Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
1.
Elife ; 132024 May 20.
Article in English | MEDLINE | ID: mdl-38767331

ABSTRACT

Wound infections are highly prevalent and can lead to delayed or failed healing, causing significant morbidity and adverse economic impacts. These infections occur in various contexts, including diabetic foot ulcers, burns, and surgical sites. Enterococcus faecalis is often found in persistent non-healing wounds, but its contribution to chronic wounds remains understudied. To address this, we employed single-cell RNA sequencing (scRNA-seq) on infected wounds in comparison to uninfected wounds in a mouse model. Examining over 23,000 cells, we created a comprehensive single-cell atlas that captures the cellular and transcriptomic landscape of these wounds. Our analysis revealed unique transcriptional and metabolic alterations in infected wounds, elucidating the distinct molecular changes associated with bacterial infection compared to the normal wound healing process. We identified dysregulated keratinocyte and fibroblast transcriptomes in response to infection, jointly contributing to an anti-inflammatory environment. Notably, E. faecalis infection prompted a premature, incomplete epithelial-mesenchymal transition in keratinocytes. Additionally, E. faecalis infection modulated M2-like macrophage polarization by inhibiting pro-inflammatory resolution in vitro, in vivo, and in our scRNA-seq atlas. Furthermore, we discovered macrophage crosstalk with neutrophils, which regulates chemokine signaling pathways, while promoting anti-inflammatory interactions with endothelial cells. Overall, our findings offer new insights into the immunosuppressive role of E. faecalis in wound infections.


If wounds get infected, they heal much more slowly, sometimes leading to skin damage and other complications, including disseminated infections or even amputation. Infections can happen in many types of wounds, ranging from ulcers in patients with diabetes to severe burns. If infections are not cleared quickly, the wounds can become 'chronic' and are unable to heal without intervention. Enterococcus faecalis is a type of bacteria that normally lives in the gut. Within that environment, in healthy people, it is not harmful. However, if it comes into contact with wounds ­ particularly diabetic ulcers or the site of a surgery ­ it can cause persistent infections and prevent healing. Although researchers are beginning to understand how E. faecalis initially colonises wounds, the biological mechanisms that transform these infections into chronic wounds are still largely unknown. Celik et al. therefore set out to investigate exactly how E. faecalis interferes with wound healing. To do this, Celik et al. looked at E. faecalis-infected wounds in mice and compared them to uninfected ones. Using a genetic technique called single-cell RNA sequencing, Celik et al. were able to determine which genes were switched on in individual skin and immune cells at the site of the wounds. This in turn allowed the researchers to determine how those cells were behaving in both infected and uninfected conditions. The experiments revealed that when E. faecalis was present in wounds, several important cell types in the wounds did not behave normally. For example, although the infected skin cells still underwent a change in behaviour required for healing (called an epithelial-mesenchymal transition), the change was both premature and incomplete. In other words, the skin cells in infected wounds started changing too early and did not finish the healing process properly. E. faecalis also changed the way macrophages and neutrophils worked within the wounds. These are cells in our immune system that normally promote inflammation, a process involved in both uninfected wounds or during infections and is a key part of wound healing when properly controlled. In the E. faecalis-infected wounds, these cells' inflammatory properties were suppressed, making them less helpful for healing. These results shed new light on how E. faecalis interacts with skin cells and the immune system to disrupt wound healing. Celik et al. hope that this knowledge will allow us to find new ways to target E. faecalis infections, and ultimately develop treatments to help chronic wounds heal better and faster.


Subject(s)
Enterococcus faecalis , Gram-Positive Bacterial Infections , Keratinocytes , Wound Healing , Enterococcus faecalis/physiology , Enterococcus faecalis/genetics , Animals , Mice , Gram-Positive Bacterial Infections/microbiology , Keratinocytes/microbiology , Keratinocytes/metabolism , Macrophages/microbiology , Macrophages/metabolism , Macrophages/immunology , Disease Models, Animal , Wound Infection/microbiology , Transcriptome , Mice, Inbred C57BL , Single-Cell Analysis , Epithelial-Mesenchymal Transition/genetics , Male , Fibroblasts/microbiology , Fibroblasts/metabolism
2.
ACS Infect Dis ; 10(5): 1725-1738, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38602352

ABSTRACT

Host-acting compounds are emerging as potential alternatives to combating antibiotic resistance. Here, we show that bosutinib, an FDA-approved chemotherapeutic for treating chronic myelogenous leukemia, does not possess any antibiotic activity but enhances macrophage responses to bacterial infection. In vitro, bosutinib stimulates murine and human macrophages to kill bacteria more effectively. In a murine wound infection with vancomycin-resistant Enterococcus faecalis, a single intraperitoneal bosutinib injection or multiple topical applications on the wound reduce the bacterial load by approximately 10-fold, which is abolished by macrophage depletion. Mechanistically, bosutinib stimulates macrophage phagocytosis of bacteria by upregulating surface expression of bacterial uptake markers Dectin-1 and CD14 and promoting actin remodeling. Bosutinib also stimulates bacterial killing by elevating the intracellular levels of reactive oxygen species. Moreover, bosutinib drives NF-κB activation, which protects infected macrophages from dying. Other Src kinase inhibitors such as DMAT and tirbanibulin also upregulate expression of bacterial uptake markers in macrophages and enhance intracellular bacterial killing. Finally, cotreatment with bosutinib and mitoxantrone, another chemotherapeutic in clinical use, results in an additive effect on bacterial clearance in vitro and in vivo. These results show that bosutinib stimulates macrophage clearance of bacterial infections through multiple mechanisms and could be used to boost the host innate immunity to combat drug-resistant bacterial infections.


Subject(s)
Aniline Compounds , Anti-Bacterial Agents , Cell Survival , Macrophages , Phagocytosis , Animals , Humans , Mice , Aniline Compounds/pharmacology , Anti-Bacterial Agents/pharmacology , Cell Survival/drug effects , Enterococcus faecalis/drug effects , Gram-Positive Bacterial Infections/drug therapy , Macrophages/drug effects , Mice, Inbred C57BL , NF-kappa B/metabolism , Nitriles/pharmacology , Phagocytosis/drug effects , Quinolines/pharmacology , Reactive Oxygen Species/metabolism
3.
Mol Microbiol ; 121(5): 1021-1038, 2024 05.
Article in English | MEDLINE | ID: mdl-38527904

ABSTRACT

Daptomycin is a last-line antibiotic commonly used to treat vancomycin-resistant Enterococci, but resistance evolves rapidly and further restricts already limited treatment options. While genetic determinants associated with clinical daptomycin resistance (DAPR) have been described, information on factors affecting the speed of DAPR acquisition is limited. The multiple peptide resistance factor (MprF), a phosphatidylglycerol-modifying enzyme involved in cationic antimicrobial resistance, is linked to DAPR in pathogens such as methicillin-resistant Staphylococcus aureus. Since Enterococcus faecalis encodes two paralogs of mprF and clinical DAPR mutations do not map to mprF, we hypothesized that functional redundancy between the paralogs prevents mprF-mediated resistance and masks other evolutionary pathways to DAPR. Here, we performed in vitro evolution to DAPR in mprF mutant background. We discovered that the absence of mprF results in slowed DAPR evolution and is associated with inactivating mutations in ftsH, resulting in the depletion of the chaperone repressor HrcA. We also report that ftsH is essential in the parental, but not in the ΔmprF, strain where FtsH depletion results in growth impairment in the parental strain, a phenotype associated with reduced extracellular acidification and reduced ability for metabolic reduction. This presents FtsH and HrcA as enticing targets for developing anti-resistance strategies.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Daptomycin , Enterococcus faecalis , Microbial Sensitivity Tests , Enterococcus faecalis/genetics , Enterococcus faecalis/drug effects , Enterococcus faecalis/metabolism , Enterococcus faecalis/enzymology , Daptomycin/pharmacology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Anti-Bacterial Agents/pharmacology , Mutation , Drug Resistance, Bacterial/genetics , Peptide Hydrolases/metabolism , Peptide Hydrolases/genetics , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/metabolism
4.
mBio ; 15(1): e0238423, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38078746

ABSTRACT

IMPORTANCE: Although E. faecalis is a common wound pathogen, its pathogenic mechanisms during wound infection are unexplored. Here, combining a mouse wound infection model with in vivo transposon and RNA sequencing approaches, we identified the E. faecalis purine biosynthetic pathway and galactose/mannose MptABCD phosphotransferase system as essential for E. faecalis acute replication and persistence during wound infection, respectively. The essentiality of purine biosynthesis and the MptABCD PTS is driven by the consumption of purine metabolites by E. faecalis during acute replication and changing carbohydrate availability during the course of wound infection. Overall, our findings reveal the importance of the wound microenvironment in E. faecalis wound pathogenesis and how these metabolic pathways can be targeted to better control wound infections.


Subject(s)
Urinary Tract Infections , Wound Infection , Animals , Mice , Enterococcus faecalis/genetics , Enterococcus faecalis/metabolism , Carbohydrates , Purines
5.
Article in English | MEDLINE | ID: mdl-38010482

ABSTRACT

Black, Hispanic, and Asian individuals, the three largest US racial/ethnic minorities, continue to suffer disproportionately from breast, cervical, and colon cancers largely because cancer screening continues to be underutilized even after decades of availability. This study examined the utility of theoretically grounded and culturally adapted in-person theater monologues aimed at promoting early detection screening among the three highest population racial/ethnic groups in Harris County, Houston, TX. Nine monologues were created to promote cancer screening and early detection for breast, cervical, and colorectal cancers in three different languages (English, Spanish, Vietnamese) and targeting underserved Black, Hispanic, and Vietnamese adult Harris County residents. From January 2014 to March 2020, 265 live monologue outreach events were held with 110 focused on prevention and screening for breast cancer, 75 for colorectal cancer, and 80 for cervical cancer. A total of 5989 individuals attended these outreach events and 86.3% completed the post-performance evaluation survey. Overall for all monologues, 6.6% of participants reported a positive change in their intent to screen from 75.7 to 82.3% after intervention (p < 0.001) and audience member scores on knowledge questions for all three cancers were mostly positive. Importantly, early detection questions for all three cancers were over 90% correct for all respondents, and well over 70% for the various groups. The findings revealed opportunities for improving monologue content to cultivate cancer early detection and screening knowledge. Results suggest that a theater-based approach may be an effective strategy to disseminate cancer screening education, improve knowledge, and increase intent to obtain screening among medically underserved communities.

6.
FEMS Microbes ; 4: xtad019, 2023.
Article in English | MEDLINE | ID: mdl-37900578

ABSTRACT

Enterococcus faecalis is an opportunistic pathogen that is frequently co-isolated with other microbes in wound infections. While E. faecalis can subvert the host immune response and promote the survival of other microbes via interbacterial synergy, little is known about the impact of E. faecalis-mediated immune suppression on co-infecting microbes. We hypothesized that E. faecalis can attenuate neutrophil-mediated responses in mixed-species infection to promote survival of the co-infecting species. We found that neutrophils control E. faecalis infection via phagocytosis, ROS production, and degranulation of azurophilic granules, but it does not trigger neutrophil extracellular trap formation (NETosis). However, E. faecalis attenuates Staphylococcus aureus-induced NETosis in polymicrobial infection by interfering with citrullination of histone, suggesting E. faecalis can actively suppress NETosis in neutrophils. Residual S. aureus-induced NETs that remain during co-infection do not impact E. faecalis, further suggesting that E. faecalis possess mechanisms to evade or survive NET-associated killing mechanisms. E. faecalis-driven reduction of NETosis corresponds with higher S. aureus survival, indicating that this immunomodulating effect could be a risk factor in promoting the virulence polymicrobial infection. These findings highlight the complexity of the immune response to polymicrobial infections and suggest that attenuated pathogen-specific immune responses contribute to pathogenesis in the mammalian host.

7.
Nat Commun ; 14(1): 4093, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37433804

ABSTRACT

Bacteria possess elaborate systems to manage reactive oxygen and nitrogen species (ROS) arising from exposure to the mammalian immune system and environmental stresses. Here we report the discovery of an ROS-sensing RNA-modifying enzyme that regulates translation of stress-response proteins in the gut commensal and opportunistic pathogen Enterococcus faecalis. We analyze the tRNA epitranscriptome of E. faecalis in response to reactive oxygen species (ROS) or sublethal doses of ROS-inducing antibiotics and identify large decreases in N2-methyladenosine (m2A) in both 23 S ribosomal RNA and transfer RNA. This we determine to be due to ROS-mediated inactivation of the Fe-S cluster-containing methyltransferase, RlmN. Genetic knockout of RlmN gives rise to a proteome that mimics the oxidative stress response, with an increase in levels of superoxide dismutase and decrease in virulence proteins. While tRNA modifications were established to be dynamic for fine-tuning translation, here we report the discovery of a dynamically regulated, environmentally responsive rRNA modification. These studies lead to a model in which RlmN serves as a redox-sensitive molecular switch, directly relaying oxidative stress to modulating translation through the rRNA and the tRNA epitranscriptome, adding a different paradigm in which RNA modifications can directly regulate the proteome.


Subject(s)
Enterococcus faecalis , Proteome , Animals , Reactive Oxygen Species , Enterococcus faecalis/genetics , Proteome/genetics , Oxidative Stress/genetics , RNA Processing, Post-Transcriptional , Adenosine , Heat-Shock Proteins , Mammals
8.
Vaccines (Basel) ; 11(4)2023 Mar 25.
Article in English | MEDLINE | ID: mdl-37112640

ABSTRACT

Adolescents living in rural areas are less likely to be up to date on the human papillomavirus (HPV) vaccine, which can prevent cervical cancer. We administered a telephone survey to 27 clinics in rural East Texas to assess perceived barriers to HPV vaccination and current use of evidence-based interventions to promote HPV vaccination. Perceived barriers were assessed using a 5-point Likert scale and clinical implementation of evidence-based practices was determined. Findings are reported using descriptive statistics. The most commonly reported barriers were missed vaccination opportunities due to the pandemic (66.7%), followed by vaccine hesitancy due to the pandemic (44.4%) and due to the HPV vaccine specifically (33.3%). Fewer than a third of clinics reported using the evidence-based strategies of use of a "refusal to vaccinate" form (29.6%), having an identified HPV vaccine champion (29.6%), and recommending the HPV vaccine at age 9 (22.2%). While many clinics surveyed currently implement evidence-based practices to promote HPV vaccination, there is a need and desire for additional HPV vaccination interventions in East Texas clinics.

9.
Sci Adv ; 9(8): eadd9280, 2023 02 22.
Article in English | MEDLINE | ID: mdl-36812322

ABSTRACT

Antibiotic resistance critically limits treatment options for infection caused by opportunistic pathogens such as enterococci. Here, we investigate the antibiotic and immunological activity of the anticancer agent mitoxantrone (MTX) in vitro and in vivo against vancomycin-resistant Enterococcus faecalis (VRE). We show that, in vitro, MTX is a potent antibiotic against Gram-positive bacteria through induction of reactive oxygen species and DNA damage. MTX also synergizes with vancomycin against VRE, rendering the resistant strains more permeable to MTX. In a murine wound infection model, single-dose MTX treatment effectively reduces VRE numbers, with further reduction when combined with vancomycin. Multiple MTX treatments accelerate wound closure. MTX also promotes macrophage recruitment and proinflammatory cytokine induction at the wound site and augments intracellular bacterial killing in macrophages by up-regulating the expression of lysosomal enzymes. These results show that MTX represents a promising bacterium- and host-targeted therapeutic for overcoming vancomycin resistance.


Subject(s)
Enterococcus faecalis , Vancomycin-Resistant Enterococci , Animals , Mice , Enterococcus faecalis/genetics , Vancomycin Resistance/genetics , Vancomycin/pharmacology , Mitoxantrone/pharmacology , Anti-Bacterial Agents/pharmacology , Vancomycin-Resistant Enterococci/genetics
10.
mBio ; 14(1): e0307322, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36629455

ABSTRACT

The bacterial cell membrane is an interface for cell envelope synthesis, protein secretion, virulence factor assembly, and a target for host cationic antimicrobial peptides (CAMPs). To resist CAMP killing, several Gram-positive pathogens encode the multiple peptide resistance factor (MprF) enzyme that covalently attaches cationic amino acids to anionic phospholipids in the cell membrane. While E. faecalis encodes two mprF paralogs, MprF2 plays a dominant role in conferring resistance to killing by the CAMP human ß-defensin 2 (hBD-2) in E. faecalis strain OG1RF. The goal of the current study is to understand the broader lipidomic and functional roles of E. faecalis mprF. We analyzed the lipid profiles of parental wild-type and mprF mutant strains and show that while ΔmprF2 and ΔmprF1 ΔmprF2 mutants completely lacked cationic lysyl-phosphatidylglycerol (L-PG), the ΔmprF1 mutant synthesized ~70% of L-PG compared to the parent. Unexpectedly, we also observed a significant reduction of PG in ΔmprF2 and ΔmprF1 ΔmprF2. In the mprF mutants, particularly ΔmprF1 ΔmprF2, the decrease in L-PG and phosphatidylglycerol (PG) is compensated by an increase in a phosphorus-containing lipid, glycerophospho-diglucosyl-diacylglycerol (GPDGDAG), and D-ala-GPDGDAG. These changes were accompanied by a downregulation of de novo fatty acid biosynthesis and an accumulation of long-chain acyl-acyl carrier proteins (long-chain acyl-ACPs), suggesting that the suppression of fatty acid biosynthesis was mediated by the transcriptional repressor FabT. Growth in chemically defined media lacking fatty acids revealed severe growth defects in the ΔmprF1 ΔmprF2 mutant strain, but not the single mutants, which was partially rescued through supplementation with palmitic and stearic acids. Changes in lipid homeostasis correlated with lower membrane fluidity, impaired protein secretion, and increased biofilm formation in both ΔmprF2 and ΔmprF1 ΔmprF2, compared to the wild type and ΔmprF1. Collectively, our findings reveal a previously unappreciated role for mprF in global lipid regulation and cellular physiology, which could facilitate the development of novel therapeutics targeting MprF. IMPORTANCE The cell membrane plays a pivotal role in protecting bacteria against external threats, such as antibiotics. Cationic phospholipids such as lysyl-phosphatidyglycerol (L-PG) resist the action of cationic antimicrobial peptides through electrostatic repulsion. Here we demonstrate that L-PG depletion has several unexpected consequences in Enterococcus faecalis, including a reduction of phosphatidylglycerol (PG), enrichment of a phosphorus-containing lipid, reduced fatty acid synthesis accompanied by an accumulation of long-chain acyl-acyl carrier proteins (long chain acyl-ACPs), lower membrane fluidity, and impaired secretion. These changes are not deleterious to the organism as long as exogenous fatty acids are available for uptake from the culture medium. Our findings suggest an adaptive mechanism involving compensatory changes across the entire lipidome upon removal of a single phospholipid modification. Such adaptations must be considered when devising antimicrobial strategies that target membrane lipids.


Subject(s)
Anti-Bacterial Agents , Anti-Infective Agents , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Enterococcus faecalis/metabolism , Drug Resistance, Bacterial , Phospholipids/metabolism , Anti-Infective Agents/metabolism , Fatty Acids/metabolism , Phosphatidylglycerols/metabolism , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/metabolism , Cations/metabolism , Carrier Proteins/metabolism , Bacterial Proteins/metabolism
11.
Mol Microbiol ; 119(1): 1-18, 2023 01.
Article in English | MEDLINE | ID: mdl-36420961

ABSTRACT

Enterococcus faecalis virulence requires cell wall-associated proteins, including the sortase-assembled endocarditis and biofilm associated pilus (Ebp), important for biofilm formation in vitro and in vivo. The current paradigm for sortase-assembled pilus biogenesis in Gram-positive bacteria is that sortases attach substrates to lipid II peptidoglycan (PG) precursors, prior to their incorporation into the growing cell wall. Contrary to prevailing dogma, by following the distribution of Ebp and PG throughout the E. faecalis cell cycle, we found that cell surface Ebp do not co-localize with newly synthesized PG. Instead, surface-exposed Ebp are localized to the older cell hemisphere and excluded from sites of new PG synthesis at the septum. Moreover, Ebp deposition on the younger hemisphere of the E. faecalis diplococcus appear as foci adjacent to the nascent septum. We propose a new model whereby sortase substrate deposition can occur on older PG rather than at sites of new cell wall synthesis. Consistent with this model, we demonstrate that sequestering lipid II to block PG synthesis via ramoplanin, does not impact new Ebp deposition at the cell surface. These data support an alternative paradigm for sortase substrate deposition in E. faecalis, in which Ebp are anchored directly onto uncrosslinked cell wall, independent of new PG synthesis.


Subject(s)
Aminoacyltransferases , Fimbriae Proteins , Fimbriae Proteins/metabolism , Enterococcus faecalis/metabolism , Bacterial Proteins/metabolism , Fimbriae, Bacterial/metabolism , Cell Wall/metabolism , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism
12.
PLoS Pathog ; 18(9): e1010766, 2022 09.
Article in English | MEDLINE | ID: mdl-36067266

ABSTRACT

Wound infections are often polymicrobial in nature, biofilm associated and therefore tolerant to antibiotic therapy, and associated with delayed healing. Escherichia coli and Staphylococcus aureus are among the most frequently cultured pathogens from wound infections. However, little is known about the frequency or consequence of E. coli and S. aureus polymicrobial interactions during wound infections. Here we show that E. coli kills Staphylococci, including S. aureus, both in vitro and in a mouse excisional wound model via the genotoxin, colibactin. Colibactin biosynthesis is encoded by the pks locus, which we identified in nearly 30% of human E. coli wound infection isolates. While it is not clear how colibactin is released from E. coli or how it penetrates target cells, we found that the colibactin intermediate N-myristoyl-D-Asn (NMDA) disrupts the S. aureus membrane. We also show that the BarA-UvrY two component system (TCS) senses the environment created during E. coli and S. aureus mixed species interaction, leading to upregulation of pks island genes. Further, we show that BarA-UvrY acts via the carbon storage global regulatory (Csr) system to control pks expression. Together, our data demonstrate the role of colibactin in interspecies competition and show that it is regulated by BarA-UvrY TCS during interspecies competition.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Membrane Proteins , Phosphotransferases , Polyketides , Staphylococcus aureus , Transcription Factors , Animals , Anti-Bacterial Agents/metabolism , Carbon/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mutagens/metabolism , N-Methylaspartate/metabolism , Peptides , Phosphotransferases/genetics , Polyketides/metabolism , Staphylococcus/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Transcription Factors/metabolism , Wound Infection/microbiology
13.
J Bacteriol ; 204(7): e0061521, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35758750

ABSTRACT

Enterococcus faecalis is often coisolated with Pseudomonas aeruginosa in polymicrobial biofilm-associated infections of wounds and the urinary tract. As a defense strategy, the host innately restricts iron availability at infection sites. Despite their coprevalence, the polymicrobial interactions of these two species in biofilms and under iron-restricted conditions remain unexplored. Here, we show that E. faecalis inhibits P. aeruginosa growth within biofilms when iron is restricted. E. faecalis lactate dehydrogenase (ldh1) gives rise to l-lactate production during fermentative growth. We find that an E. faecalis ldh1 mutant fails to inhibit P. aeruginosa growth. Additionally, we demonstrate that ldh1 expression is induced under iron-restricted conditions, resulting in increased lactic acid exported and, consequently, a reduction in local environmental pH. Together, our results suggest that E. faecalis synergistically inhibits P. aeruginosa growth by decreasing environmental pH and l-lactate-mediated iron chelation. Overall, this study emphasizes the importance of the microenvironment in polymicrobial interactions and how manipulating the microenvironment can impact the growth trajectory of bacterial communities. IMPORTANCE Many infections are polymicrobial and biofilm-associated in nature. Iron is essential for many metabolic processes and plays an important role in controlling infections, where the host restricts iron as a defense mechanism against invading pathogens. However, polymicrobial interactions between pathogens are underexplored under iron-restricted conditions. Here, we explore the polymicrobial interactions between commonly coisolated E. faecalis and P. aeruginosa within biofilms. We find that E. faecalis modulates the microenvironment by exporting lactic acid which further chelates already limited iron and also lowers the environmental pH to antagonize P. aeruginosa growth under iron-restricted conditions. Our findings provide insights into polymicrobial interactions between bacteria and how manipulating the microenvironment can be taken advantage of to better control infections.


Subject(s)
Enterococcus faecalis , Pseudomonas aeruginosa , Biofilms , Enterococcus faecalis/metabolism , Iron/metabolism , Lactic Acid/metabolism , Pseudomonas aeruginosa/metabolism
14.
ISME J ; 16(8): 2015-2026, 2022 08.
Article in English | MEDLINE | ID: mdl-35589966

ABSTRACT

The contribution of biofilms to virulence and as a barrier to treatment is well-established for Staphylococcus aureus and Enterococcus faecalis, both nosocomial pathogens frequently isolated from biofilm-associated infections. Despite frequent co-isolation, their interactions in biofilms have not been well-characterized. We report that in combination, these two species can give rise to augmented biofilms biomass that is dependent on the activation of E. faecalis aerobic respiration. In E. faecalis, respiration requires both exogenous heme to activate the cydAB-encoded heme-dependent cytochrome bd, and the availability of O2. We determined that the ABC transporter encoded by cydDC contributes to heme import. In dual species biofilms, S. aureus provides the heme to activate E. faecalis respiration. S. aureus mutants deficient in heme biosynthesis were unable to augment biofilms whereas heme alone is sufficient to augment E. faecalis mono-species biofilms. Our results demonstrate that S. aureus-derived heme, likely in the form of released hemoproteins, promotes E. faecalis biofilm formation, and that E. faecalis gelatinase activity facilitates heme extraction from hemoproteins. This interspecies interaction and metabolic cross-feeding may explain the frequent co-occurrence of these microbes in biofilm-associated infections.


Subject(s)
Enterococcus faecalis , Staphylococcus aureus , Biofilms , Enterococcus faecalis/genetics , Heme , Staphylococcus aureus/genetics , Virulence
15.
PLoS Pathog ; 18(4): e1010434, 2022 04.
Article in English | MEDLINE | ID: mdl-35390107

ABSTRACT

Enterococcus faecalis is a frequent opportunistic pathogen of wounds, whose infections are associated with biofilm formation, persistence, and recalcitrance toward treatment. We have previously shown that E. faecalis wound infection persists for at least 7 days. Here we report that viable E. faecalis are present within both immune and non-immune cells at the wound site up to 5 days after infection, raising the prospect that intracellular persistence contributes to chronic E. faecalis infection. Using in vitro keratinocyte and macrophage infection models, we show that E. faecalis becomes internalized and a subpopulation of bacteria can survive and replicate intracellularly. E. faecalis are internalized into keratinocytes primarily via macropinocytosis into single membrane-bound compartments and can persist in late endosomes up to 24 h after infection in the absence of colocalization with the lysosomal protease Cathepsin D or apparent fusion with the lysosome, suggesting that E. faecalis blocks endosomal maturation. Indeed, intracellular E. faecalis infection results in heterotypic intracellular trafficking with partial or absent labelling of E. faecalis-containing compartments with Rab5 and Rab7, small GTPases required for the endosome-lysosome trafficking. In addition, E. faecalis infection results in marked reduction of Rab5 and Rab7 protein levels which may also contribute to attenuated Rab incorporation into E. faecalis-containing compartments. Finally, we demonstrate that intracellular E. faecalis derived from infected keratinocytes are significantly more efficient in reinfecting new keratinocytes. Together, these data suggest that intracellular proliferation of E. faecalis may contribute to its persistence in the face of a robust immune response, providing a primed reservoir of bacteria for subsequent reinfection.


Subject(s)
Enterococcus faecalis , rab GTP-Binding Proteins , Animals , Endosomes/metabolism , Enterococcus faecalis/metabolism , Lysosomes/metabolism , Mammals , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
16.
Curr Opin Microbiol ; 63: 117-125, 2021 10.
Article in English | MEDLINE | ID: mdl-34333239

ABSTRACT

Biofilm formation is a multifactorial process and often a multi-species endeavour that involves complex signalling networks, chemical gradients, bacterial adhesion, and production or acquisition of matrix components. Antibiotics remain the main choice when treating bacterial biofilm-associated infections despite their intrinsic tolerance to antimicrobials, and propensity for acquisition and rapid dissemination of antimicrobial resistance within the biofilm. Eliminating hard to treat biofilm-associated infections that are antibiotic resistant will demand a holistic and multi-faceted approach, targeting multiple stages of biofilm formation, many of which are already in development. This mini review will highlight the current approaches that are employed to treat bacterial biofilm infections and discuss new approaches in development that have promise to reach clinical practice.


Subject(s)
Anti-Infective Agents , Bacterial Infections , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Biofilms , Humans , Prospective Studies
17.
J Eval Clin Pract ; 27(6): 1398-1402, 2021 12.
Article in English | MEDLINE | ID: mdl-33624429

ABSTRACT

OBJECTIVE: To assess knowledge, attitudes, and perceived barriers (KAP) regarding e-cigarette use counselling among adolescent healthcare clinical staff in an urban system, and to compare results between providers and rooming staff. METHODS: Primary care clinical staff (n = 169) completed an anonymous survey. Descriptive statistics and Chi-square tests were used to summarize data and compare KAP between medical providers and rooming staff. RESULTS: Staff wanted to learn more about e-cigarettes (87.6%). The most common knowledge deficits were how to use the 5As + 5Rs model for tobacco cessation counselling (66.7%) and the chemical content of e-liquids (55.4%), with no differences across groups. Overall, 58% of providers expressed confidence in their ability to talk with adolescent patients about e-cigarette use. The most common barriers to counselling were low knowledge about e-cigarettes (74.0%) and how to refer adolescent patients for cessation support (43.8%). CONCLUSIONS: Provider and rooming staff expressed similar educational needs surrounding e-cigarettes, counselling, and treatment for adolescent patients. Clinical staff expressed confidence in their ability to affect change. There were no differences in the identified knowledge gaps or barriers to care between rooming staff and providers, suggesting that the same educational format can be used to target both groups.


Subject(s)
Electronic Nicotine Delivery Systems , Smoking Cessation , Adolescent , Counseling , Health Knowledge, Attitudes, Practice , Humans , Primary Health Care
18.
Microlife ; 2: uqab005, 2021.
Article in English | MEDLINE | ID: mdl-37223253

ABSTRACT

[This corrects the article DOI: 10.1093/femsml/uqab002.].

19.
Microlife ; 2: uqab002, 2021.
Article in English | MEDLINE | ID: mdl-37223255

ABSTRACT

Membrane vesicles (MVs) contribute to various biological processes in bacteria, including virulence factor delivery, antimicrobial resistance, host immune evasion and cross-species communication. MVs are frequently released from the surface of both Gram-negative and Gram-positive bacteria during growth. In some Gram-positive bacteria, genes affecting MV biogenesis have been identified, but the mechanism of MV formation is unknown. In Enterococcus faecalis, a causative agent of life-threatening bacteraemia and endocarditis, neither mechanisms of MV formation nor their role in virulence has been examined. Since MVs of many bacterial species are implicated in host-pathogen interactions, biofilm formation, horizontal gene transfer, and virulence factor secretion in other species, we sought to identify, describe and functionally characterize MVs from E. faecalis. Here, we show that E. faecalis releases MVs that possess unique lipid and protein profiles, distinct from the intact cell membrane and are enriched in lipoproteins. MVs of E. faecalis are specifically enriched in unsaturated lipids that might provide membrane flexibility to enable MV formation, providing the first insights into the mechanism of MV formation in this Gram-positive organism.

20.
FEMS Microbes ; 2: xtab010, 2021.
Article in English | MEDLINE | ID: mdl-37334228

ABSTRACT

Staphylococcus aureus (SA) is the most common bacterial species in chronic wounds. However, there is a lack of understanding of how SA secretions affect the cell biology during the healing process. We studied the effects of biofilm-secretions from SA strain SA29213 on 3T3 fibroblasts. SA29213 is a chronic wound isolate and widely used as a reference strain. We used a series of concentrations of biofilm-conditioned media (BCM) and found 100% BCM is lethal within 10 h. Cells survived in ≤75% BCM but the rate of closure in scratch wound assays was reduced. Treatment with 75% and 50% BCM caused fibroblasts to change shape and develop dendrite like processes. Prolonged treatment with 75% and 50% BCM reduced cell proliferation and increased the 4n deoxyribonucleic acid cell population with cell cycle arrest. There was also an elevation in the senescence marker beta galactosidase and the number of multinucleated cells. Shorter treatments with 75% and 50% SA BCM caused an increase in cell-cell adhesion and a redistribution of ß-catenin from the cell membrane to the cytoplasm along with a change in the appearance and decrease in size of ZO-1, vinculin and paxillin structures. Fibroblasts in the edge of chronic wounds exposed to the secretions of SA may suffer similar effects such as induction of senescence, reduced proliferation and migration, which may contribute to the delayed healing of these chronic infected wounds.

SELECTION OF CITATIONS
SEARCH DETAIL
...