Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 195
Filter
1.
Proc Natl Acad Sci U S A ; 121(28): e2402543121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38959031

ABSTRACT

The outer membrane (OM) of gram-negative bacteria serves as a vital organelle that is densely populated with OM proteins (OMPs) and plays pivotal roles in cellular functions and virulence. The assembly and insertion of these OMPs into the OM represent a fundamental process requiring specialized molecular chaperones. One example is the translocation and assembly module (TAM), which functions as a transenvelope chaperone promoting the folding of specific autotransporters, adhesins, and secretion systems. The catalytic unit of TAM, TamA, comprises a catalytic ß-barrel domain anchored within the OM and three periplasmic polypeptide-transport-associated (POTRA) domains that recruit the TamB subunit. The latter acts as a periplasmic ladder that facilitates the transport of unfolded OMPs across the periplasm. In addition to their role in recruiting the auxiliary protein TamB, our data demonstrate that the POTRA domains mediate interactions with the inner surface of the OM, ultimately modulating the membrane properties. Through the integration of X-ray crystallography, molecular dynamic simulations, and biomolecular interaction methodologies, we located the membrane-binding site on the first and second POTRA domains. Our data highlight a binding preference for phosphatidylglycerol, a minor lipid constituent present in the OM, which has been previously reported to facilitate OMP assembly. In the context of the densely OMP-populated membrane, this association may serve as a mechanism to secure lipid accessibility for nascent OMPs through steric interactions with existing OMPs, in addition to creating favorable conditions for OMP biogenesis.


Subject(s)
Bacterial Outer Membrane Proteins , Escherichia coli Proteins , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/chemistry , Escherichia coli/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Models, Molecular , Molecular Chaperones/metabolism , Molecular Chaperones/chemistry , Periplasm/metabolism , Protein Domains , Protein Folding
2.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2258-2269, 2024 Jul 25.
Article in Chinese | MEDLINE | ID: mdl-39044589

ABSTRACT

This study aims to prepare bacterial outer membrane vesicles (OMVs) with anti-glypican-3 (GPC3) single-chain antibody and analyze their targeting effects on Hep G2 hepatocellular carcinoma (HCC) cells and tissue. The recombinant plasmid pET28a-Hbp-hGC 33-scFv was constructed by ligating Hbp-hGC 33-scFv to pET28a. Western blotting was employed to determine the prokaryotic expression of the fusion protein Hbp-hGC 33-scFv, on the basis of which the optimal induction conditions were determined. Hbp-hGC 33-OMVs secreted from the recombinant expressing strains were collected by ultrafiltration concentration and then characterized. The localization of Hbp-hGC 33-scFv in bacteria and Hbp-hGC 33-OMVs was analyzed by immune electron microscopy. The binding of Hbp-hGC 33-scFv to Hep G2 cells was observed by immunofluorescence. The Hep G2 tumor-bearing mouse model was established, and the targeted retention of Hbp-hGC 33-OMVs in the tumor site of mice was observed by a fluorescence imaging system in vivo. The results showed that the actual molecular weight of the fusion protein was 175.3 kDa, and the optimal induction conditions were as follows: OD600=0.5, IPTG added at a final concentration of 0.5 mmol/L, and overnight induction at 16 ℃. The prepared Hbp-hGC 33-OMVs were irregular spherical structures with an average particle size of (112.3±4.6) nm, expressing OmpC, OmpA, and the fusion protein Hbp-hGC 33-scFv. The Hbp-hGC 33-OMVs prepared in this study demonstrated stronger ability of binding to Hep G2 cells than the wild-type OMVs (P=0.008). All the data indicated that Hbp-hGC 33-OMVs with anti-GPC3 single-chain antibody were successfully prepared and could be used for research on the targeted therapy of hepatocellular carcinoma.


Subject(s)
Bacterial Outer Membrane , Carcinoma, Hepatocellular , Glypicans , Liver Neoplasms , Single-Chain Antibodies , Single-Chain Antibodies/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/chemistry , Animals , Mice , Humans , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane/immunology , Hep G2 Cells , Glypicans/immunology , Glypicans/metabolism , Glypicans/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/biosynthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Drug Delivery Systems , Mice, Nude
3.
Proc Natl Acad Sci U S A ; 121(30): e2403460121, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39008666

ABSTRACT

Autonomous nanorobots represent an advanced tool for precision therapy to improve therapeutic efficacy. However, current nanorobotic designs primarily rely on inorganic materials with compromised biocompatibility and limited biological functions. Here, we introduce enzyme-powered bacterial outer membrane vesicle (OMV) nanorobots. The immobilized urease on the OMV membrane catalyzes the decomposition of bioavailable urea, generating effective propulsion for nanorobots. This OMV nanorobot preserves the unique features of OMVs, including intrinsic biocompatibility, immunogenicity, versatile surface bioengineering for desired biofunctionalities, capability of cargo loading and protection. We present OMV-based nanorobots designed for effective tumor therapy by leveraging the membrane properties of OMVs. These involve surface bioengineering of robotic body with cell-penetrating peptide for tumor targeting and penetration, which is further enhanced by active propulsion of nanorobots. Additionally, OMV nanorobots can effectively safeguard the loaded gene silencing tool, small interfering RNA (siRNA), from enzymatic degradation. Through systematic in vitro and in vivo studies using a rodent model, we demonstrate that these OMV nanorobots substantially enhanced siRNA delivery and immune stimulation, resulting in the utmost effectiveness in tumor suppression when juxtaposed with static groups, particularly evident in the orthotopic bladder tumor model. This OMV nanorobot opens an inspiring avenue to design advanced medical robots with expanded versatility and adaptability, broadening their operation scope in practical biomedical domains.


Subject(s)
Bacterial Outer Membrane , Animals , Humans , Bacterial Outer Membrane/metabolism , Mice , Robotics/methods , Urease/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism
4.
J Nanobiotechnology ; 22(1): 310, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831378

ABSTRACT

Radiotherapy (RT), including external beam radiation therapy (EBRT) and radionuclide therapy (RNT), realizes physical killing of local tumors and activates systemic anti-tumor immunity. However, these effects need to be further strengthened and the difference between EBRT and RNT should be discovered. Herein, bacterial outer membrane (OM) was biomineralized with manganese oxide (MnO2) to obtain OM@MnO2-PEG nanoparticles for enhanced radio-immunotherapy via amplifying EBRT/RNT-induced immunogenic cell death (ICD) and cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) activation. OM@MnO2-PEG can react with H2O2 and then gradually produce O2, Mn2+ and OM fragments in the tumor microenvironment. The relieved tumor hypoxia improves the radio-sensitivity of tumor cells, resulting in enhanced ICD and DNA damage. Mn2+ together with the DNA fragments in the cytoplasm activate the cGAS-STING pathway, further exhibiting a positive role in various aspects of innate immunity and adaptive immunity. Besides, OM fragments promote tumor antigen presentation and anti-tumor macrophages polarization. More importantly, our study reveals that OM@MnO2-PEG-mediated RNT triggers much stronger cGAS-STING pathway-involved immunotherapy than that of EBRT, owing to the duration difference of RT. Therefore, this study develops a powerful sensitizer of radio-immunotherapy and uncovers some differences between EBRT and RNT in the activation of cGAS-STING pathway-related anti-tumor immunity.


Subject(s)
Bacterial Outer Membrane , Immunotherapy , Manganese Compounds , Membrane Proteins , Nucleotidyltransferases , Oxides , Nucleotidyltransferases/metabolism , Manganese Compounds/chemistry , Membrane Proteins/metabolism , Mice , Immunotherapy/methods , Oxides/chemistry , Animals , Bacterial Outer Membrane/metabolism , Tumor Microenvironment , Cell Line, Tumor , Signal Transduction , Humans , Radiotherapy/methods , Nanoparticles/chemistry , Biomineralization , Immunogenic Cell Death/drug effects , Neoplasms/therapy , Hydrogen Peroxide/metabolism , Immunity, Innate
5.
Science ; 384(6701): eado0713, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38870284

ABSTRACT

Bacteria can repurpose their own bacteriophage viruses (phage) to kill competing bacteria. Phage-derived elements are frequently strain specific in their killing activity, although there is limited evidence that this specificity drives bacterial population dynamics. Here, we identified intact phage and their derived elements in a metapopulation of wild plant-associated Pseudomonas genomes. We discovered that the most abundant viral cluster encodes a phage remnant resembling a phage tail called a tailocin, which bacteria have co-opted to kill bacterial competitors. Each pathogenic Pseudomonas strain carries one of a few distinct tailocin variants that target the variable polysaccharides in the outer membrane of co-occurring pathogenic Pseudomonas strains. Analysis of herbarium samples from the past 170 years revealed that the same tailocin and bacterial receptor variants have persisted in Pseudomonas populations. These results suggest that tailocin genetic diversity can be mined to develop targeted "tailocin cocktails" for microbial control.


Subject(s)
Bacteriocins , Pseudomonas Phages , Pseudomonas , Viral Tail Proteins , Antibiosis , Bacterial Outer Membrane/metabolism , Bacteriocins/genetics , Bacteriocins/metabolism , Genetic Variation , Genome, Bacterial , Polysaccharides, Bacterial/metabolism , Pseudomonas/metabolism , Pseudomonas/virology , Pseudomonas Phages/genetics , Pseudomonas Phages/metabolism , Viral Tail Proteins/metabolism , Viral Tail Proteins/genetics , Phage Therapy/methods
6.
Microbiol Res ; 285: 127774, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38833829

ABSTRACT

Extended-spectrumß-lactam producing Escherichia coli (ESBL-EC) readily colonizes live poultry and serves as a major source of contamination in retail chicken meat, posing significant threats to public health. This study aims to investigate the impact of inappropriate antibiotic use on the dissemination and exacerbation of antibiotic resistance in ESBL-EC and explore the underlying molecular mechanisms. Through experimental analysis, we propose a hypothesis that inappropriate antibiotic use may exacerbate resistance by affecting vesicle formation and protein secretion. Experimental results demonstrate that under the influence of amoxicillin, the concentration of proteins secreted in outer membrane vehicles (OMVs) by ESBL-EC significantly increases, along with a significant upregulation in the expression of the CTX-M-55-type Extended-spectrum beta-lactamase (CTX-M-55). Proteomic analysis and differential gene knockout experiments identified the key protein YdcZ, associated with OMVs formation and protein transportation in ESBL-EC under amoxicillin treatment. Further investigations reveal direct interactions between YdcZ and other proteins (YdiH and BssR). Upon ydcz gene knockout, a significant decrease in protein concentration within OMVs is observed, accompanied by a noticeable reduction in protection against sensitive bacteria. These findings suggest a critical role of YdcZ in regulating the process of protein transportation to OMVs in ESBL-EC under the influence of amoxicillin. In summary, our research uncovers the significant role of inappropriate antibiotic use in promoting the secretion of OMVs by ESBL-EC, aiding the survival of antibiotic-sensitive bacteria in the vicinity of infection sites. These findings provide new insights into the mechanisms underlying antibiotic-induced bacterial resistance dissemination and offer novel avenues for exploring prevention and control strategies against bacterial resistance propagation.


Subject(s)
Amoxicillin , Anti-Bacterial Agents , Escherichia coli Proteins , Escherichia coli , Protein Transport , beta-Lactamases , Anti-Bacterial Agents/pharmacology , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , beta-Lactamases/metabolism , beta-Lactamases/genetics , Amoxicillin/pharmacology , Animals , Microbial Sensitivity Tests , Proteomics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/genetics , Chickens/microbiology , Drug Resistance, Bacterial , Bacterial Outer Membrane/drug effects , Bacterial Outer Membrane/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/drug therapy
7.
Nat Commun ; 15(1): 4733, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830951

ABSTRACT

Polymyxins are gram-negative antibiotics that target lipid A, the conserved membrane anchor of lipopolysaccharide in the outer membrane. Despite their clinical importance, the molecular mechanisms underpinning polymyxin activity remain unresolved. Here, we use surface plasmon resonance to kinetically interrogate interactions between polymyxins and lipid A and derive a phenomenological model. Our analyses suggest a lipid A-catalyzed, three-state mechanism for polymyxins: transient binding, membrane insertion, and super-stoichiometric cluster accumulation with a long residence time. Accumulation also occurs for brevicidine, another lipid A-targeting antibacterial molecule. Lipid A modifications that impart polymyxin resistance and a non-bactericidal polymyxin derivative exhibit binding that does not evolve into long-lived species. We propose that transient binding to lipid A permeabilizes the outer membrane and cluster accumulation enables the bactericidal activity of polymyxins. These findings could establish a blueprint for discovery of lipid A-targeting antibiotics and provide a generalizable approach to study interactions with the gram-negative outer membrane.


Subject(s)
Anti-Bacterial Agents , Lipid A , Polymyxin B , Surface Plasmon Resonance , Polymyxin B/pharmacology , Polymyxin B/metabolism , Lipid A/metabolism , Lipid A/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Microbial Sensitivity Tests , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane/drug effects , Kinetics
8.
Microb Pathog ; 193: 106749, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38879140

ABSTRACT

Bacteria-derived outer membrane vesicles (OMVs) can be engineered to incorporate foreign antigens. This study explored the potential of ClearColi™-derived OMVs as a natural adjuvant and a carrier (recombinant OMVs or rOMVs) for development of an innovative therapeutic vaccine candidate harboring HIV-1 Nef and Nef-Tat antigens. Herein, the rOMVs containing CytolysinA (ClyA)-Nef and ClyA-Nef-Tat fusion proteins were isolated from ClearColi™ strain. The presence of Nef and Nef-Tat proteins on their surface (rOMVNef and rOMVNef-Tat) was confirmed by western blotting after proteinase K treatment. Immune responses induced by Nef and Nef-Tat proteins emulsified with Montanide® ISA720 or mixed with OMVs, and also rOMVNef and rOMVNef-Tat were investigated in BALB/c mice. Additionally, the potency of splenocytes exposed to single-cycle replicable (SCR) HIV-1 virions was assessed for the secretion of cytokines in vitro. Our findings showed that the rOMVs as an antigen carrier (rOMVNef and rOMVNef-Tat) induced higher levels of IgG2a, IFN-γ and granzyme B compared to OMVs as an adjuvant (Nef + OMV and Nef-Tat + OMV), and also Montanide® ISA720 (Nef + Montanide and Nef-Tat + Montanide). Moreover, IFN-γ level in splenocytes isolated from mice immunized with rOMVNef-Tat was higher than other regimens after exposure to SCR virions. Generally, ClearColi™-derived rOMVs can serve as potent carriers for developing effective vaccines against HIV-1 infection.


Subject(s)
AIDS Vaccines , Adjuvants, Immunologic , HIV Infections , HIV-1 , Mice, Inbred BALB C , nef Gene Products, Human Immunodeficiency Virus , Animals , AIDS Vaccines/immunology , AIDS Vaccines/genetics , HIV-1/genetics , HIV-1/immunology , nef Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/immunology , Mice , Adjuvants, Immunologic/administration & dosage , HIV Infections/prevention & control , HIV Infections/immunology , Female , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/immunology , Cytokines/metabolism , Immunoglobulin G/blood , HIV Antibodies/immunology , Bacterial Outer Membrane/metabolism , Vaccine Development , Humans , Drug Carriers , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Spleen/immunology
9.
J Proteome Res ; 23(7): 2505-2517, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38845157

ABSTRACT

Escherichia coli Nissle 1917 (EcN 1917) exhibits distinct tumor-targeting activity, and early studies demonstrated that outer membrane vesicles (OMVs) mediate bacteria-host interactions. To decipher the molecular mechanism underlying the interaction between EcN 1917 and host cells via OMV-mediated communication, we investigated the phenotypic changes in Caco-2 cells perturbed by EcN 1917-derived OMVs and constructed proteomic maps of the EcN 1917-derived OMV components and OMV-perturbed host cells. Our findings revealed that the size of the EcN 1917-derived OMV proteome increased 4-fold. Treatment with EcN 1917-derived OMVs altered the proteomic and phosphoproteomic profiles of host cells. Importantly, for the first time, we found that treatment with EcN 1917-derived OMVs inhibited cancer cell migration by suppressing the expression of ANXA9. In addition, phosphoproteomic data suggested that the ErbB pathway may be involved in OMV-mediated cell migration. Taken together, our study provides valuable data for further investigations of OMV-mediated bacteria-host interactions and offers great insights into the underlying mechanism of probiotic-assisted colorectal cancer therapy.


Subject(s)
Cell Movement , Escherichia coli , Proteome , Proteomics , Humans , Caco-2 Cells , Proteomics/methods , Escherichia coli/metabolism , Proteome/analysis , Proteome/metabolism , Bacterial Outer Membrane/metabolism
10.
Nat Commun ; 15(1): 4389, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782915

ABSTRACT

Members of the Omp85 superfamily of outer membrane proteins (OMPs) found in Gram-negative bacteria, mitochondria and chloroplasts are characterized by a distinctive 16-stranded ß-barrel transmembrane domain and at least one periplasmic POTRA domain. All previously studied Omp85 proteins promote critical OMP assembly and/or protein translocation reactions. Pseudomonas aeruginosa PlpD is the prototype of an Omp85 protein family that contains an N-terminal patatin-like (PL) domain that is thought to be translocated across the OM by a C-terminal ß-barrel domain. Challenging the current dogma, we find that the PlpD PL-domain resides exclusively in the periplasm and, unlike previously studied Omp85 proteins, PlpD forms a homodimer. Remarkably, the PL-domain contains a segment that exhibits unprecedented dynamicity by undergoing transient strand-swapping with the neighboring ß-barrel domain. Our results show that the Omp85 superfamily is more structurally diverse than currently believed and suggest that the Omp85 scaffold was utilized during evolution to generate novel functions.


Subject(s)
Bacterial Outer Membrane Proteins , Protein Multimerization , Pseudomonas aeruginosa , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Periplasm/metabolism , Protein Domains , Bacterial Outer Membrane/metabolism , Models, Molecular , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics
11.
J Chem Inf Model ; 64(10): 4204-4217, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38733348

ABSTRACT

Membranes─cells' essential scaffolds─are valid molecular targets for substances with an antimicrobial effect. While certain substances, such as octenidine, have been developed to target membranes for antimicrobial purposes, the recently reported molecule, fabimycin (F2B)─a novel agent targeting drug-resistant Gram-negative bacteria─has not received adequate attention regarding its activity on membranes in the literature. The following study aims to investigate the effects of F2B on different bacterial membrane models, including simple planar bilayers and more complex bilayer systems that mimic the Escherichia coli shell equipped with double inner and outer bilayers. Our results show that F2B exhibited more pronounced interactions with bacterial membrane systems compared to the control PC system. Furthermore, we observed significant changes in local membrane property homeostasis in both the inner and outer membrane models, specifically in the case of lateral diffusion, membrane thickness, and membrane resilience (compressibility, tilt). Finally, our results showed that the effect of F2B differed in a complex system and a single membrane system. Our study provides new insights into the multifaceted activity of F2B, demonstrating its potential to disrupt bacterial membrane homeostasis, indicating that its activity extends the currently known mechanism of FabI enzyme inhibition. This disruption, coupled with the ability of F2B to penetrate the outer membrane layers, sheds new light on the behavior of this antimicrobial molecule. This highlights the importance of the interaction with the membrane, crucial in combating bacterial infections, particularly those caused by drug-resistant strains.


Subject(s)
Cell Membrane , Lipid Bilayers , Molecular Dynamics Simulation , Cell Membrane/metabolism , Cell Membrane/drug effects , Lipid Bilayers/metabolism , Lipid Bilayers/chemistry , Escherichia coli/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane/drug effects
12.
Microbiol Spectr ; 12(6): e0316823, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38722177

ABSTRACT

Vitamin B12 (B12) serves as a critical cofactor within mycobacterial metabolism. While some pathogenic strains can synthesize B12 de novo, others rely on host-acquired B12. In this investigation, we studied the transport of vitamin B12 in Mycobacterium marinum using B12-auxotrophic and B12-sensitive strains by deleting metH or metE, respectively. These two enzymes rely on B12 in different ways to function as methionine synthases. We used these strains to select mutants affecting B12 scavenging and confirmed their phenotypes during growth experiments in vitro. Our analysis of B12 uptake mechanisms revealed that membrane lipids and cell wall integrity play an essential role in cell envelope transport. Furthermore, we identified a potential transcription regulator that responds to B12. Our study demonstrates that M. marinum can take up exogenous B12 and that altering mycobacterial membrane integrity affects B12 uptake. Finally, during zebrafish infection using B12-auxotrophic and B12-sensitive strains, we found that B12 is available for virulent mycobacteria in vivo.IMPORTANCEOur study investigates how mycobacteria acquire essential vitamin B12. These microbes, including those causing tuberculosis, face challenges in nutrient uptake due to their strong outer layer. We focused on Mycobacterium marinum, similar to TB bacteria, to uncover its vitamin B12 absorption. We used modified strains unable to produce their own B12 and discovered that M. marinum can indeed absorb it from the environment, even during infections. Changes in the outer layer composition affect this process, and genes related to membrane integrity play key roles. These findings illuminate the interaction between mycobacteria and their environment, offering insights into combatting diseases like tuberculosis through innovative strategies. Our concise research underscores the pivotal role of vitamin B12 in microbial survival and its potential applications in disease control.


Subject(s)
Bacterial Outer Membrane , Mycobacterium marinum , Vitamin B 12 , Zebrafish , Mycobacterium marinum/genetics , Mycobacterium marinum/metabolism , Vitamin B 12/metabolism , Animals , Zebrafish/microbiology , Bacterial Outer Membrane/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Cell Membrane Permeability , Biological Transport , Cell Membrane/metabolism , Mycobacterium Infections, Nontuberculous/microbiology
13.
J Phys Chem B ; 128(22): 5371-5377, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38787347

ABSTRACT

The cell envelope of Gram-negative bacteria is composed of an outer membrane (OM) and an inner membrane (IM) and a peptidoglycan cell wall (CW) between them. Combined with Braun's lipoprotein (Lpp), which connects the OM and the CW, and numerous membrane proteins that exist in both OM and IM, the cell envelope creates a mechanically stable environment that resists various physical and chemical perturbations to the cell, including turgor pressure caused by the solute concentration difference between the cytoplasm of the cell and the extracellular environment. Previous computational studies have explored how individual components (OM, IM, and CW) can resist turgor pressure although combinations of them have been less well studied. To that end, we constructed multiple OM-CW systems, including the Lpp connections with the CW under increasing degrees of strain. The results show that the OM can effectively resist the tension imposed by the CW, shrinking by only 3-5% in area even when the CW is stretched to 2.5× its relaxed area. The area expansion modulus of the system increases with increasing CW strain, although the OM remains a significant contributor to the envelope's mechanical stability. Additionally, we find that when the protein TolC is embedded in the OM, its stiffness increases.


Subject(s)
Bacterial Outer Membrane Proteins , Cell Wall , Peptidoglycan , Cell Wall/chemistry , Cell Wall/metabolism , Peptidoglycan/chemistry , Peptidoglycan/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane/chemistry , Bacterial Outer Membrane/metabolism , Molecular Dynamics Simulation
14.
J Microbiol ; 62(4): 261-275, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38816673

ABSTRACT

Lipopolysaccharide (LPS) is a critical component of the extracellular leaflet within the bacterial outer membrane, forming an effective physical barrier against environmental threats in Gram-negative bacteria. After LPS is synthesized and matured in the bacterial cytoplasm and the inner membrane (IM), LPS is inserted into the outer membrane (OM) through the ATP-driven LPS transport (Lpt) pathway, which is an energy-intensive process. A trans-envelope complex that contains seven Lpt proteins (LptA-LptG) is crucial for extracting LPS from the IM and transporting it across the periplasm to the OM. The last step in LPS transport involves the mediation of the LptDE complex, facilitating the insertion of LPS into the outer leaflet of the OM. As the Lpt system plays an essential role in maintaining the impermeability of the OM via LPS decoration, the interactions between these interconnected subunits, which are meticulously regulated, may be potential targets for the development of new antibiotics to combat multidrug-resistant Gram-negative bacteria. In this review, we aimed to provide an overview of current research concerning the structural interactions within the Lpt system and their implications to clarify the function and regulation of LPS transport in the overall process of OM biogenesis. Additionally, we explored studies on the development of therapeutic inhibitors of LPS transport, the factors that limit success, and future prospects.


Subject(s)
Anti-Bacterial Agents , Gram-Negative Bacteria , Lipopolysaccharides , Lipopolysaccharides/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/chemistry , Gram-Negative Bacteria/metabolism , Gram-Negative Bacteria/drug effects , Biological Transport , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/chemistry , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/chemistry , Bacterial Outer Membrane/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial
15.
Eur J Pharm Biopharm ; 200: 114336, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38795784

ABSTRACT

Antimicrobial resistance is becoming more prominent day after day due to a number of mechanisms by microbes, especially the sophisticated biological barriers of bacteria, especially in Gram-negatives. There, the lipopolysaccharides (LPS) layer is a unique component of the outer leaflet of the outer membrane which is highly impermeable and prevents antibiotics from passing passively into the intracellular compartments. Biodynamers, a novel class of dynamically bio-responsive polymers, may open new perspectives to overcome this particular barrier by accommodating various secondary structures and form supramolecular structures in such bacterial microenvironments. Generally, bio-responsive polymers are not only candidates as bio-active molecules against bacteria but also carriers via their interactions with the cargo. Based on their dynamicity, design flexibility, biodegradability, biocompatibility, and pH-responsiveness, we investigated the potential of two peptide-based biodynamers for improving antimicrobial drug delivery. By a range of experimental methods, we discovered a greater affinity of Arg-biodynamers for bacterial membranes than for mammalian membranes as well as an enhanced LPS targeting on the bacterial membrane, opening perspectives for enhancing the delivery of antimicrobials across the Gram-negative bacterial cell envelope. This could be explained by the change of the secondary structure of Arg-biodynamers into a predominant ß-sheet character in the LPS microenvironment, by contrast to the α-helical structure typically observed for most lipid membrane-permeabilizing peptides. In comparison to poly-L-arginine, the intrinsic antibacterial activity of Arg-biodynamers was nearly unchanged, but its toxicity against mammalian cells was >128-fold reduced. When used in bacterio as an antibiotic potentiator, however, Arg-biodynamers improved the minimum inhibitory concentration (MIC) against Escherichia coli by 32 times compared to colistin alone. Similar effect has also been observed in two stains of Pseudomonas aeruginosa. Arg-biodynamers may therefore represent an interesting option as an adjuvant for antibiotics against Gram-negative bacteria and to overcome antimicrobial resistance.


Subject(s)
Anti-Bacterial Agents , Gram-Negative Bacteria , Lipopolysaccharides , Microbial Sensitivity Tests , Lipopolysaccharides/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Gram-Negative Bacteria/drug effects , Bacterial Outer Membrane/drug effects , Bacterial Outer Membrane/metabolism , Humans , Escherichia coli/drug effects , Polymers/chemistry , Arginine/chemistry , Drug Delivery Systems/methods
16.
J Biosci Bioeng ; 138(2): 137-143, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38796341

ABSTRACT

Shewanella vesiculosa HM13, a psychrotrophic gram-negative bacterium isolated from the intestinal contents of horse mackerel, produces abundant extracellular membrane vesicles (EMVs) by budding the outer membrane. The EMVs of this bacterium carry a single major cargo protein, P49, of unknown function, which may be useful as a carrier for the secretory production of heterologous proteins as cargoes of EMVs. In this study, to increase the utility of S. vesiculosa HM13 as a host for EMV-mediated protein production, we improved its EMV productivity by weakening the linkage between the outer membrane and underlying peptidoglycan layer. In gram-negative bacteria, the outer membrane is connected to peptidoglycans predominantly through Braun's lipoprotein (Lpp), and the formation of this linkage is catalyzed by an l,d-transpeptidase (Ldt). We constructed gene-disrupted mutants of Lpp and Ldt and assessed their EMV productivity. The EMVs of the lpp- and ldt-disrupted mutants grown at 18 °C were evaluated using nanoparticle tracking analysis, and their morphologies were observed using transmission electron microscopy. As a result, an approximately 2.5-fold increase in EMV production was achieved, whereas the morphology of the EMVs of these mutants remained almost identical to that of the parent strain. In accordance with the increase in EMV production, the mutants secreted approximately 2-fold higher amounts of P49 than the parent strain into the culture broth as the EMV cargo. These findings will contribute to the development of an EMV-based secretory production system for heterologous proteins using S. vesiculosa HM13 as a host.


Subject(s)
Extracellular Vesicles , Peptidoglycan , Shewanella , Shewanella/metabolism , Shewanella/genetics , Extracellular Vesicles/metabolism , Peptidoglycan/metabolism , Bacterial Outer Membrane/metabolism , Protein Transport , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Lipoproteins/metabolism , Lipoproteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/genetics
17.
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
18.
J Nanobiotechnology ; 22(1): 273, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773507

ABSTRACT

BACKGROUND: The outer membrane vesicles (OMVs) produced by Gram-negative bacteria can modulate the immune system and have great potentials for bacterial vaccine development. RESULTS: A highly active Acinetobacter baumannii phage lysin, LysP53, can stimulate the production of OMVs after interacting with A. baumannii, Escherichia coli, and Salmonella. The OMVs prepared by the lysin (LOMVs) from A. baumannii showed better homogeneity, higher protein yield, lower endotoxin content, and lower cytotoxicity compared to the naturally produced OMVs (nOMVs). The LOMVs contain a significantly higher number of cytoplasmic and cytoplasmic membrane proteins but a smaller number of periplasmic and extracellular proteins compared to nOMVs. Intramuscular immunization with either LOMVs or nOMVs three times provided robust protection against A. baumannii infections in both pneumonia and bacteremia mouse models. Intranasal immunization offered good protection in the pneumonia model but weaker protection (20-40%) in the bacteremia model. However, with a single immunization, LOMVs demonstrated better protection than the nOMVs in the pneumonia mouse model. CONCLUSIONS: The novel lysin approach provides a superior choice compared to current methods for OMV production, especially for vaccine development.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Bacteriophages , Animals , Acinetobacter Infections/prevention & control , Mice , Female , Mice, Inbred BALB C , Bacterial Vaccines/immunology , Immunization , Extracellular Vesicles , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane Proteins/immunology , Disease Models, Animal , Humans , Administration, Intranasal , Viral Proteins
19.
J Extracell Vesicles ; 13(5): e12447, 2024 May.
Article in English | MEDLINE | ID: mdl-38766978

ABSTRACT

The continuous emergence of multidrug-resistant bacterial pathogens poses a major global healthcare challenge, with Klebsiella pneumoniae being a prominent threat. We conducted a comprehensive study on K. pneumoniae's antibiotic resistance mechanisms, focusing on outer membrane vesicles (OMVs) and polymyxin, a last-resort antibiotic. Our research demonstrates that OMVs protect bacteria from polymyxins. OMVs derived from Polymyxin B (PB)-stressed K. pneumoniae exhibited heightened protective efficacy due to increased vesiculation, compared to OMVs from unstressed Klebsiella. OMVs also shield bacteria from different bacterial families. This was validated ex vivo and in vivo using precision cut lung slices (PCLS) and Galleria mellonella. In all models, OMVs protected K. pneumoniae from PB and reduced the associated stress response on protein level. We observed significant changes in the lipid composition of OMVs upon PB treatment, affecting their binding capacity to PB. The altered binding capacity of single OMVs from PB stressed K. pneumoniae could be linked to a reduction in the lipid A amount of their released vesicles. Although the amount of lipid A per vesicle is reduced, the overall increase in the number of vesicles results in an increased protection because the sum of lipid A and therefore PB binding sites have increased. This unravels the mechanism of the altered PB protective efficacy of OMVs from PB stressed K. pneumoniae compared to control OMVs. The lipid A-dependent protective effect against PB was confirmed in vitro using artificial vesicles. Moreover, artificial vesicles successfully protected Klebsiella from PB ex vivo and in vivo. The findings indicate that OMVs act as protective shields for bacteria by binding to polymyxins, effectively serving as decoys and preventing antibiotic interaction with the cell surface. Our findings provide valuable insights into the mechanisms underlying antibiotic cross-protection and offer potential avenues for the development of novel therapeutic interventions to address the escalating threat of multidrug-resistant bacterial infections.


Subject(s)
Anti-Bacterial Agents , Klebsiella pneumoniae , Polymyxin B , Klebsiella pneumoniae/metabolism , Klebsiella pneumoniae/drug effects , Anti-Bacterial Agents/pharmacology , Animals , Polymyxin B/pharmacology , Bacterial Outer Membrane/metabolism , Polymyxins/pharmacology , Extracellular Vesicles/metabolism , Klebsiella Infections/microbiology , Klebsiella Infections/metabolism , Microbial Sensitivity Tests , Drug Resistance, Multiple, Bacterial/drug effects
20.
BMC Microbiol ; 24(1): 186, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38802775

ABSTRACT

The outer membrane (OM) of Gram-negative bacteria acts as an effective barrier to protect against toxic compounds. By nature, the OM is asymmetric with the highly packed lipopolysaccharide (LPS) at the outer leaflet and glycerophospholipids at the inner leaflet. OM asymmetry is maintained by the Mla system, in which is responsible for the retrograde transport of glycerophospholipids from the OM to the inner membrane. This system is comprised of six Mla proteins, including MlaA, an OM lipoprotein involved in the removal of glycerophospholipids that are mis-localized at the outer leaflet of the OM. Interestingly, MlaA was initially identified - and called VacJ - based on its role in the intracellular spreading of Shigella flexneri.Many open questions remain with respect to the Mla system and the mechanism involved in the translocation of mislocated glycerophospholipids at the outer leaflet of the OM, by MlaA. After summarizing the current knowledge on MlaA, we focus on the impact of mlaA deletion on OM lipid composition and biophysical properties of the OM. How changes in OM lipid composition and biophysical properties can impact the generation of membrane vesicles and membrane permeability is discussed. Finally, we explore whether and how MlaA might be a candidate for improving the activity of antibiotics and as a vaccine candidate.Efforts dedicated to understanding the relationship between the OM lipid composition and the mechanical strength of the bacterial envelope and, in turn, how such properties act against external stress, are needed for the design of new targets or drugs for Gram-negative infections.


Subject(s)
Bacterial Outer Membrane Proteins , Bacterial Outer Membrane , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/genetics , Membrane Lipids/metabolism , Gram-Negative Bacteria/metabolism , Glycerophospholipids/metabolism , Shigella flexneri/metabolism , Shigella flexneri/physiology , Shigella flexneri/genetics
SELECTION OF CITATIONS
SEARCH DETAIL