Microbial challenges, such as widespread bacterial infection in sepsis, induce endotoxin tolerance, a state of hyporesponsiveness to subsequent infections. The participation of DNA methylation in this process is poorly known. In this study, we perform integrated analysis of DNA methylation and transcriptional changes following in vitro exposure to gram-negative bacterial lipopolysaccharide, together with analysis of ex vivo monocytes from septic patients. We identify TET2-mediated demethylation and transcriptional activation of inflammation-related genes that is specific to toll-like receptor stimulation. Changes also involve phosphorylation of STAT1, STAT3 and STAT5, elements of the JAK2 pathway. JAK2 pathway inhibition impairs the activation of tolerized genes on the first encounter with lipopolysaccharide. We then confirm the implication of the JAK2-STAT pathway in the aberrant DNA methylome of patients with sepsis caused by gram-negative bacteria. Finally, JAK2 inhibition in monocytes partially recapitulates the expression changes produced in the immunosuppressive cellular state acquired by monocytes from gram-negative sepsis, as described by single cell-RNA-sequencing. Our study evidences both the crucial role the JAK2-STAT pathway in epigenetic regulation and initial response of the tolerized genes to gram-negative bacterial endotoxins and provides a pharmacological target to prevent exacerbated responses.


Subject(s)
Endotoxin Tolerance/genetics , Gram-Negative Bacteria/immunology , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/immunology , Monocytes/immunology , Monocytes/microbiology , Sepsis/genetics , Sepsis/immunology , Case-Control Studies , DNA Methylation/genetics , DNA Methylation/immunology , Endotoxin Tolerance/drug effects , Endotoxin Tolerance/immunology , Endotoxins/toxicity , Epigenesis, Genetic , Female , Gram-Negative Bacterial Infections/microbiology , Humans , In Vitro Techniques , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Lipopolysaccharides/toxicity , Male , Monocytes/drug effects , STAT Transcription Factors/genetics , STAT Transcription Factors/immunology , Sepsis/microbiology , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology
13.
Front Immunol ; 12: 750496, 2021.
Article in English | MEDLINE | ID: mdl-34867981

ABSTRACT

One of the main hallmarks of tuberculosis (TB) is the ability of the causative agent to transform into a stage of dormancy and the capability of long persistence in the host phagocytes. It is believed that approximately one-third of the population of the world is latently infected with Mycobacterium tuberculosis (Mtb), and 5%-10% of these individuals can develop clinical manifestations of active TB even decades after the initial infection. In this latent, intracellular form, the bacillus is shielded by an extremely robust cell wall and becomes phenotypically resistant to most antituberculars. Therefore, there is a clear rationale to develop novel compounds or carrier-conjugated constructs of existing drugs that are effective against the intracellular form of the bacilli. In this paper, we describe an experimental road map to define optimal candidates against intracellular Mtb and potential compounds effective in the therapy of latent TB. To validate our approach, isoniazid, a first-line antitubercular drug was employed, which is active against extracellular Mtb in the submicromolar range, but ineffective against the intracellular form of the bacteria. Cationic peptide conjugates of isoniazid were synthesized and employed to study the host-directed drug delivery. To measure the intracellular killing activity of the compounds, Mtb-infected MonoMac-6 human monocytic cells were utilized. We have assessed the antitubercular activity, cytotoxicity, membrane interactions in combination with internalization efficacy, localization, and penetration ability on interface and tissue-mimicking 3D models. Based on these in vitro data, most active compounds were further evaluated in vivo in a murine model of TB. Intraperitoneal infectious route was employed to induce a course of slowly progressive and systemic disease. The well-being of the animals, monitored by the body weight, allows a prolonged experimental setup and provides a great opportunity to test the long-term activity of the drug candidates. Having shown the great potency of this simple and suitable experimental design for antimicrobial research, the proposed novel assay platform could be used in the future to develop further innovative and highly effective antituberculars.


Subject(s)
Antimicrobial Peptides/administration & dosage , Antitubercular Agents/administration & dosage , Biological Assay/methods , Cell-Penetrating Peptides/administration & dosage , Isoniazid/administration & dosage , Mycobacterium tuberculosis/drug effects , Animals , Antimicrobial Peptides/chemistry , Antitubercular Agents/chemistry , Bronchi , Cell Line , Cell-Penetrating Peptides/chemistry , Endocytosis , Female , Humans , Isoniazid/chemistry , Mice, Inbred BALB C , Monocytes/microbiology , Mycobacterium tuberculosis/growth & development , Reproducibility of Results , Spheroids, Cellular , Tuberculosis/drug therapy
14.
mBio ; 12(6): e0215821, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34933448

ABSTRACT

Shigellosis causes most diarrheal deaths worldwide, particularly affecting children. Shigella invades and replicates in the epithelium of the large intestine, eliciting inflammation and tissue destruction. To understand how Shigella rewires macrophages prior to epithelium invasion, we performed genome-wide and focused secondary CRISPR knockout and CRISPR interference (CRISPRi) screens in Shigella flexneri-infected human monocytic THP-1 cells. Knockdown of the Toll-like receptor 1/2 signaling pathway significantly reduced proinflammatory cytokine and chemokine production, enhanced host cell survival, and controlled intracellular pathogen growth. Knockdown of the enzymatic component of the mitochondrial pyruvate dehydrogenase complex enhanced THP-1 cell survival. Small-molecule inhibitors blocking key components of these pathways had similar effects; these were validated with human monocyte-derived macrophages, which closely mimic the in vivo physiological state of macrophages postinfection. High-throughput CRISPR screens can elucidate how S. flexneri triggers inflammation and redirects host pyruvate catabolism for energy acquisition before killing macrophages, pointing to new shigellosis therapies. IMPORTANCE Treatment for shigellosis is becoming increasingly difficult as resistance to antibiotics becomes more prevalent. One way to prevent this significant public health problem from developing into a full-blown crisis is to approach shigellosis intervention from the point of view of the host. So far, little is known about the specific biological pathways that might be modulated in macrophages, sentinel cells of the innate immune system, to strengthen the response to Shigella infection. In this work, we conducted CRISPR screens to comprehensively decipher the complexity of macrophage-Shigella interactions and to discover new potential therapeutic interventions against Shigella flexneri infection. Our work highlights systematic genetic perturbation strategies to provide direct causal evidence showing how intracellular pathogens manipulate innate immune cells.


Subject(s)
Dysentery, Bacillary/genetics , Dysentery, Bacillary/microbiology , Macrophages/microbiology , Shigella flexneri/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Cytokines/genetics , Cytokines/immunology , Dysentery, Bacillary/immunology , High-Throughput Nucleotide Sequencing , Humans , Macrophages/immunology , Monocytes/immunology , Monocytes/microbiology , Shigella flexneri/physiology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology
15.
Front Immunol ; 12: 751683, 2021.
Article in English | MEDLINE | ID: mdl-34804037

ABSTRACT

Innate immune memory, the ability of innate cells to react in a more protective way to secondary challenges, is induced by exposure to infectious and other exogeous and endogenous agents. Engineered nanoparticles are particulate exogenous agents that, as such, could trigger an inflammatory reaction in monocytes and macrophages and could therefore be also able to induce innate memory. Here, we have evaluated the capacity of engineered gold nanoparticles (AuNPs) to induce a memory response or to modulate the memory responses induced by microbial agents. Microbial agents used were in soluble vs. particulate form (MDP and the gram-positive bacteria Staphylococcus aureus; ß-glucan and the ß-glucan-producing fungi C. albicans), and as whole microrganisms that were either killed (S. aureus, C. albicans) or viable (the gram-negative bacteria Helicobacter pylori). The memory response was assessed in vitro, by exposing human primary monocytes from 2-7 individual donors to microbial agents with or without AuNPs (primary response), then resting them for 6 days to allow return to baseline, and eventually challenging them with LPS (secondary memory response). Primary and memory responses were tested as production of the innate/inflammatory cytokine TNFα and other inflammatory and anti-inflammatory factors. While inactive on the response induced by soluble microbial stimuli (muramyl dipeptide -MDP-, ß-glucan), AuNPs partially reduced the primary response induced by whole microorganisms. AuNPs were also unable to directly induce a memory response but could modulate stimulus-induced memory in a circumscribed fashion, limited to some agents and some cytokines. Thus, the MDP-induced tolerance in terms of TNFα production was further exacerbated by co-priming with AuNPs, resulting in a less inflammatory memory response. Conversely, the H. pylori-induced tolerance was downregulated by AuNPs only relative to the anti-inflammatory cytokine IL-10, which would lead to an overall more inflammatory memory response. These effects of AuNPs may depend on a differential interaction/association between the reactive particle surfaces and the microbial components and agents, which may lead to a change in the exposure profiles. As a general observation, however, the donor-to-donor variability in memory response profiles and reactivity to AuNPs was substantial, suggesting that innate memory depends on the individual history of exposures.


Subject(s)
Candida albicans , Gold/administration & dosage , Helicobacter pylori , Immunologic Memory/drug effects , Metal Nanoparticles/administration & dosage , Monocytes/drug effects , Staphylococcus aureus , beta-Glucans/pharmacology , Cells, Cultured , Cytokines/immunology , Humans , Immunity, Innate/drug effects , Monocytes/immunology , Monocytes/microbiology
16.
Dis Markers ; 2021: 4537760, 2021.
Article in English | MEDLINE | ID: mdl-34691286

ABSTRACT

BACKGROUND: Sepsis is a critical medical condition that requires additional diagnostic considerations. Recently, focus has shifted to the diagnosis of sepsis using new markers to overcome the limitations of traditional laboratory diagnostic modalities. Neutrophil CD11b (nCD11b) and monocyteCD14 (mCD14) cell surface antigens have been shown to be useful in such diagnostic consideration. AIM: To investigate the diagnostic, monitoring, prognostic, and predictive roles of nCD11b and mCD14 as sepsis biomarkers in comparison to each other and to traditional laboratory sepsis parameters in order to select the best fit for routine daily use in neonatal intensive care units (NICUs). SUBJECT: The study included 188 neonates from Ain Shams University Hospitals' NICUs, who were divided into two groups: the control group (n = 100) and the sepsis group (n = 88). Highly sensitive CRP (hs-CRP), complete blood count (CBC), blood culture, and nCD11b and mCD14 evaluations were all part of the laboratory sepsis evaluation (done by flow cytometry technology). Positive blood culture results (BACT/ALERT system) confirmed the sepsis diagnosis. Twenty-four enrolled sepsis neonates were subjected to follow-up assessments, and they were divided into two groups based on clinical improvement: improved sepsis and sepsis without improvement. In order to predict performance evaluation, the subjected neonates were reclassified according to their outcome into survivors' and nonsurvivors' group. RESULTS: Sepsis patients had a significant increase in mCD14 MFI values when compared to controls. With sensitivity 75.4 percent, specificity 71.9 percent, efficacy 73.3 percent, and AUC 0.703, mCD14 MFI at cutoff 9.36 could distinguish the presence of septicemia. Significant increases in both mCD14 MFI and nCD11b MFI (P = 0.001) were observed in the severe sepsis/septic shock group compared to the nonsevere sepsis group. The combined measurement of CD14 MFI at cutoff 9.97 and CD14 percent at cutoff 44.7 percent yielded the best predictive performance. CONCLUSION: Sepsis patients had a significant increase in mCD14 MFI comparable to the controls. mCD14 MFI demonstrated better diagnostic, prognostic, and predictive results than nCD11b. hs-CRP outperformed mCD14 and nCD11b in terms of diagnostic efficacy and AUC. In the monitoring of sepsis patients, both mCD14 and nCD11b produced unsatisfactory results. Currently, the routine use of mCD14 or nCD11b as sepsis biomarkers in neonatal ICUs is not justified.


Subject(s)
Monocytes , Neonatal Sepsis/blood , Neonatal Sepsis/mortality , Neutrophils , Area Under Curve , Biomarkers/blood , CD11b Antigen/metabolism , Case-Control Studies , Female , Humans , Infant , Intensive Care Units, Neonatal , Lipopolysaccharide Receptors/metabolism , Male , Monocytes/metabolism , Monocytes/microbiology , Neonatal Sepsis/microbiology , Neutrophils/metabolism , Neutrophils/microbiology , Prognosis
17.
Front Immunol ; 12: 695227, 2021.
Article in English | MEDLINE | ID: mdl-34484192

ABSTRACT

Aims: Periodontitis is an independent risk factor for cardiovascular disease, but the mechanistic link is not fully understood. In atherosclerotic cardiovascular disease, monocytes can adopt a persistent hyperresponsive phenotype, termed trained immunity. We hypothesized that periodontitis-associated bacteria can induce trained immunity in monocytes, which subsequently accelerate atherosclerosis development. Materials and Methods: We combined in vitro experiments on human primary monocytes and in vivo techniques in patients with periodontitis to test this hypothesis. Adherent peripheral blood mononuclear cells (PBMCs) were transiently exposed in vitro to Porphyromonas gingivalis for 24 hours, and restimulated with lipopolysaccharide (LPS) or Pam3CysK4 (P3C) six days later, to measure interleukin-6 (IL-6) and tumor necrosis factor α (TNFα) production. In an exploratory observational study, patients with severe periodontitis (63 ± 6 years, n=14) and control subjects with no-to-mild periodontitis (54 ± 10 years, n=14) underwent venipuncture and 2'-deoxy-2'-[18F]fluoro-D-glucose positron-emission-tomography ([18F]FDG PET/CT) scanning. Results: When adherent peripheral blood mononuclear cells (PBMCs) were transiently exposed in vitro to Porphyromonas gingivalis for 24 hours, and restimulated with LPS or P3C six days later, IL-6 and TNFα production was significantly increased (TNFα/P3C, p<0.01). Circulating leukocytes, IL-6 and interleukin-1 receptor antagonist (IL-1Ra) concentrations were generally higher in patients compared to controls (leukocytes: p<0.01; IL-6: p=0.08; IL-1Ra: p=0.10). Cytokine production capacity in PBMCs after 24h stimulation revealed no differences between groups. [18F]FDG PET/CT imaging showed a trend for increased [18F]FDG-uptake in the periodontium [mean standard uptake value (SUVmean), p=0.11] and in femur bone marrow (SUVmean, p=0.06), but no differences were observed for vascular inflammation. Positive correlations between severity of periodontitis, measured by The Dutch Periodontal Screening Index and pocket depth, with circulating inflammatory markers and tissue inflammation were found. Conclusions: P. gingivalis induces long-term activation of human monocytes in vitro (trained immunity). Patients with severe periodontitis did have signs of increased systemic inflammation and hematopoietic tissue activation. However, their circulating monocytes did not show a hyperresponsive phenotype. Together we suggest that trained immunity might contribute to local periodontal inflammation which warrants further investigation.


Subject(s)
Atherosclerosis/immunology , Cytokines/metabolism , Inflammation Mediators/metabolism , Monocytes/immunology , Periodontitis/immunology , Porphyromonas gingivalis/immunology , Adult , Aged , Aged, 80 and over , Atherosclerosis/diagnostic imaging , Atherosclerosis/metabolism , Atherosclerosis/microbiology , Case-Control Studies , Cells, Cultured , Female , Host-Pathogen Interactions , Humans , Lipopeptides/pharmacology , Lipopolysaccharides/pharmacology , Male , Middle Aged , Monocytes/drug effects , Monocytes/metabolism , Monocytes/microbiology , Periodontitis/diagnostic imaging , Periodontitis/metabolism , Periodontitis/microbiology , Phenotype , Porphyromonas gingivalis/pathogenicity , Positron Emission Tomography Computed Tomography , Risk Assessment , Risk Factors , Severity of Illness Index
18.
Front Cell Infect Microbiol ; 11: 711144, 2021.
Article in English | MEDLINE | ID: mdl-34350134

ABSTRACT

Most bacteria naturally release spherical lipid-bilayered extracellular vesicles (EVs) containing proteins, nucleic acids, and virulence-related molecules, thus contributing to diverse biological functions including transport of virulence factors. The group A streptococcus, Streptococcus pyogenes (GAS), a major human pathogen, also releases EVs; however, it remains unclear how GAS EVs interact physiologically and pathologically with host cells, and what the differences are between invasive and non-invasive strains. The proteome profile in this study revealed that GAS EVs enclosed many virulence-related proteins such as streptolysin O and NAD-glycohydrolase, facilitating their pathogenicity, and invasive GAS EVs were more abundant than non-invasive counterparts. In terms of biological effects, invasive GAS EVs showed slo-dependent cytotoxic activity and the induction of cytokine expression, contributing to GAS pathogenicity directly. Although non-invasive GAS EVs did not show cytotoxic activity, they may be utilized as a means to prevent antibacterial mechanisms such as autophagy, leading to enhancement of their own survival in the intracellular environment after the infection. These results suggest that invasive and non-invasive GAS EVs play different roles in GAS infection strategy and pathogenicity. Our findings also indicate that EVs could be a key factor for GAS pathogenicity in GAS-host interactions.


Subject(s)
Extracellular Vesicles , Monocytes/microbiology , Streptococcus pyogenes , Bacterial Proteins , Humans , Inflammation , NAD+ Nucleosidase , Virulence , Virulence Factors
19.
Front Immunol ; 12: 712678, 2021.
Article in English | MEDLINE | ID: mdl-34413857

ABSTRACT

Mycobacterium tuberculosis (Mtb), the pathological agent that causes tuberculosis (TB) is the number one infectious killer worldwide with one fourth of the world's population currently infected. Data indicate that γ9δ2 T cells secrete Granzyme A (GzmA) in the extracellular space triggering the infected monocyte to inhibit growth of intracellular mycobacteria. Accordingly, deletion of GZMA from γ9δ2 T cells reverses their inhibitory capacity. Through mechanistic studies, GzmA's action was investigated in monocytes from human PBMCs. The use of recombinant human GzmA expressed in a mammalian system induced inhibition of intracellular mycobacteria to the same degree as previous human native protein findings. Our data indicate that: 1) GzmA is internalized within mycobacteria-infected cells, suggesting that GzmA uptake could prevent infection and 2) that the active site is not required to inhibit intracellular replication. Global proteomic analysis demonstrated that the ER stress response and ATP producing proteins were upregulated after GzmA treatment, and these proteins abundancies were confirmed by examining their expression in an independent set of patient samples. Our data suggest that immunotherapeutic host interventions of these pathways may contribute to better control of the current TB epidemic.


Subject(s)
Adenosine Triphosphate/biosynthesis , Endoplasmic Reticulum Stress/immunology , Granzymes/physiology , Monocytes/microbiology , Mycobacterium bovis/physiology , T-Lymphocyte Subsets/immunology , Blotting, Western , Cell Division , Granzymes/biosynthesis , Granzymes/genetics , Granzymes/pharmacology , HEK293 Cells , Humans , Memory T Cells/immunology , Memory T Cells/metabolism , Proteome , Receptors, Antigen, T-Cell, gamma-delta/analysis , Recombinant Proteins/pharmacology , T-Lymphocyte Subsets/metabolism , Two-Dimensional Difference Gel Electrophoresis
20.
Gut Microbes ; 13(1): 1939598, 2021.
Article in English | MEDLINE | ID: mdl-34224309

ABSTRACT

Gut microbiota is a constant source of antigens and stimuli to which the resident immune system has developed tolerance. However, the mechanisms by which mononuclear phagocytes, specifically monocytes/macrophages, cope with these usually pro-inflammatory signals are poorly understood. Here, we show that innate immune memory promotes anti-inflammatory homeostasis, using as model strains of the commensal bacterium Lactiplantibacillus plantarum. Priming of monocytes/macrophages with bacteria, especially in its live form, enhances bacterial intracellular survival and decreases the release of pro-inflammatory signals to the environment, with lower production of TNF and higher levels of IL-10. Analysis of the transcriptomic landscape of these cells shows downregulation of pathways associated with the production of reactive oxygen species (ROS) and the release of cytokines, chemokines and antimicrobial peptides. Indeed, the induction of ROS prevents memory-induced bacterial survival. In addition, there is a dysregulation in gene expression of several metabolic pathways leading to decreased glycolytic and respiratory rates in memory cells. These data support commensal microbe-specific metabolic changes in innate immune memory cells that might contribute to homeostasis in the gut.


Subject(s)
Immunity, Innate , Lactobacillaceae/immunology , Macrophages/immunology , Monocytes/immunology , Adult , Aged , Animals , Antimicrobial Peptides/immunology , Female , Humans , Immunologic Memory , Interleukin-10/immunology , Macrophages/microbiology , Male , Mice , Microbiota , Middle Aged , Monocytes/microbiology , RAW 264.7 Cells , Saliva/microbiology , Symbiosis
SELECTION OF CITATIONS
SEARCH DETAIL