Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Proteomics ; 23(10): e2200464, 2023 05.
Article in English | MEDLINE | ID: mdl-36781972

ABSTRACT

Gram-negative bacteria produce outer membrane vesicles (OMVs) and contain bacterial cargo including nucleic acids and proteins. The proteome of OMVs can be altered by various factors including bacterial growth stage, growth conditions, and environmental factors. However, it is currently unknown if the mechanism of OMV biogenesis can determine their proteome. In this study, we examined whether the mechanisms of OMV biogenesis influenced the production and protein composition of Pseudomonas aeruginosa OMVs. OMVs were isolated from three P. aeruginosa strains that produced OMVs either by budding alone, by explosive cell lysis, or by both budding and explosive cell lysis. We identified that the mechanism of OMV biogenesis dictated OMV quantity. Furthermore, a global proteomic analysis comparing the proteome of OMVs to their parent bacteria showed significant differences in the identification of proteins in bacteria and OMVs. Finally, we determined that the mechanism of OMV biogenesis influenced the protein composition of OMVs, as OMVs released by distinct mechanisms of biogenesis differed significantly from one another in their proteome and functional enrichment analysis. Overall, our findings reveal that the mechanism of OMV biogenesis is a main factor that determines the OMV proteome which may affect their subsequent biological functions.


Subject(s)
Exosomes , Pseudomonas aeruginosa , Pseudomonas aeruginosa/metabolism , Proteome/metabolism , Proteomics , Exosomes/metabolism , Gram-Negative Bacteria/metabolism , Bacterial Outer Membrane Proteins/metabolism
2.
Proteomics ; : e2300269, 2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37991474

ABSTRACT

Gram-negative bacteria release outer membrane vesicles (OMVs) that contain cargo derived from their parent bacteria. Helicobacter pylori is a Gram-negative human pathogen that produces urease to increase the pH of the surrounding environment to facilitate colonization of the gastric mucosa. However, the effect of acidic growth conditions on the production and composition of H. pylori OMVs is unknown. In this study, we examined the production, composition, and proteome of H. pylori OMVs produced during acidic and neutral pH growth conditions. H. pylori growth in acidic conditions reduced the quantity and size of OMVs produced. Additionally, OMVs produced during acidic growth conditions had increased protein, DNA, and RNA cargo compared to OMVs produced during neutral conditions. Proteomic analysis comparing the proteomes of OMVs to their parent bacteria demonstrated significant differences in the enrichment of beta-lactamases and outer membrane proteins between bacteria and OMVs, supporting that differing growth conditions impacts OMV composition. We also identified differences in the enrichment of proteins between OMVs produced during different pH growth conditions. Overall, our findings reveal that growth of H. pylori at different pH levels is a factor that alters OMV proteomes, which may affect their subsequent functions.

3.
Proteomics ; : e2300087, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38059892

ABSTRACT

The sexually transmitted pathogen Neisseria gonorrhoeae releases membrane vesicles including outer membrane vesicles (OMVs) during infections. OMVs traffic outer membrane molecules, such as the porin PorB and lipo-oligosaccharide (LOS), into host innate immune cells, eliciting programmed cell death pathways, and inflammation. Little is known, however, about the proteome and LOS content of OMVs released by clinical strains isolated from different infection sites, and whether these vesicles similarly activate immune responses. Here, we characterized OMVs from four N. gonorrhoeae isolates and determined their size, abundance, proteome, LOS content, and activation of inflammatory responses in macrophages. The overall proteome of the OMVs was conserved between the four different isolates, which included major outer membrane and periplasm proteins. Despite this, we observed differences in the rate of OMV biogenesis and the relative abundance of membrane proteins and LOS. Consequently, OMVs from clinical isolates induced varying rates of macrophage cell death and the secretion of interleukin-1 family members, such as IL-1α and IL-1ß. Overall, these findings demonstrate that clinical isolates of N. gonorrhoeae utilize membrane vesicles to release proteins and lipids, which affects innate immune responses.

4.
Subcell Biochem ; 97: 101-150, 2021.
Article in English | MEDLINE | ID: mdl-33779916

ABSTRACT

The release of extracellular vesicles (EVs) is a process conserved across the three domains of life. Amongst prokaryotes, EVs produced by Gram-negative bacteria, termed outer membrane vesicles (OMVs), were identified more than 50 years ago and a wealth of literature exists regarding their biogenesis, composition and functions. OMVs have been implicated in benefiting numerous metabolic functions of their parent bacterium. Additionally, OMVs produced by pathogenic bacteria have been reported to contribute to pathology within the disease setting. By contrast, the release of EVs from Gram-positive bacteria, known as membrane vesicles (MVs), has only been widely accepted within the last decade. As such, there is a significant disproportion in knowledge regarding MVs compared to OMVs. Here we provide an overview of the literature regarding bacterial membrane vesicles (BMVs) produced by pathogenic and commensal bacteria. We highlight the mechanisms of BMV biogenesis and their roles in assisting bacterial survival, in addition to discussing their functions in promoting disease pathologies and their potential use as novel therapeutic strategies.


Subject(s)
Gram-Negative Bacteria , Gram-Positive Bacteria , Prokaryotic Cells
5.
Int J Mol Sci ; 22(3)2021 Jan 20.
Article in English | MEDLINE | ID: mdl-33498269

ABSTRACT

Bacterial membrane vesicles (BMVs) are nanoparticles produced by both Gram-negative and Gram-positive bacteria that can function to modulate immunity in the host. Both outer membrane vesicles (OMVs) and membrane vesicles (MVs), which are released by Gram-negative and Gram-positive bacteria, respectively, contain cargo derived from their parent bacterium, including immune stimulating molecules such as proteins, lipids and nucleic acids. Of these, peptidoglycan (PG) and lipopolysaccharide (LPS) are able to activate host innate immune pattern recognition receptors (PRRs), known as NOD-like receptors (NLRs), such as nucleotide-binding oligomerisation domain-containing protein (NOD) 1, NOD2 and NLRP3. NLR activation is a key driver of inflammation in the host, and BMVs derived from both pathogenic and commensal bacteria have been shown to package PG and LPS in order to modulate the host immune response using NLR-dependent mechanisms. Here, we discuss the packaging of immunostimulatory cargo within OMVs and MVs, their detection by NLRs and the cytokines produced by host cells in response to their detection. Additionally, commensal derived BMVs are thought to shape immunity and contribute to homeostasis in the gut, therefore we also highlight the interactions of commensal derived BMVs with NLRs and their roles in limiting inflammatory diseases.


Subject(s)
Bacterial Outer Membrane/immunology , NLR Proteins/metabolism , Nanoparticles/chemistry , Adjuvants, Immunologic/administration & dosage , Animals , Bacterial Outer Membrane/chemistry , Humans , Immunity, Innate , Inflammasomes/immunology , Nanoparticles/metabolism
6.
Proteomics ; 19(1-2): e1800209, 2019 01.
Article in English | MEDLINE | ID: mdl-30488570

ABSTRACT

Gram-negative bacteria release outer membrane vesicles (OMVs) as part of their normal growth that contain a range of cargo from their parent bacterium, including DNA, RNA, and proteins. The protein content of OMVs is suggested to be similar in composition to various sub-cellular locations of their parent bacterium. However, very little is known regarding the effect of bacterial growth stage on the size, content, and selective packaging of proteins into OMVs. In this study, the global proteome of Helicobacter pylori and their OMVs throughout bacterial growth are examined to determine if bacterial growth stage affected OMV cargo composition. Analysis of OMVs produced by H. pylori reveals that bacterial growth stage affects the size, composition, and selection of protein cargo into OMVs. Proteomic analysis identifies that the proteome of H. pylori OMVs is vastly different throughout bacterial growth and that OMVs contain a range of proteins compared to their parent bacteria. In addition, bacterial growth stage affects the ability of OMVs to induce the production of IL-8 by human epithelial cells. Therefore, the findings identify that the size, proteome, and immunogenicity of OMVs produced during various stages of bacterial growth is not comparable. Collectively, these findings highlight the importance of considering the bacterial growth stage from which OMVs are isolated, as this will impact their size, protein composition, and ultimately their biological functions.


Subject(s)
Extracellular Vesicles/metabolism , Helicobacter pylori/metabolism , Proteome/metabolism , Proteomics/methods , Bacterial Outer Membrane Proteins/metabolism , Gram-Negative Bacteria/metabolism
7.
Proc Natl Acad Sci U S A ; 112(5): 1535-40, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25605927

ABSTRACT

Inflammation is critical for host defense, but without appropriate control, it can cause chronic disease or even provoke fatal responses. Here we identify a mechanism that limits the inflammatory response. Probing the responses of macrophages to the key sensory Toll-like receptors, we identify that the Broad-complex, Tramtrack and Bric-a-brac/poxvirus and zinc finger (BTB/POZ), transcriptional regulator promyelocytic leukemia zinc finger (PLZF) limits the expression of inflammatory gene products. In accord with this finding, PLZF-deficient animals express higher levels of potent inflammatory cytokines and mount exaggerated inflammatory responses to infectious stimuli. Temporal quantitation of inflammatory gene transcripts shows increased gene induction in the absence of PLZF. Genome-wide analysis of histone modifications distinguish that PLZF establishes basal activity states of early response genes to maintain immune homeostasis and limit damaging inflammation. We show that PLZF stabilizes a corepressor complex that encompasses histone deacetylase activity to control chromatin. Together with our previous demonstration that PLZF promotes the antiviral response, these results suggest a strategy that could realize one of the major goals of immune therapy to retain immune resistance to pathogens while curbing damaging inflammation.


Subject(s)
Chromatin/metabolism , Inflammation/metabolism , Kruppel-Like Transcription Factors/metabolism , Signal Transduction , Animals , Bacterial Infections/metabolism , Chromatin Immunoprecipitation , Fluorescence Resonance Energy Transfer , Histone Deacetylases/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Promyelocytic Leukemia Zinc Finger Protein , Real-Time Polymerase Chain Reaction
8.
Cell Microbiol ; 18(11): 1518-1524, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27564529

ABSTRACT

Outer membrane vesicles were first described approximately 50 years ago and for many years were considered to be an artifact of bacterial growth. Since that initial discovery, it has become evident that outer membrane vesicles are produced by almost all Gram-negative bacteria as part of their normal growth in addition to driving pathogenesis within the host. More recently, the identification of membrane vesicle (MV) production by some Gram-positive bacteria, parasites, fungi, mycobacteria and infected host cells has significantly broadened the field of MV research and emphasized their importance to pathogenesis. In this review, we will focus on discussing recent advances in the field of bacterial MV biogenesis and the mechanisms whereby they modulate immunity and contribute to pathogenesis. We will highlight findings identifying the contribution of extracellular vesicles produced by Gram-positive bacteria, fungi, parasites, and infected host cells in mediating pathogenesis in addition to the functions of MVs produced by commensal bacteria. Finally, we will discuss recent progress in the development of bacterial MVs as novel vaccines capable of mediating cellular and humoral immune responses.


Subject(s)
Cell-Derived Microparticles/physiology , Gram-Negative Bacteria/physiology , Gram-Negative Bacterial Infections/microbiology , Adaptive Immunity , Animals , Epithelial Cells/microbiology , Gram-Negative Bacteria/immunology , Host-Pathogen Interactions , Humans , Immunity, Innate
9.
Int J Mol Sci ; 18(6)2017 Jun 16.
Article in English | MEDLINE | ID: mdl-28621731

ABSTRACT

The therapeutic potential of extracellular vesicles from eukaryotes has gained strong interest in recent years. However, research into the therapeutic application of their bacterial counterparts, known as bacterial membrane vesicles, is only just beginning to be appreciated. Membrane vesicles (MVs) from both Gram-positive and Gram-negative bacteria offer significant advantages in therapeutic development, including large-scale, cost effective production and ease of molecular manipulation to display foreign antigens. The nanoparticle size of MVs enables their dissemination through numerous tissue types, and their natural immunogenicity and self-adjuvanting capability can be harnessed to induce both cell-mediated and humoral immunity in vaccine design. Moreover, the ability to target MVs to specific tissues through the display of surface receptors raises their potential use as targeted MV-based anti-cancer therapy. This review discusses recent advances in MV research with particular emphasis on exciting new possibilities for the application of MVs in therapeutic design.


Subject(s)
Bacteria/immunology , Bacterial Vaccines/immunology , Cancer Vaccines/immunology , Extracellular Vesicles/immunology , Adaptive Immunity , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/pharmacology , Animals , Bacteria/chemistry , Bacteria/genetics , Bacterial Infections/immunology , Bacterial Infections/prevention & control , Bacterial Vaccines/chemistry , Bacterial Vaccines/genetics , Bioengineering/methods , Cancer Vaccines/chemistry , Cancer Vaccines/genetics , Extracellular Vesicles/chemistry , Extracellular Vesicles/genetics , Genetic Engineering/methods , Humans , Immunity, Innate , Neoplasms/immunology , Neoplasms/prevention & control
10.
Cytokine ; 74(2): 207-12, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25801093

ABSTRACT

The host has developed an array of systems that enables protection against infection and response to injury, ultimately resulting in the generation of a pro-inflammatory response. The most rapid immune response is mediated via the innate immune system, which is comprised of germ line encoded pathogen recognition receptors (PRRs). This PRR mediated system functions by specifically recognizing conserved structures of microbial molecules or products, known as microbial-associated molecular patterns (MAMPs), ultimately enabling transduction of signaling cascades, gene transcription and the development of a pro-inflammatory innate immune response. The intracellular PRRs nucleotide-binding oligomerization domain protein 1 (NOD1) and NOD2 will be the focus of this review. A brief overview of NOD1 and NOD2 and recent advances in the field regarding the intracellular location and mechanisms of NOD1 signaling will be discussed. These new findings have broadened our understanding of the mechanisms whereby NOD1 signaling results in the induction of the cellular degradation pathway of autophagy and the development of pro-inflammatory responses that activate the adaptive immune system.


Subject(s)
Adaptive Immunity , Autophagy/immunology , Nod1 Signaling Adaptor Protein/immunology , Receptors, Pattern Recognition/immunology , Signal Transduction/immunology , Animals , Humans , Nod2 Signaling Adaptor Protein/immunology
11.
Helicobacter ; 20(4): 269-83, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25669590

ABSTRACT

BACKGROUND: Multiple studies have established the importance of the tol-pal gene cluster in bacterial cell membrane integrity and outer membrane vesicle (OMV) formation in Escherichia coli. In contrast, the functions of Tol-Pal proteins in pathogenic organisms, including those of the Epsilonproteobacteria, remain poorly if at all defined. The aim of this study was to characterize the roles of two key components of the Tol-Pal system, TolB and Pal, in OMV formation in the pathogenic bacterium, Helicobacter pylori. METHODS: H. pylori ΔtolB, Δpal and ΔtolBpal mutants, as well as complemented strains, were generated and assessed for changes in morphology and OMV production by scanning electron microscopy and enzyme-linked immunoassay (ELISA), respectively. The protein content and pro-inflammatory properties of OMVs were determined by mass spectroscopy and interleukin-8 (IL-8) ELISA on culture supernatants from OMV-stimulated cells, respectively. RESULTS: H. pylori ΔtolB and Δpal bacteria exhibited aberrant cell morphology and/or flagella biosynthesis. Importantly, the disruption of H. pylori tolB but not pal resulted in a significant increase in OMV production. The OMVs from H. pylori ΔtolB and Δpal bacteria harbored many of the major outer membrane and virulence proteins observed in wild-type (WT) OMVs. Interestingly, ΔtolB, Δpal and ΔtolBpal OMVs induced significantly higher levels of IL-8 production by host cells, compared with WT OMVs. CONCLUSIONS: This work demonstrates that TolB and Pal are important for membrane integrity in H. pylori. Moreover, it shows how H. pylori tolB-pal genes may be manipulated to develop "hypervesiculating" strains for vaccine purposes.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Interleukin-8/metabolism , Periplasmic Proteins/metabolism , Cell Membrane/physiology , Enzyme-Linked Immunospot Assay , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Mass Spectrometry , Microscopy, Electron, Scanning , Periplasmic Proteins/genetics
12.
J Immunol ; 190(7): 3706-15, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23460743

ABSTRACT

Virulent Helicobacter pylori strains that specifically activate signaling in epithelial cells via the innate immune molecule, nucleotide oligomerization domain 1 (NOD1), are more frequently associated with IFN-γ-dependent inflammation and with severe clinical outcomes (i.e., gastric cancer and peptic ulceration). In cell culture models, we showed that H. pylori activation of the NOD1 pathway caused enhanced proinflammatory signaling in epithelial cells in response to IFN-γ stimulation through the direct effects of H. pylori on two components of the IFN-γ signaling pathway, STAT1 and IFN regulatory factor 1 (IRF1). Specifically, H. pylori activation of the NOD1 pathway was shown to increase the levels of STAT1-Tyr(701)/Ser(727) phosphorylation and IRF1 expression/synthesis in cells, resulting in enhanced production of the NOD1- and IFN-γ-regulated chemokines, IL-8- and IFN-γ-induced protein 10, respectively. Consistent with the notion that heightened proinflammatory signaling in epithelial cells may have an impact on disease severity, we observed significantly increased expression levels of NOD1, CXCL8, IRF1, and CXCL10 in human gastric biopsies displaying severe gastritis, when compared with those without gastritis (p < 0.05, p < 0.001, p < 0.01, and p < 0.05, respectively). Interestingly, NOD1, CXCL8, and IRF1 expression levels were also significantly upregulated in gastric tumor tissues, when compared with paired nontumor samples (p < 0.0001, p < 0.05, and p < 0.05, respectively). Thus, we propose that cross-talk between NOD1 and IFN-γ signaling pathways contribute to H. pylori-induced inflammatory responses, potentially revealing a novel mechanism whereby virulent H. pylori strains promote more severe disease.


Subject(s)
Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Helicobacter Infections/metabolism , Interferon-gamma/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Signal Transduction , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cell Line , Chemokines/biosynthesis , Disease Progression , Epithelial Cells/microbiology , Gastric Mucosa/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Interferon Regulatory Factor-1/genetics , Phosphorylation , STAT1 Transcription Factor/metabolism , Transcription, Genetic
13.
Nucleic Acids Res ; 40(16): 8048-58, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22684508

ABSTRACT

Fine-tuning of inflammatory responses by microRNAs (miRNAs) is complex, as they can both enhance and repress expression of pro-inflammatory mediators. In this study, we investigate inflammatory responses following global miRNA depletion, to better define the overall contribution of miRNAs to inflammation. We demonstrate that miRNAs positively regulate Toll-like receptor signaling using inducible Dicer1 deletion and global miRNA depletion. We establish an important contribution of miR-19b in this effect, which potentiates nuclear factor-κB (NF-κB) activity in human and mouse cells. Positive regulation of NF-κB signaling by miR-19b involves the coordinated suppression of a regulon of negative regulators of NF-κB signaling (including A20/Tnfaip3, Rnf11, Fbxl11/Kdm2a and Zbtb16). Transfection of miR-19b mimics exacerbated the inflammatory activation of rheumatoid arthritis primary fibroblast-like synoviocytes, demonstrating its physiological importance in the pathology of this disease. This study constitutes, to our knowledge, the first description of a miR-19 regulon that controls NF-κB signaling, and suggests that targeting this miRNA and linked family members could regulate the activity of NF-κB signaling in inflammation.


Subject(s)
MicroRNAs/metabolism , NF-kappa B/metabolism , Regulon , Signal Transduction , Animals , Bone Marrow Cells/metabolism , Cells, Cultured , Fibroblasts/metabolism , Humans , Inflammation/genetics , Inflammation Mediators/metabolism , Macrophages/metabolism , Mice , Synovial Membrane/cytology , Synovial Membrane/metabolism , Toll-Like Receptors/metabolism
14.
J Parkinsons Dis ; 14(2): 227-244, 2024.
Article in English | MEDLINE | ID: mdl-38427502

ABSTRACT

Parkinson's disease (PD) is an increasingly common neurodegenerative disease. It has been suggested that the etiology of idiopathic PD is complex and multifactorial involving environmental contributions, such as viral or bacterial infections and microbial dysbiosis, in genetically predisposed individuals. With advances in our understanding of the gut-brain axis, there is increasing evidence that the intestinal microbiota and the mammalian immune system functionally interact. Recent findings suggest that a shift in the gut microbiome to a pro-inflammatory phenotype may play a role in PD onset and progression. While there are links between gut bacteria, inflammation, and PD, the bacterial products involved and how they traverse the gut lumen and distribute systemically to trigger inflammation are ill-defined. Mechanisms emerging in other research fields point to a role for small, inherently stable vesicles released by Gram-negative bacteria, called outer membrane vesicles in disease pathogenesis. These vesicles facilitate communication between bacteria and the host and can shuttle bacterial toxins and virulence factors around the body to elicit an immune response in local and distant organs. In this perspective article, we hypothesize a role for bacterial outer membrane vesicles in PD pathogenesis. We present evidence suggesting that these outer membrane vesicles specifically from Gram-negative bacteria could potentially contribute to PD by traversing the gut lumen to trigger local, systemic, and neuroinflammation. This perspective aims to facilitate a discussion on outer membrane vesicles in PD and encourage research in the area, with the goal of developing strategies for the prevention and treatment of the disease.


Subject(s)
Gastrointestinal Microbiome , Neurodegenerative Diseases , Parkinson Disease , Animals , Humans , Parkinson Disease/pathology , Bacterial Outer Membrane/pathology , Inflammation/complications , Gastrointestinal Microbiome/physiology , Mammals
15.
Nat Rev Microbiol ; 21(7): 415-430, 2023 07.
Article in English | MEDLINE | ID: mdl-36932221

ABSTRACT

Extracellular vesicles are produced by species across all domains of life, suggesting that vesiculation represents a fundamental principle of living matter. In Gram-negative bacteria, membrane vesicles (MVs) can originate either from blebs of the outer membrane or from endolysin-triggered explosive cell lysis, which is often induced by genotoxic stress. Although less is known about the mechanisms of vesiculation in Gram-positive and Gram-neutral bacteria, recent research has shown that both lysis and blebbing mechanisms also exist in these organisms. Evidence has accumulated over the past years that different biogenesis routes lead to distinct types of MV with varied structure and composition. In this Review, we discuss the different types of MV and their potential cargo packaging mechanisms. We summarize current knowledge regarding how MV composition determines their various functions including support of bacterial growth via the disposal of waste material, nutrient scavenging, export of bioactive molecules, DNA transfer, neutralization of phages, antibiotics and bactericidal functions, delivery of virulence factors and toxins to host cells and inflammatory and immunomodulatory effects. We also discuss the advantages of MV-mediated secretion compared with classic bacterial secretion systems and we introduce the concept of quantal secretion.


Subject(s)
Bacteriophages , Extracellular Vesicles , Bacteria , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Gram-Negative Bacteria/metabolism , Virulence Factors/metabolism , Cell Membrane/metabolism
16.
Microbiol Spectr ; : e0517922, 2023 Mar 22.
Article in English | MEDLINE | ID: mdl-36946779

ABSTRACT

Outer membrane vesicles (OMVs) produced by Gram-negative bacteria package various cargo, including DNA that can be transferred to other bacteria or to host cells. OMV-associated DNA has been implicated in mediating horizontal gene transfer (HGT) between bacteria, which includes the dissemination of antibiotic resistance genes within and between bacterial species. Despite the known ability of OMVs to mediate HGT, the mechanisms of DNA packaging into OMVs remain poorly characterized, as does the effect of bacterial growth conditions on the DNA cargo composition of OMVs and their subsequent abilities to mediate HGT. In this study, we examined the DNA content of OMVs produced by the opportunistic pathogen Pseudomonas aeruginosa grown in either planktonic or biofilm conditions. Analysis of planktonic growth-derived OMVs revealed their ability to package and protect plasmid DNA from DNase degradation and to transfer plasmid-encoded antibiotic resistance genes to recipient, antibiotic-sensitive P. aeruginosa bacteria at a greater efficiency than transformation with plasmid alone. Comparisons of planktonic and biofilm-derived P. aeruginosa OMVs demonstrated that biofilm-derived OMVs were smaller but were associated with more plasmid DNA than planktonic-derived OMVs. Additionally, biofilm-derived P. aeruginosa OMVs were more efficient in the transformation of competent P. aeruginosa bacteria, compared to transformations with an equivalent number of planktonic-derived OMVs. The findings of this study highlight the importance of bacterial growth conditions for the packaging of DNA within P. aeruginosa OMVs and their ability to facilitate HGT, thus contributing to the spread of antibiotic resistance genes between P. aeruginosa bacteria. IMPORTANCE Bacterial membrane vesicles (BMVs) mediate interbacterial communication, and their ability to package DNA specifically contributes to biofilm formation, antibiotic resistance, and HGT between bacteria. However, the ability of P. aeruginosa OMVs to mediate HGT has not yet been demonstrated. Here, we reveal that P. aeruginosa planktonic and biofilm-derived OMVs can deliver plasmid-encoded antibiotic resistance to recipient P. aeruginosa. Additionally, we demonstrated that P. aeruginosa biofilm-derived OMVs were associated with more plasmid DNA compared to planktonic-derived OMVs and were more efficient in the transfer of plasmid DNA to recipient bacteria. Overall, this demonstrated the ability of P. aeruginosa OMVs to facilitate the dissemination of antibiotic resistance genes, thereby enabling the survival of susceptible bacteria during antibiotic treatment. Investigating the roles of biofilm-derived BMVs may contribute to furthering our understanding of the role of BMVs in HGT and the spread of antibiotic resistance in the environment.

17.
Cells ; 12(20)2023 10 20.
Article in English | MEDLINE | ID: mdl-37887335

ABSTRACT

Milk is a complex biological fluid that has high-quality proteins including growth factors and also contains extracellular vesicles (EVs). EVs are a lipid bilayer containing vesicles that contain proteins, metabolites and nucleic acids. Several studies have proposed that EVs in cow milk can survive the gut and can illicit cross-species communication in the consuming host organism. In this study, we isolated and characterized extracellular vesicles from the raw milk of the four species of the Bovidae family, namely cow, sheep, goat and buffalo, that contribute 99% of the total milk consumed globally. A comparative proteomic analysis of these vesicles was performed to pinpoint their potential functional role in health and disease. Vesicles sourced from buffalo and cow milk were particularly enriched with proteins implicated in modulating the immune system. Furthermore, functional studies were performed to determine the anti-cancer effects of these vesicles. The data obtained revealed that buffalo-milk-derived EVs induced significantly higher cell death in colon cancer cells. Overall, the results from this study highlight the potent immunoregulatory and anti-cancer nature of EVs derived from the milk of Bovidae family members.


Subject(s)
Extracellular Vesicles , Milk , Female , Cattle , Animals , Sheep , Buffaloes , Proteomics/methods , Extracellular Vesicles/metabolism , Goats
18.
Helicobacter ; 17(1): 68-76, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22221619

ABSTRACT

BACKGROUND: Growth of Helicobacter pyloriin vitro depends on supplementation of the medium with blood or serum. However, these supplements often require frozen storage and can show batch-to-batch variation, resulting in differences in bacterial growth. In this study, we introduce the use of a commercially available, lipid-rich supplement called AlbuMAX II(®) (Gibco BRL, Grand Island, NY, USA) for use as a serum/blood replacement for H. pylori culture. MATERIALS AND METHODS: The growth of H. pylori on solid and liquid media was examined by comparing growth after supplementation with horse blood, fetal calf serum, ß-cyclodextrin or AlbuMAX II(®) (Gibco BRL). Human gastric adenocarcinoma (AGS) cellular responses to H. pylori were measured by NF-κB luciferase assays and IL-8 ELISA. RESULTS: We show that the growth of H. pylori on both solid and liquid media containing AlbuMAX II(®) (Gibco BRL) were comparable to levels obtained on blood agar or liquid media supplemented with serum. Growth was consistently higher in media supplemented with AlbuMAX II(®) (Gibco BRL) than media containing ß-cyclodextrin. Furthermore, bacteria grown in AlbuMAX II(®) (Gibco BRL) induced proinflammatory responses in AGS cells. CONCLUSIONS: AlbuMAX II(®) (Gibco BRL) can be used as a serum/blood replacement for the cultivation of H. pylori in solid and liquid media. This medium could be useful for an improved understanding of H. pylori metabolism or for antigen production. Furthermore, AlbuMAX II(®) (Gibco BRL) may be suitable for use in remote locations, particularly in areas where frozen storage of serum may be a problem.


Subject(s)
Culture Media , Helicobacter pylori/growth & development , Agar , Cell Line, Tumor , HEK293 Cells , Helicobacter pylori/metabolism , Humans , Serum
19.
Methods Mol Biol ; 2523: 1-8, 2022.
Article in English | MEDLINE | ID: mdl-35759187

ABSTRACT

Detection of microbes by the host is essential to restrict microbial colonization, to clear pathogens, and to mount adapted defense reactions, and thus is the key function of the innate immune systems of plants and mammals. Here we provide an introduction into pathogen recognition by the innate immune system of both plants and animals. We will particularly focus on the concept of effector-triggered immunity, and similarities and differences in its function between plants and animals.


Subject(s)
Plant Immunity , Plants , Animals , Immunity, Innate , Mammals , Signal Transduction
20.
Methods Mol Biol ; 2523: 43-61, 2022.
Article in English | MEDLINE | ID: mdl-35759190

ABSTRACT

Bacterial membrane vesicles (BMVs) released by Gram-negative and Gram-positive bacteria are a bona fide secretion system that enable the dissemination of bacterial effector molecules, and can trigger a range of responses in the host. The study of BMV production, composition, and functions can give insights into their roles in mediating bacterial survival, pathogenesis, and disease. Furthermore, BMVs can be harnessed to develop cutting-edge nano-therapeutics including targeted chemotherapy delivery, antimicrobials, and novel vaccines. Here we describe routine methods that can be used for small- or large-scale production, isolation, and purification of outer membrane vesicles produced by Gram-negative bacteria, and membrane vesicles produced by Gram-positive bacteria, which we collectively refer to as BMVs. We discuss methods that can be used to visualize BMVs by electron microscopy, and to quantify their DNA, RNA, and protein cargo. We outline a method for the fluorescent labeling of BMVs that can be applied to examine their ability to interact with and enter host cells using a range of in vitro and in vivo biological assays. Finally, we provide a cell culture-based method that can be used to examine a range of immunogenic properties of BMVs, including their cytotoxicity, ability to activate pathogen-recognition receptors (PRRs), induce autophagy and cytokine responses, and modulate cellular pathways.


Subject(s)
Bacteria , Gram-Negative Bacteria , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria , Membranes
SELECTION OF CITATIONS
SEARCH DETAIL