Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.215
Filter
1.
Front Cell Infect Microbiol ; 14: 1346087, 2024.
Article in English | MEDLINE | ID: mdl-38736751

ABSTRACT

Epithelial cells form a resilient barrier and orchestrate defensive and reparative mechanisms to maintain tissue stability. This review focuses on gut and airway epithelia, which are positioned where the body interfaces with the outside world. We review the many signaling pathways and mechanisms by which epithelial cells at the interface respond to invading pathogens to mount an innate immune response and initiate adaptive immunity and communicate with other cells, including resident microbiota, to heal damaged tissue and maintain homeostasis. We compare and contrast how airway and gut epithelial cells detect pathogens, release antimicrobial effectors, collaborate with macrophages, Tregs and epithelial stem cells to mount an immune response and orchestrate tissue repair. We also describe advanced research models for studying epithelial communication and behaviors during inflammation, tissue injury and disease.


Subject(s)
Homeostasis , Immunity, Innate , Intestinal Mucosa , Humans , Intestinal Mucosa/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Animals , Respiratory Mucosa/microbiology , Respiratory Mucosa/immunology , Epithelial Cells/microbiology , Signal Transduction , Adaptive Immunity , Macrophages/immunology , Macrophages/microbiology , Host-Pathogen Interactions
2.
PLoS Genet ; 20(5): e1011281, 2024 May.
Article in English | MEDLINE | ID: mdl-38743788

ABSTRACT

CgHog1, terminal kinase of the high-osmolarity glycerol signalling pathway, orchestrates cellular response to multiple external stimuli including surplus-environmental iron in the human fungal pathogen Candida glabrata (Cg). However, CgHog1 substrates remain unidentified. Here, we show that CgHog1 adversely affects Cg adherence to host stomach and kidney epithelial cells in vitro, but promotes Cg survival in the iron-rich gastrointestinal tract niche. Further, CgHog1 interactome and in vitro phosphorylation analysis revealed CgSub2 (putative RNA helicase) to be a CgHog1 substrate, with CgSub2 also governing iron homeostasis and host adhesion. CgSub2 positively regulated EPA1 (encodes a major adhesin) expression and host adherence via its interactor CgHtz1 (histone H2A variant). Notably, both CgHog1 and surplus environmental iron had a negative impact on CgSub2-CgHtz1 interaction, with CgHTZ1 or CgSUB2 deletion reversing the elevated adherence of Cghog1Δ to epithelial cells. Finally, the surplus-extracellular iron led to CgHog1 activation, increased CgSub2 phosphorylation, elevated CgSub2-CgHta (canonical histone H2A) interaction, and EPA1 transcriptional activation, thereby underscoring the iron-responsive, CgHog1-induced exchange of histone partners of CgSub2. Altogether, our work mechanistically defines how CgHog1 couples Epa1 adhesin expression with iron abundance, and point towards specific chromatin composition modification programs that probably aid fungal pathogens align their adherence to iron-rich (gut) and iron-poor (blood) host niches.


Subject(s)
Candida glabrata , Cell Adhesion , Epithelial Cells , Fungal Proteins , Histones , Candida glabrata/genetics , Candida glabrata/metabolism , Humans , Histones/metabolism , Histones/genetics , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Cell Adhesion/genetics , Fungal Proteins/metabolism , Fungal Proteins/genetics , Phosphorylation , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/genetics , Iron/metabolism , Gene Expression Regulation, Fungal , Candidiasis/microbiology , Candidiasis/genetics , Signal Transduction
3.
PLoS Pathog ; 20(5): e1012214, 2024 May.
Article in English | MEDLINE | ID: mdl-38722857

ABSTRACT

Epithelial cells function as the primary line of defense against invading pathogens. However, bacterial pathogens possess the ability to compromise this barrier and facilitate the transmigration of bacteria. Nonetheless, the specific molecular mechanism employed by Mycobacterium tuberculosis (M.tb) in this process is not fully understood. Here, we investigated the role of Rv2569c in M.tb translocation by assessing its ability to cleave E-cadherin, a crucial component of cell-cell adhesion junctions that are disrupted during bacterial invasion. By utilizing recombinant Rv2569c expressed in Escherichia coli and subsequently purified through affinity chromatography, we demonstrated that Rv2569c exhibited cell wall-associated serine protease activity. Furthermore, Rv2569c was capable of degrading a range of protein substrates, including casein, fibrinogen, fibronectin, and E-cadherin. We also determined that the optimal conditions for the protease activity of Rv2569c occurred at a temperature of 37°C and a pH of 9.0, in the presence of MgCl2. To investigate the function of Rv2569c in M.tb, a deletion mutant of Rv2569c and its complemented strains were generated and used to infect A549 cells and mice. The results of the A549-cell infection experiments revealed that Rv2569c had the ability to cleave E-cadherin and facilitate the transmigration of M.tb through polarized A549 epithelial cell layers. Furthermore, in vivo infection assays demonstrated that Rv2569c could disrupt E-cadherin, enhance the colonization of M.tb, and induce pathological damage in the lungs of C57BL/6 mice. Collectively, these results strongly suggest that M.tb employs the serine protease Rv2569c to disrupt epithelial defenses and facilitate its systemic dissemination by crossing the epithelial barrier.


Subject(s)
Bacterial Proteins , Cadherins , Epithelial Cells , Mycobacterium tuberculosis , Serine Proteases , Cadherins/metabolism , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/metabolism , Animals , Humans , Mice , Serine Proteases/metabolism , Serine Proteases/genetics , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , A549 Cells , Tuberculosis/microbiology , Tuberculosis/metabolism , Female
4.
Nat Commun ; 15(1): 3666, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38693120

ABSTRACT

Respiratory viral infection increases host susceptibility to secondary bacterial infections, yet the precise dynamics within airway epithelia remain elusive. Here, we elucidate the pivotal role of CD47 in the airway epithelium during bacterial super-infection. We demonstrated that upon influenza virus infection, CD47 expression was upregulated and localized on the apical surface of ciliated cells within primary human nasal or bronchial epithelial cells. This induced CD47 exposure provided attachment sites for Staphylococcus aureus, thereby compromising the epithelial barrier integrity. Through bacterial adhesion assays and in vitro pull-down assays, we identified fibronectin-binding proteins (FnBP) of S. aureus as a key component that binds to CD47. Furthermore, we found that ciliated cell-specific CD47 deficiency or neutralizing antibody-mediated CD47 inactivation enhanced in vivo survival rates. These findings suggest that interfering with the interaction between airway epithelial CD47 and pathogenic bacterial FnBP holds promise for alleviating the adverse effects of super-infection.


Subject(s)
CD47 Antigen , Epithelial Cells , Staphylococcal Infections , Staphylococcus aureus , Superinfection , CD47 Antigen/metabolism , CD47 Antigen/genetics , Humans , Animals , Superinfection/microbiology , Mice , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/virology , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Influenza, Human/metabolism , Influenza, Human/immunology , Influenza, Human/virology , Bacterial Adhesion , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Respiratory Mucosa/virology , Mice, Inbred C57BL , Bronchi/metabolism , Bronchi/cytology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Mice, Knockout , Influenza A Virus, H1N1 Subtype
5.
Front Cell Infect Microbiol ; 14: 1397940, 2024.
Article in English | MEDLINE | ID: mdl-38751999

ABSTRACT

Non-typeable Haemophilus influenzae (NTHi) and Moraxella catarrhalis (Mcat) are two common respiratory tract pathogens often associated with acute exacerbations in Chronic Obstructive Pulmonary Disease (COPD) as well as with otitis media (OM) in children. Although there is evidence that these pathogens can adopt persistence mechanisms such as biofilm formation, the precise means through which they contribute to disease severity and chronicity remains incompletely understood, posing challenges for their effective eradication. The identification of potential vaccine candidates frequently entails the characterization of the host-pathogen interplay in vitro even though this approach is limited by the fact that conventional models do not permit long term bacterial infections. In the present work, by using air-liquid-interface (ALI) human airway in vitro models, we aimed to recreate COPD-related persistent bacterial infections. In particular, we explored an alternative use of the ALI system consisting in the assembly of an inverted epithelium grown on the basal part of a transwell membrane with the aim to enable the functionality of natural defense mechanisms such as mucociliary clearance and cellular extrusion that are usually hampered during conventional ALI infection experiments. The inversion of the epithelium did not affect tissue differentiation and considerably delayed NTHi or Mcat infection progression, allowing one to monitor host-pathogen interactions for up to three weeks. Notably, the use of these models, coupled with confocal and transmission electron microscopy, revealed unique features associated with NTHi and Mcat infection, highlighting persistence strategies including the formation of intracellular bacterial communities (IBCs) and surface-associated biofilm-like structures. Overall, this study demonstrates the possibility to perform long term host-pathogen investigations in vitro with the aim to define persistence mechanisms adopted by respiratory pathogens and individuate potential new vaccine targets.


Subject(s)
Biofilms , Haemophilus influenzae , Moraxella catarrhalis , Moraxellaceae Infections , Moraxella catarrhalis/physiology , Humans , Haemophilus influenzae/physiology , Haemophilus influenzae/pathogenicity , Biofilms/growth & development , Moraxellaceae Infections/microbiology , Persistent Infection/microbiology , Host-Pathogen Interactions , Haemophilus Infections/microbiology , Pulmonary Disease, Chronic Obstructive/microbiology , Models, Biological , Respiratory Tract Infections/microbiology , Epithelial Cells/microbiology
6.
Front Cell Infect Microbiol ; 14: 1389527, 2024.
Article in English | MEDLINE | ID: mdl-38756230

ABSTRACT

Neisseria meningitidis (Nm, the meningococcus) is considered an asymptomatic colonizer of the upper respiratory tract and a transient member of its microbiome. It is assumed that the spread of N. meningitidis into the bloodstream occurs via transcytosis of the nasopharyngeal epithelial barrier without destroying the barrier layer. Here, we used Calu-3 respiratory epithelial cells that were grown under air-liquid-interface conditions to induce formation of pseudostratified layers and mucus production. The number of bacterial localizations in the outer mucus, as well as cellular adhesion, invasion and transmigration of different carrier and disease N. meningitidis isolates belonging to MenB:cc32 and MenW:cc22 lineages was assessed. In addition, the effect on barrier integrity and cytokine release was determined. Our findings showed that all strains tested resided primarily in the outer mucus layer after 24 h of infection (>80%). Nonetheless, both MenB:cc32 and MenW:cc22 carrier and disease isolates reached the surface of the epithelial cells and overcame the barrier. Interestingly, we observed a significant difference in the number of bacteria transmigrating the epithelial cell barrier, with the representative disease isolates being more efficient to transmigrate compared to carrier isolates. This could be attributed to the capacity of the disease isolates to invade, however could not be assigned to expression of the outer membrane protein Opc. Moreover, we found that the representative meningococcal isolates tested in this study did not damage the epithelial barrier, as shown by TEER measurement, FITC-dextran permeability assays, and expression of cell-junction components.


Subject(s)
Bacterial Adhesion , Carrier State , Epithelial Cells , Meningococcal Infections , Nasopharynx , Neisseria meningitidis , Epithelial Cells/microbiology , Humans , Nasopharynx/microbiology , Neisseria meningitidis/metabolism , Meningococcal Infections/microbiology , Carrier State/microbiology , Cell Line , Cytokines/metabolism
7.
PLoS One ; 19(5): e0303449, 2024.
Article in English | MEDLINE | ID: mdl-38768097

ABSTRACT

Candida albicans (C. albicans) can behave as a commensal yeast colonizing the vaginal mucosa, and in this condition is tolerated by the epithelium. When the epithelial tolerance breaks down, due to C. albicans overgrowth and hyphae formation, the generated inflammatory response and cell damage lead to vulvovaginal candidiasis (VVC) symptoms. Here, we focused on the induction of mitochondrial reactive oxygen species (mtROS) in vaginal epithelial cells after C. albicans infection and the involvement of fungal burden, morphogenesis and candidalysin (CL) production in such induction. Bioluminescent (BLI) C. albicans, C. albicans PCA-2 and C. albicans 529L strains were employed in an in vitro infection model including reconstituted vaginal epithelium cells (RVE), produced starting from A-431 cell line. The production of mtROS was kinetically measured by using MitoSOX™ Red probe. The potency of C. albicans to induced cell damage to RVE and C. albicans proliferation have also been evaluated. C. albicans induces a rapid mtROS release from vaginal epithelial cells, in parallel with an increase of the fungal load and hyphal formation. Under the same experimental conditions, the 529L C. albicans strain, known to be defective in CL production, induced a minor mtROS release showing the key role of CL in causing epithelial mithocondrial activation. C. albicans PCA-2, unable to form hyphae, induced comparable but slower mtROS production as compared to BLI C. albicans yeasts. By reducing mtROS through a ROS scavenger, an increased fungal burden was observed during RVE infection but not in fungal cultures grown on abiotic surface. Collectively, we conclude that CL, more than fungal load and hyphae formation, seems to play a key role in the rapid activation of mtROS by epithelial cells and in the induction of cell-damage and that mtROS are key elements in the vaginal epithelial cells response to C. albicans.


Subject(s)
Candida albicans , Candidiasis, Vulvovaginal , Epithelial Cells , Fungal Proteins , Mitochondria , Reactive Oxygen Species , Vagina , Candida albicans/metabolism , Candida albicans/physiology , Female , Humans , Mitochondria/metabolism , Vagina/microbiology , Reactive Oxygen Species/metabolism , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Fungal Proteins/metabolism , Candidiasis, Vulvovaginal/microbiology , Hyphae/metabolism , Hyphae/growth & development , Cell Line
8.
Front Cell Infect Microbiol ; 14: 1391758, 2024.
Article in English | MEDLINE | ID: mdl-38716194

ABSTRACT

Campylobacter jejuni, a Gram-negative bacterium, is one of the most common causes of foodborne illness worldwide. Its adhesion mechanism is mediated by several bacterial factors, including flagellum, protein adhesins, lipooligosaccharides, proteases, and host factors, such as surface glycans on epithelial cells and mucins. Fungal lectins, specialized carbohydrate-binding proteins, can bind to specific glycans on host and bacterial cells and thus influence pathogenesis. In this study, we investigated the effects of fungal lectins and protease inhibitors on the adhesion of C. jejuni to model biotic surfaces (mucin, fibronectin, and collagen) and Caco-2 cells as well as the invasion of Caco-2 cells. The lectins Marasmius oreades agglutinin (MOA) and Laccaria bicolor tectonin 2 (Tec2) showed remarkable efficacy in all experiments. In addition, different pre-incubations of lectins with C. jejuni or Caco-2 cells significantly inhibited the ability of C. jejuni to adhere to and invade Caco-2 cells, but to varying degrees. Pre-incubation of Caco-2 cells with selected lectins reduced the number of invasive C. jejuni cells the most, while simultaneous incubation showed the greatest reduction in adherent C. jejuni cells. These results suggest that fungal lectins are a promising tool for the prevention and treatment of C. jejuni infections. Furthermore, this study highlights the potential of fungi as a rich reservoir for novel anti-adhesive agents.


Subject(s)
Bacterial Adhesion , Campylobacter jejuni , Lectins , Protease Inhibitors , Campylobacter jejuni/drug effects , Campylobacter jejuni/physiology , Campylobacter jejuni/metabolism , Humans , Caco-2 Cells , Bacterial Adhesion/drug effects , Lectins/metabolism , Lectins/pharmacology , Protease Inhibitors/pharmacology , Protease Inhibitors/metabolism , Fungi/drug effects , Mucins/metabolism , Epithelial Cells/microbiology , Fibronectins/metabolism
9.
J Med Invest ; 71(1.2): 102-112, 2024.
Article in English | MEDLINE | ID: mdl-38735705

ABSTRACT

Vibrio vulnificus (V. vulnificus) is a halophilic gram-negative bacterium that inhabits coastal warm water and induce severe diseases such as primary septicemia. To investigate the mechanisms of rapid bacterial translocation on intestinal infection, we focused on outer membrane vesicles (OMVs), which are extracellular vesicles produced by Gram-negative bacteria and deliver virulence factors. However, there are very few studies on the pathogenicity or contents of V. vulnificus OMVs (Vv-OMVs). In this study, we investigated the effects of Vv-OMVs on host cells. Epithelial cells INT407 were stimulated with purified OMVs and morphological alterations and levels of lactate dehydrogenase (LDH) release were observed. In cells treated with OMVs, cell detachment without LDH release was observed, which exhibited different characteristics from cytotoxic cell detachment observed in V. vulnificus infection. Interestingly, OMVs from a Vibrio Vulnificus Hemolysin (VVH) and Multifunctional-autoprocessing repeats-in -toxin (MARTX) double-deletion mutant strain also caused cell detachment without LDH release. Our results suggested that the proteolytic function of a serine protease contained in Vv-OMVs may contribute to pathogenicity of V. vulnificus by assisting bacterial translocation. This study reveals a new pathogenic mechanism during V. vulnificus infections. J. Med. Invest. 71 : 102-112, February, 2024.


Subject(s)
Extracellular Vesicles , Vibrio vulnificus , Vibrio vulnificus/pathogenicity , Vibrio vulnificus/metabolism , Humans , Extracellular Vesicles/metabolism , Hemolysin Proteins/metabolism , L-Lactate Dehydrogenase/metabolism , Bacterial Outer Membrane/metabolism , Epithelial Cells/microbiology
10.
Virulence ; 15(1): 2357670, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38804638

ABSTRACT

Salmonella enterica subspecies enterica serovar Typhimurium is an intracellular pathogen that invades and colonizes the intestinal epithelium. Following bacterial invasion, Salmonella is enclosed within a membrane-bound vacuole known as a Salmonella-containing vacuole (SCV). However, a subset of Salmonella has the capability to prematurely rupture the SCV and escape, resulting in Salmonella hyper-replication within the cytosol of epithelial cells. A recently published RNA-seq study provides an overview of cytosolic and vacuolar upregulated genes and highlights pagN vacuolar upregulation. Here, using transcription kinetics, protein production profile, and immunofluorescence microscopy, we showed that PagN is exclusively produced by Salmonella in SCV. Gentamicin protection and chloroquine resistance assays were performed to demonstrate that deletion of pagN affects Salmonella replication by affecting the cytosolic bacterial population. This study presents the first example of a Salmonella virulence factor expressed within the endocytic compartment, which has a significant impact on the dynamics of Salmonella cytosolic hyper-replication.


Subject(s)
Bacterial Proteins , Cytosol , Salmonella typhimurium , Vacuoles , Virulence Factors , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity , Cytosol/microbiology , Vacuoles/microbiology , Vacuoles/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Humans , Virulence , Salmonella Infections/microbiology , HeLa Cells , Epithelial Cells/microbiology , Gene Expression Regulation, Bacterial
11.
Microbiol Res ; 284: 127727, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636241

ABSTRACT

Heme oxygenase HO-1 (HMOX) regulates cellular inflammation and apoptosis, but its role in regulation of autophagy in Mycoplasma bovis infection is unknown. The objective was to determine how the HO-1/CO- Protein kinase RNA-like endoplasmic reticulum kinase (PERK)-Ca2+- transcription factor EB (TFEB) signaling axis induces autophagy and regulates clearance of M. bovis by bovine mammary epithelial cells (bMECs). M. bovis inhibited autophagy and lysosomal biogenesis in bMECs and suppressed HO-1 protein and expression of related proteins, namely nuclear factor erythroid 2-related factor 2 (Nrf2) and Kelch-like ECH-associated protein 1 (keap1). Activation of HO-1 and its production of carbon monoxide (CO) were required for induction of autophagy and clearance of intracellular M. bovis. Furthermore, when HO-1 was deficient, CO sustained cellular autophagy. HO-1 activation increased intracellular calcium (Ca2+) and cytosolic localization activity of TFEB via PERK. Knockdown of PERK or chelation of intracellular Ca2+ inhibited HO-1-induced M. bovis autophagy and clearance. M. bovis infection affected nuclear localization of lysosomal TFEB in the MiT/TFE transcription factor subfamily, whereas activation of HO-1 mediated dephosphorylation and intranuclear localization of TFEB, promoting autophagy, lysosomal biogenesis and autophagic clearance of M. bovis. Nuclear translocation of TFEB in HO-1 was critical to induce M. bovis transport and survival of infected bMECs. Furthermore, the HO-1/CO-PERK-Ca2+-TFEB signaling axis induced autophagy and M. bovis clearance, providing a viable approach to treat persistent M. bovis infections.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Calcium , Cell Nucleus , Endoplasmic Reticulum , Epithelial Cells , Mammary Glands, Animal , Mycoplasma bovis , Animals , Cattle , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Calcium/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Endoplasmic Reticulum/metabolism , Mammary Glands, Animal/microbiology , Mammary Glands, Animal/metabolism , Cell Nucleus/metabolism , Female , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Mycoplasma Infections/microbiology , Mycoplasma Infections/veterinary , Mycoplasma Infections/metabolism , Lysosomes/metabolism , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Carbon Monoxide/metabolism , Signal Transduction , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics
12.
Cell Rep ; 43(4): 114051, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38564334

ABSTRACT

Uropathogenic Escherichia coli (UPEC) is the most common causative agent of urinary tract infection (UTI). UPEC invades bladder epithelial cells (BECs) via fusiform vesicles, escapes into the cytosol, and establishes biofilm-like intracellular bacterial communities (IBCs). Nucleoside-diphosphate kinase (NDK) is secreted by pathogenic bacteria to enhance virulence. However, whether NDK is involved in UPEC pathogenesis remains unclear. Here, we find that the lack of ndk impairs the colonization of UPEC CFT073 in mouse bladders and kidneys owing to the impaired ability of UPEC to form IBCs. Furthermore, we demonstrate that NDK inhibits caspase-1-dependent pyroptosis by consuming extracellular ATP, preventing superficial BEC exfoliation, and promoting IBC formation. UPEC utilizes the reactive oxygen species (ROS) sensor OxyR to indirectly activate the regulator integration host factor, which then directly activates ndk expression in response to intracellular ROS. Here, we reveal a signaling transduction pathway that UPEC employs to inhibit superficial BEC exfoliation, thus facilitating acute UTI.


Subject(s)
Caspase 1 , Escherichia coli Infections , Nucleoside-Diphosphate Kinase , Pyroptosis , Urinary Tract Infections , Uropathogenic Escherichia coli , Uropathogenic Escherichia coli/pathogenicity , Animals , Urinary Tract Infections/microbiology , Urinary Tract Infections/pathology , Mice , Caspase 1/metabolism , Nucleoside-Diphosphate Kinase/metabolism , Nucleoside-Diphosphate Kinase/genetics , Escherichia coli Infections/microbiology , Escherichia coli Infections/metabolism , Escherichia coli Infections/pathology , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL , Humans , Female , Urinary Bladder/microbiology , Urinary Bladder/pathology , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Signal Transduction
13.
Microb Pathog ; 190: 106642, 2024 May.
Article in English | MEDLINE | ID: mdl-38599551

ABSTRACT

The intestinal and respiratory tracts of healthy individuals serve as habitats for a diverse array of microorganisms, among which Klebsiella oxytoca holds significance as a causative agent in numerous community- and hospital-acquired infections, often manifesting in polymicrobial contexts. In specific circumstances, K. oxytoca, alongside other constituents of the gut microbiota, undergoes translocation to distinct physiological niches. In these new environments, it engages in close interactions with other microbial community members. As this interaction may progress to co-infection where the virulence of involved pathogens may be promoted and enhance disease severity, we investigated how K. oxytoca affects the adhesion of commonly co-isolated bacteria and vice versa during co-incubation of different biotic and abiotic surfaces. Co-incubation was beneficial for the adhesion of at least one of the two co-cultured strains. K. oxytoca enhanced the adhesion of other enterobacteria strains to polystyrene and adhered more efficiently to bladder or lung epithelial cell lines in the presence of most enterobacteria strains and S. aureus. This effect was accompanied by bacterial coaggregation mediated by carbohydrate-protein interactions occurring between bacteria. These interactions occur only in sessile, but not planktonic populations, and depend on the features of the surface. The data are of particular importance for the risk assessment of the urinary and respiratory tract infections caused by K. oxytoca, including those device-associated. In this paper, we present the first report on K. oxytoca ability to acquire increased adhesive capacities on epithelial cells through interactions with common causal agents of urinary and respiratory tract infections.


Subject(s)
Bacterial Adhesion , Epithelial Cells , Klebsiella Infections , Klebsiella oxytoca , Lung , Urinary Bladder , Klebsiella oxytoca/physiology , Humans , Epithelial Cells/microbiology , Lung/microbiology , Klebsiella Infections/microbiology , Urinary Bladder/microbiology , Staphylococcus aureus/physiology , Staphylococcus aureus/pathogenicity , Coculture Techniques , Coinfection/microbiology , Cell Line , Microbial Interactions , Opportunistic Infections/microbiology , Respiratory Tract Infections/microbiology , Virulence
14.
PLoS Biol ; 22(4): e3002597, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38684033

ABSTRACT

Intestinal epithelial cells (IECs) play pivotal roles in nutrient uptake and in the protection against gut microorganisms. However, certain enteric pathogens, such as Salmonella enterica serovar Typhimurium (S. Tm), can invade IECs by employing flagella and type III secretion systems (T3SSs) with cognate effector proteins and exploit IECs as a replicative niche. Detection of flagella or T3SS proteins by IECs results in rapid host cell responses, i.e., the activation of inflammasomes. Here, we introduce a single-cell manipulation technology based on fluidic force microscopy (FluidFM) that enables direct bacteria delivery into the cytosol of single IECs within a murine enteroid monolayer. This approach allows to specifically study pathogen-host cell interactions in the cytosol uncoupled from preceding events such as docking, initiation of uptake, or vacuole escape. Consistent with current understanding, we show using a live-cell inflammasome reporter that exposure of the IEC cytosol to S. Tm induces NAIP/NLRC4 inflammasomes via its known ligands flagellin and T3SS rod and needle. Injected S. Tm mutants devoid of these invasion-relevant ligands were able to grow in the cytosol of IECs despite the absence of T3SS functions, suggesting that, in the absence of NAIP/NLRC4 inflammasome activation and the ensuing cell death, no effector-mediated host cell manipulation is required to render the epithelial cytosol growth-permissive for S. Tm. Overall, the experimental system to introduce S. Tm into single enteroid cells enables investigations into the molecular basis governing host-pathogen interactions in the cytosol with high spatiotemporal resolution.


Subject(s)
Calcium-Binding Proteins , Cytosol , Flagellin , Host-Pathogen Interactions , Inflammasomes , Salmonella typhimurium , Type III Secretion Systems , Cytosol/metabolism , Cytosol/microbiology , Animals , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/metabolism , Type III Secretion Systems/metabolism , Inflammasomes/metabolism , Mice , Flagellin/metabolism , Neuronal Apoptosis-Inhibitory Protein/metabolism , Neuronal Apoptosis-Inhibitory Protein/genetics , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Mice, Inbred C57BL , CARD Signaling Adaptor Proteins/metabolism , CARD Signaling Adaptor Proteins/genetics , Single-Cell Analysis/methods , Salmonella Infections/microbiology , Salmonella Infections/metabolism , Salmonella Infections/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism
15.
Front Immunol ; 15: 1374293, 2024.
Article in English | MEDLINE | ID: mdl-38680489

ABSTRACT

Introduction: Shigella is the etiologic agent of a bacillary dysentery known as shigellosis, which causes millions of infections and thousands of deaths worldwide each year due to Shigella's unique lifestyle within intestinal epithelial cells. Cell adhesion/invasion assays have been extensively used not only to identify targets mediating host-pathogen interaction, but also to evaluate the ability of Shigella-specific antibodies to reduce virulence. However, these assays are time-consuming and labor-intensive and fail to assess differences at the single-cell level. Objectives and methods: Here, we developed a simple, fast and high-content method named visual Adhesion/Invasion Inhibition Assay (vAIA) to measure the ability of anti-Shigellaantibodies to inhibit bacterial adhesion to and invasion of epithelial cells by using the confocal microscope Opera Phenix. Results: We showed that vAIA performed well with a pooled human serum from subjects challenged with S. sonnei and that a specific anti-IpaD monoclonal antibody effectively reduced bacterial virulence in a dose-dependent manner. Discussion: vAIA can therefore inform on the functionality of polyclonal and monoclonal responses thereby supporting the discovery of pathogenicity mechanisms and the development of candidate vaccines and immunotherapies. Lastly, this assay is very versatile and may be easily applied to other Shigella species or serotypes and to different pathogens.


Subject(s)
Antibodies, Bacterial , Bacterial Adhesion , Dysentery, Bacillary , Humans , Bacterial Adhesion/immunology , Dysentery, Bacillary/immunology , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/diagnosis , Antibodies, Bacterial/immunology , Host-Pathogen Interactions/immunology , Shigella/immunology , Shigella/pathogenicity , Epithelial Cells/microbiology , Epithelial Cells/immunology , Shigella sonnei/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , HeLa Cells
16.
Microbiol Spectr ; 12(5): e0425522, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38587411

ABSTRACT

tRNA modifications play important roles in maintaining translation accuracy in all domains of life. Disruptions in the tRNA modification machinery, especially of the anticodon stem loop, can be lethal for many bacteria and lead to a broad range of phenotypes in baker's yeast. Very little is known about the function of tRNA modifications in host-pathogen interactions, where rapidly changing environments and stresses require fast adaptations. We found that two closely related fungal pathogens of humans, the highly pathogenic Candida albicans and its much less pathogenic sister species, Candida dubliniensis, differ in the function of a tRNA-modifying enzyme. This enzyme, Hma1, exhibits species-specific effects on the ability of the two fungi to grow in the hypha morphology, which is central to their virulence potential. We show that Hma1 has tRNA-threonylcarbamoyladenosine dehydratase activity, and its deletion alters ribosome occupancy, especially at 37°C-the body temperature of the human host. A C. albicans HMA1 deletion mutant also shows defects in adhesion to and invasion into human epithelial cells and shows reduced virulence in a fungal infection model. This links tRNA modifications to host-induced filamentation and virulence of one of the most important fungal pathogens of humans.IMPORTANCEFungal infections are on the rise worldwide, and their global burden on human life and health is frequently underestimated. Among them, the human commensal and opportunistic pathogen, Candida albicans, is one of the major causative agents of severe infections. Its virulence is closely linked to its ability to change morphologies from yeasts to hyphae. Here, this ability is linked-to our knowledge for the first time-to modifications of tRNA and translational efficiency. One tRNA-modifying enzyme, Hma1, plays a specific role in C. albicans and its ability to invade the host. This adds a so-far unknown layer of regulation to the fungal virulence program and offers new potential therapeutic targets to fight fungal infections.


Subject(s)
Candida albicans , Candidiasis , Fungal Proteins , Hyphae , RNA, Transfer , Candida albicans/genetics , Candida albicans/pathogenicity , Candida albicans/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Virulence/genetics , Humans , Fungal Proteins/genetics , Fungal Proteins/metabolism , Candidiasis/microbiology , Hyphae/growth & development , Hyphae/genetics , Hyphae/metabolism , Animals , Candida/pathogenicity , Candida/genetics , Candida/metabolism , Host-Pathogen Interactions , Mice , Epithelial Cells/microbiology
17.
Infect Immun ; 92(5): e0044723, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38629841

ABSTRACT

Streptococcus pneumoniae, a common colonizer of the upper respiratory tract, invades nasopharyngeal epithelial cells without causing disease in healthy participants of controlled human infection studies. We hypothesized that surface expression of pneumococcal lipoproteins, recognized by the innate immune receptor TLR2, mediates epithelial microinvasion. Mutation of lgt in serotype 4 (TIGR4) and serotype 6B (BHN418) pneumococcal strains abolishes the ability of the mutants to activate TLR2 signaling. Loss of lgt also led to the concomitant decrease in interferon signaling triggered by the bacterium. However, only BHN418 lgt::cm but not TIGR4 lgt::cm was significantly attenuated in epithelial adherence and microinvasion compared to their respective wild-type strains. To test the hypothesis that differential lipoprotein repertoires in TIGR4 and BHN418 lead to the intraspecies variation in epithelial microinvasion, we employed a motif-based genome analysis and identified an additional 525 a.a. lipoprotein (pneumococcal accessory lipoprotein A; palA) encoded by BHN418 that is absent in TIGR4. The gene encoding palA sits within a putative genetic island present in ~10% of global pneumococcal isolates. While palA was enriched in the carriage and otitis media pneumococcal strains, neither mutation nor overexpression of the gene encoding this lipoprotein significantly changed microinvasion patterns. In conclusion, mutation of lgt attenuates epithelial inflammatory responses during pneumococcal-epithelial interactions, with intraspecies variation in the effect on microinvasion. Differential lipoprotein repertoires encoded by the different strains do not explain these differences in microinvasion. Rather, we postulate that post-translational modifications of lipoproteins may account for the differences in microinvasion.IMPORTANCEStreptococcus pneumoniae (pneumococcus) is an important mucosal pathogen, estimated to cause over 500,000 deaths annually. Nasopharyngeal colonization is considered a necessary prerequisite for disease, yet many people are transiently and asymptomatically colonized by pneumococci without becoming unwell. It is therefore important to better understand how the colonization process is controlled at the epithelial surface. Controlled human infection studies revealed the presence of pneumococci within the epithelium of healthy volunteers (microinvasion). In this study, we focused on the regulation of epithelial microinvasion by pneumococcal lipoproteins. We found that pneumococcal lipoproteins induce epithelial inflammation but that differing lipoprotein repertoires do not significantly impact the magnitude of microinvasion. Targeting mucosal innate immunity and epithelial microinvasion alongside the induction of an adaptive immune response may be effective in preventing pneumococcal colonization and disease.


Subject(s)
Epithelial Cells , Lipoproteins , Pneumococcal Infections , Streptococcus pneumoniae , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/pathogenicity , Humans , Lipoproteins/genetics , Lipoproteins/metabolism , Lipoproteins/immunology , Epithelial Cells/microbiology , Epithelial Cells/immunology , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Nasopharynx/microbiology , Mutation , Bacterial Adhesion
18.
Int Immunopharmacol ; 132: 111780, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38603853

ABSTRACT

BACKGROUND: Glycopeptide antibiotic vancomycin is a bactericidal antibiotic available for the infection to Staphylococcus aureus (SA), however, SA has a strong adaptive capacity and thereby acquires resistance to vancomycin. This study aims to illuminate the possible molecular mechanism of vancomycin resistance of SA based on the 16S rRNA sequencing data and microarray profiling data. METHODS: 16S rRNA sequencing data of control samples and urinary tract infection samples were retrieved from the EMBL-EBI (European Molecular Biology Laboratory - European Bioinformatics Institute) database. Correlation of gut flora and clinical indicators was evaluated. The possible targets regulated by SA were predicted by microarray profiling and subjected to KEGG (Kyoto Encyclopedia of Genes and Genomes) enrichment analysis. CXCL10 gene knockout and overexpression were introduced to evaluate the effect of CXCL10 on the virulence of SA and the resistance to vancomycin. SA strains were co-cultured with urethral epithelial cells in vitro. The presence of SA virulence factors was detected using PCR. Biofilm formation of SA strains was assessed using the microtiter plate method. Furthermore, the antibiotic sensitivity of SA strains was evaluated through vancomycin testing. RESULTS: Gut flora and its species abundance had significant difference between urinary tract infection and control samples. SA was significantly differentially expressed in urinary tract infection samples. Resistance of SA to vancomycin mainly linked to the D-alanine metabolism pathway. SA may participate in the occurrence of urinary tract infection by upregulating CXCL10. In addition, CXCL10 mainly affected the SA resistance to vancomycin through the TLR signaling pathway. In vitro experimental results further confirmed that the overexpression of CXCL10 in SA increased SA virulence and decreased its susceptibility to vancomycin. In vitro experimental validation demonstrated that the knockout of CXCL10 in urethral epithelial cells enhanced the sensitivity of Staphylococcus aureus (SA) to vancomycin. CONCLUSION: SA upregulates the expression of CXCL10 in urethral epithelial cells, thereby activating the TLR signaling pathway and promoting resistance to glycopeptide antibiotics in SA.


Subject(s)
Anti-Bacterial Agents , Chemokine CXCL10 , Staphylococcal Infections , Staphylococcus aureus , Urinary Tract Infections , Vancomycin Resistance , Vancomycin , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Vancomycin/pharmacology , Humans , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Chemokine CXCL10/metabolism , Chemokine CXCL10/genetics , Vancomycin Resistance/genetics , Urinary Tract Infections/microbiology , Urinary Tract Infections/drug therapy , Biofilms/drug effects , Gastrointestinal Microbiome/drug effects , RNA, Ribosomal, 16S/genetics , Epithelial Cells/microbiology , Epithelial Cells/drug effects , Female , Male
19.
Vet Microbiol ; 293: 110091, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38626624

ABSTRACT

Mastitis in dairy cows is mainly caused by bacteria, in which Staphylococcus aureus appears frequently. Epithelial cells, as a major physical barrier of mammary gland, play an important role in preventing mastitis in dairy cows. Our previous study reported that Rab11fip4 (an effector of Rab11) was significantly changed in response to stimulation by S. aureus. So, in this study, the role of Rab11A in phagocytosis of bovine mammary epithelial cells (MAC-T) against S. aureus was evaluated. First, changes of Rab11A and Rab11fip4 were analyzed in response to S. aureus by immunofluorescence and western blotting. Subsequently, the effects of Rab11A and Rab11fip4 on proliferation of S. aureus, as well as formation and function of late endosomes (LEs) and lysosomes (LYSs) were investigated. The results showed that, after infection, Rab11A and Rab11fip4 were recruited to phagosomes containing S. aureus. Rab11A promoted bacterial clearance and rescues the destruction of LEs and LYSs by S. aureus, whereas Rab11fip4 did the opposite. These findings provide new insights into phagocytosis and control of S. aureus in host cells, thus lay the foundation to elucidate the pathogenesis of S. aureus in bovine mastitis.


Subject(s)
Epithelial Cells , Mastitis, Bovine , Phagocytosis , Staphylococcal Infections , Staphylococcus aureus , rab GTP-Binding Proteins , Animals , Cattle , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Staphylococcus aureus/physiology , Female , Epithelial Cells/microbiology , Staphylococcal Infections/veterinary , Staphylococcal Infections/microbiology , Mastitis, Bovine/microbiology , Mammary Glands, Animal/microbiology , Endosomes/metabolism , Endosomes/microbiology , Lysosomes/metabolism , Lysosomes/microbiology , Cell Line , Phagosomes/microbiology
20.
Nature ; 629(8012): 669-678, 2024 May.
Article in English | MEDLINE | ID: mdl-38600382

ABSTRACT

Interleukin 22 (IL-22) has a non-redundant role in immune defence of the intestinal barrier1-3. T cells, but not innate lymphoid cells, have an indispensable role in sustaining the IL-22 signalling that is required for the protection of colonic crypts against invasion during infection by the enteropathogen Citrobacter rodentium4 (Cr). However, the intestinal epithelial cell (IEC) subsets targeted by T cell-derived IL-22, and how T cell-derived IL-22 sustains activation in IECs, remain undefined. Here we identify a subset of absorptive IECs in the mid-distal colon that are specifically targeted by Cr and are differentially responsive to IL-22 signalling. Major histocompatibility complex class II (MHCII) expression by these colonocytes was required to elicit sustained IL-22 signalling from Cr-specific T cells, which was required to restrain Cr invasion. Our findings explain the basis for the regionalization of the host response to Cr and demonstrate that epithelial cells must elicit MHCII-dependent help from IL-22-producing T cells to orchestrate immune protection in the intestine.


Subject(s)
Citrobacter rodentium , Colon , Epithelial Cells , Intestinal Mucosa , T-Lymphocytes , Animals , Female , Male , Mice , Citrobacter rodentium/immunology , Colon/cytology , Colon/immunology , Colon/microbiology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Interleukin-22/immunology , Interleukin-22/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/cytology , Mice, Inbred C3H , Mice, Inbred C57BL , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...