Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.488
Filter
1.
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
2.
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
3.
Curr Opin Microbiol ; 79: 102479, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718542

ABSTRACT

Bacteria surround themselves with complex cell envelopes to maintain their integrity and protect against external insults. The envelope of Gram-negative organisms is multilayered, with two membranes sandwiching the periplasmic space that contains the peptidoglycan cell wall. Understanding how this complicated surface architecture is assembled during cell growth and division is a major fundamental problem in microbiology. Additionally, because the envelope is an important antibiotic target and determinant of intrinsic antibiotic resistance, understanding the mechanisms governing its assembly is relevant to therapeutic development. In the last several decades, most of the factors required to build the Gram-negative envelope have been identified. However, surprisingly, little is known about how the biogenesis of the different cell surface layers is co-ordinated. Here, we provide an overview of recent work that is beginning to uncover the links connecting the different envelope biosynthetic pathways and assembly machines to ensure uniform envelope growth.


Subject(s)
Cell Membrane , Cell Wall , Gram-Negative Bacteria , Peptidoglycan , Gram-Negative Bacteria/metabolism , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/physiology , Cell Wall/metabolism , Peptidoglycan/metabolism , Cell Membrane/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics
4.
J Bacteriol ; 206(6): e0002724, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38814789

ABSTRACT

Iron acquisition systems are crucial for pathogen growth and survival in iron-limiting host environments. To overcome nutritional immunity, bacterial pathogens evolved to use diverse mechanisms to acquire iron. Here, we examine a heme acquisition system that utilizes hemophores called hemophilins which are also referred to as HphAs in several Gram-negative bacteria. In this study, we report three new HphA structures from Stenotrophomonas maltophilia, Vibrio harveyi, and Haemophilus parainfluenzae. Structural determination of HphAs revealed an N-terminal clamp-like domain that binds heme and a C-terminal eight-stranded ß-barrel domain that shares the same architecture as the Slam-dependent Neisserial surface lipoproteins. The genetic organization of HphAs consists of genes encoding a Slam homolog and a TonB-dependent receptor (TBDR). We investigated the Slam-HphA system in the native organism or the reconstituted system in Escherichia coli cells and found that the efficient secretion of HphA depends on Slam. The TBDR also played an important role in heme uptake and conferred specificity for its cognate HphA. Furthermore, bioinformatic analysis of HphA homologs revealed that HphAs are conserved in the alpha, beta, and gammaproteobacteria. Together, these results show that the Slam-dependent HphA-type hemophores are prevalent in Gram-negative bacteria and further expand the role of Slams in transporting soluble proteins. IMPORTANCE: This paper describes the structure and function of a family of Slam (Type IX secretion System) secreted hemophores that bacteria use to uptake heme (iron) while establishing an infection. Using structure-based bioinformatics analysis to define the diversity and prevalence of this heme acquisition pathway, we discovered that a large portion of gammaproteobacterial harbors this system. As organisms, including Acinetobacter baumannii, utilize this system to facilitate survival during host invasion, the identification of this heme acquisition system in bacteria species is valuable information and may represent a target for antimicrobials.


Subject(s)
Bacterial Proteins , Gram-Negative Bacteria , Heme , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/metabolism , Heme/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Iron/metabolism
5.
Curr Opin Struct Biol ; 87: 102828, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38723580

ABSTRACT

Recent advances in molecular dynamics (MD) simulations have led to rapid improvement in our understanding of the molecular details of the outer membranes (OMs) of Gram-negative bacteria. In this review, we highlight the latest discoveries from MD simulations of OMs, shedding light on the dynamic nature of these bacteria's first line of defense. With the focus on cutting-edge approaches, we explore the OM's sensitivity to structural features, including divalent cations and membrane composition, which have emerged as crucial determinants of antimicrobial passage. Additionally, studies have provided novel insights into outer-membrane proteins (OMPs), revealing their intricate roles in substrate translocation and their distinct interactions with lipopolysaccharides (LPS) in the OM. Finally, we explore the challenging process of ß-barrel membrane protein insertion, showcasing recent findings that have enhanced our grasp of this fundamental biological phenomenon.


Subject(s)
Bacterial Outer Membrane Proteins , Gram-Negative Bacteria , Molecular Dynamics Simulation , Gram-Negative Bacteria/metabolism , Gram-Negative Bacteria/chemistry , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane/chemistry , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism
6.
Proc Natl Acad Sci U S A ; 121(17): e2322363121, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38640341

ABSTRACT

Anti-microbial resistance (AMR) is one of the greatest threats to global health. The continual battle between the emergence of AMR and the development of drugs will be extremely difficult to stop as long as traditional anti-biotic approaches are taken. In order to overcome this impasse, we here focused on the type III secretion system (T3SS), which is highly conserved in many Gram-negative pathogenic bacteria. The T3SS is known to be indispensable in establishing disease processes but not essential for pathogen survival. Therefore, T3SS inhibitors may be innovative anti-infective agents that could dramatically reduce the evolutionary selective pressure on strains resistant to treatment. Based on this concept, we previously identified a polyketide natural product, aurodox (AD), as a specific T3SS inhibitor using our original screening system. However, despite its promise as a unique anti-infective drug of AD, the molecular target of AD has remained unclear. In this paper, using an innovative chemistry and genetic biology-based approach, we show that AD binds to adenylosuccinate synthase (PurA), which suppresses the production of the secreted proteins from T3SS, resulting in the expression of bacterial virulence both in vitro and in vivo experiments. Our findings illuminate the potential of PurA as a target of anti-infective drugs and vaccination and could open a avenue for application of PurA in the regulation of T3SS.


Subject(s)
Aurodox , Type III Secretion Systems , Type III Secretion Systems/metabolism , Aurodox/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Gram-Negative Bacteria/metabolism , Bacterial Proteins/metabolism
7.
Commun Biol ; 7(1): 499, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664513

ABSTRACT

Bacterial cooperation and antagonism mediated by secretion systems are among the ways in which bacteria interact with one another. Here we report the discovery of an antagonistic property of a type IV secretion system (T4SS) sourced from a conjugative plasmid, RP4, using engineering approaches. We scrutinized the genetic determinants and suggested that this antagonistic activity is independent of molecular cargos, while we also elucidated the resistance genes. We further showed that a range of Gram-negative bacteria and a mixed bacterial population can be eliminated by this T4SS-dependent antagonism. Finally, we showed that such an antagonistic property is not limited to T4SS sourced from RP4, rather it can also be observed in a T4SS originated from another conjugative plasmid, namely R388. Our results are the first demonstration of conjugative T4SS-dependent antagonism between Gram-negative bacteria on the genetic level and provide the foundation for future mechanistic studies.


Subject(s)
Conjugation, Genetic , Plasmids , Type IV Secretion Systems , Plasmids/genetics , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism
8.
Proc Natl Acad Sci U S A ; 121(15): e2317274121, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38579010

ABSTRACT

Here, we describe the identification of an antibiotic class acting via LpxH, a clinically unexploited target in lipopolysaccharide synthesis. The lipopolysaccharide synthesis pathway is essential in most Gram-negative bacteria and there is no analogous pathway in humans. Based on a series of phenotypic screens, we identified a hit targeting this pathway that had activity on efflux-defective strains of Escherichia coli. We recognized common structural elements between this hit and a previously published inhibitor, also with activity against efflux-deficient bacteria. With the help of X-ray structures, this information was used to design inhibitors with activity on efflux-proficient, wild-type strains. Optimization of properties such as solubility, metabolic stability and serum protein binding resulted in compounds having potent in vivo efficacy against bloodstream infections caused by the critical Gram-negative pathogens E. coli and Klebsiella pneumoniae. Other favorable properties of the series include a lack of pre-existing resistance in clinical isolates, and no loss of activity against strains expressing extended-spectrum-ß-lactamase, metallo-ß-lactamase, or carbapenemase-resistance genes. Further development of this class of antibiotics could make an important contribution to the ongoing struggle against antibiotic resistance.


Subject(s)
Anti-Bacterial Agents , Lipopolysaccharides , Humans , Anti-Bacterial Agents/chemistry , Escherichia coli/metabolism , Gram-Negative Bacteria/metabolism , beta-Lactamases/genetics , Microbial Sensitivity Tests
9.
Curr Opin Microbiol ; 79: 102465, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38520915

ABSTRACT

Contractile injection systems (CISs) are phage tail-like machineries found in a wide range of bacteria. They are often deployed by bacteria to translocate effectors into the extracellular space or into target cells. CISs are classified into intracellular type VI secretion systems (T6SSs) and extracellular CIS (eCISs). eCISs are assembled in cytoplasm and released into the extracellular milieu upon cell lysis, while T6SSs are the secretion systems widespread among Gram-negative bacteria and actively translocate effectors into the environment or into the adjacent cell, without lysis of T6SS-producing cells. Recently, several noncanonical CISs that exhibit distinct characteristics have been discovered. This review will provide an overview on these noncanonical CISs and their unique features, as well as new advances in reprogramming CISs for therapeutic protein delivery.


Subject(s)
Bacterial Proteins , Type VI Secretion Systems , Type VI Secretion Systems/metabolism , Type VI Secretion Systems/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacteria/metabolism , Bacteria/genetics , Gram-Negative Bacteria/metabolism , Gram-Negative Bacteria/physiology , Gram-Negative Bacteria/genetics
10.
J Cell Mol Med ; 28(6): e18146, 2024 03.
Article in English | MEDLINE | ID: mdl-38426932

ABSTRACT

Acne vulgaris represents a chronic inflammatory condition, the pathogenesis of which is closely associated with the altered skin microbiome. Recent studies have implicated a profound role of Gram-negative bacteria in acne development, but there is a lack of antiacne agents targeting these bacteria. Polyphyllins are major components of Rhizoma Paridis with great anti-inflammatory potential. In this study, we aimed to evaluate the antiacne effects and the underlying mechanisms of PPH and a PPH-enriched Rhizoma Paridis extract (RPE) in treating the Gram-negative bacteria-induced acne. PPH and RPE treatments significantly suppressed the mRNA and protein expressions of interleukin (IL)-1ß and IL-6 in lipopolysaccharide (LPS)-induced RAW 264.7 and HaCaT cells, along with the intracellular reactive oxygen species (ROS) generation. Furthermore, PPH and RPE inhibited the nuclear translocation of nuclear factor kappa-B (NF-κB) P65 in LPS-induced RAW 264.7 cells. Based on molecular docking, PPH could bind to kelch-like ECH-associated protein 1 (KEAP1) protein. PPH and RPE treatments could activate nuclear factor erythroid 2-related factor 2 (NRF2) and upregulate haem oxygenase-1 (HO-1). Moreover, RPE suppressed the mitogen-activated protein kinase (MAPK) pathway. Therefore, PPH-enriched RPE showed anti-inflammatory and antioxidative effects in vitro, which is promising for alternative antiacne therapeutic.


Subject(s)
Acne Vulgaris , Saponins , Humans , Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Lipopolysaccharides/adverse effects , Saponins/pharmacology , Saponins/therapeutic use , Molecular Docking Simulation , Anti-Inflammatory Agents/therapeutic use , NF-kappa B/metabolism , Gram-Negative Bacteria/metabolism , Acne Vulgaris/drug therapy , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Inflammation/metabolism
11.
Int J Biol Macromol ; 264(Pt 2): 130631, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38453114

ABSTRACT

Gram-negative bacteria binding proteins (GNBPs) have the ability to recognize molecular patterns associated with microbial pathogens (PAMPs), leading to the activation of immune responses downstream. In the genome of Tribolium castaneum, three GNBP genes have been identified; however, their immunological roles remain unexplored. In our study, a GNBP1, designated as TcGNBP1, were identified from the cDNA library of T. castaneum. The coding sequence of TcGNBP1 consisted of 1137 bps and resulted in the synthesis of a protein comprising 378 amino acids. This protein encompasses a signal peptide, a low-complexity region, and a glycoside hydrolase 16 domain. TcGNBP1 was strongly expressed in early adult stages, and mainly distributed in hemolymph and gut. Upon being challenged with Escherichia coli or Staphylococcus aureus, the transcript levels of TcGNBP1 were significantly changed at different time points. Through molecular docking and ELISA analysis, it was observed that TcGNBP1 has the ability to interact with lipopolysaccharides, peptidoglycan, and ß-1, 3-glucan. Based on these findings, it was further discovered that recombinant TcGNBP1 can directly bind to five different bacteria in a Ca2+-dependent manner. After knockdown of TcGNBP1 with RNA interference, expression of antimicrobial peptide genes and prophenoloxidase (proPO) activity were suppressed, the susceptibility of T. castaneum to E. coli or S. aureus infection was enhanced, leading to low survival rate. These results suggest a regulatory mechanism of TcGNBP1 in innate immunity of T. castaneum and provide a potential molecular target for dsRNA-based insect pest management.


Subject(s)
Tribolium , Animals , Tribolium/genetics , Tribolium/metabolism , Carrier Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Staphylococcus aureus/metabolism , Molecular Docking Simulation , Bacteria/metabolism , Gram-Negative Bacteria/metabolism , Immunity, Innate/genetics , Insect Proteins/genetics , Insect Proteins/metabolism
12.
Methods Mol Biol ; 2778: 1-30, 2024.
Article in English | MEDLINE | ID: mdl-38478268

ABSTRACT

ß-barrels are a class of membrane proteins made up of a cylindrical, anti-parallel ß-sheet with a hydrophobic exterior and a hydrophilic interior. The majority of proteins found in the outer membranes (OMs) of Gram-negative bacteria, mitochondria, and chloroplasts are ß-barrel outer membrane proteins (OMPs). ß-barrel OMPs have a diverse repertoire of functions, including nutrient transport, secretion, bacterial virulence, and enzymatic activity. Here, we discuss the broad functional classes of ß-barrel OMPs, how they are folded into the membrane, and the future of ß-barrel OMP research and its applications.


Subject(s)
Bacterial Outer Membrane Proteins , Escherichia coli Proteins , Bacterial Outer Membrane Proteins/metabolism , Escherichia coli Proteins/metabolism , Mitochondria/metabolism , Gram-Negative Bacteria/metabolism , Protein Conformation, beta-Strand , Protein Folding
13.
Methods Mol Biol ; 2778: 83-99, 2024.
Article in English | MEDLINE | ID: mdl-38478273

ABSTRACT

ß-barrel membrane proteins populate the outer membrane of Gram-negative bacteria, mitochondria, and chloroplasts, playing significant roles in multiple key cellular pathways. Characterizing the functions of these membrane proteins in vivo is often challenging due to the complex protein network in the periplasm of Gram-negative bacteria (or intermembrane space in mitochondria and chloroplasts) and the presence of other outer membrane proteins. In vitro reconstitution into lipid-bilayer-like environments such as nanodiscs or proteoliposomes provides an excellent method for examining the specific function and mechanism of these membrane proteins in an isolated system. Here, we describe the methodologies employed to investigate Slam, a 14-stranded ß-barrel membrane protein also known as the type XI secretion system that is responsible for translocating proteins across the outer membrane of many bacterial species.


Subject(s)
Bacterial Outer Membrane Proteins , Proteolipids , Bacterial Outer Membrane Proteins/metabolism , Proteolipids/metabolism , Mitochondria/metabolism , Protein Transport , Gram-Negative Bacteria/metabolism
14.
Methods Mol Biol ; 2778: 367-381, 2024.
Article in English | MEDLINE | ID: mdl-38478289

ABSTRACT

Biogenesis of the outer membrane (OM) of Gram-negative bacteria involves two processes essential for growth, that is, the insertion of ß-barrel outer membrane proteins (OMPs) by the Bam complex and the assembly of the LPS-containing outer leaflet of the OM by the LptD/E complex from the Lpt pathway. These processes have only recently gained attention as targets for antimicrobial drugs. Our laboratory has developed a simple screening tool to identify compounds that target processes that disrupt the biogenesis of the cell envelope, among which the activity of the Bam complex. The tool is based on the observation that such a disruption triggers cell envelope stress response systems, such as the σE, Rcs, and Cpx responses. In essence, specific stress-responsive promoters are fused to a gene encoding a bright fluorescent protein to serve as a panel of easy-to-monitor stress reporter plasmids. Using these plasmids, compounds triggering these stress systems and, therefore, putatively disrupting the biogenesis of the cell envelope can be identified by the nature and kinetics of the induced stress responses. We describe here the use of the stress reporter plasmids in high-throughput phenotypic screening using multi-well plates.


Subject(s)
Escherichia coli Proteins , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Cell Membrane/metabolism , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/metabolism
15.
J Am Chem Soc ; 146(13): 8981-8990, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38513269

ABSTRACT

The rapid development of antibiotic resistance, especially among difficult-to-treat Gram-negative bacteria, is recognized as a serious and urgent threat to public health. The detection and characterization of novel resistance mechanisms are essential to better predict the spread and evolution of antibiotic resistance. Corramycin is a novel and modified peptidic antibiotic with activity against several Gram-negative pathogens. We demonstrate that the kinase ComG, part of the corramycin biosynthetic gene cluster, phosphorylates and thereby inactivates corramycin, leading to the resistance of the host. Remarkably, we found that the closest structural homologues of ComG are aminoglycoside phosphotransferases; however, ComG shows no activity toward this class of antibiotics. The crystal structure of ComG in complex with corramycin reveals that corramycin adopts a ß-hairpin-like structure and allowed us to define the changes leading to a switch in substrate from sugar to peptide. Bioinformatic analyses suggest a limited occurrence of ComG-like proteins, which along with the absence of cross-resistance to clinically used drugs positions corramycin as an attractive antibiotic for further development.


Subject(s)
Anti-Bacterial Agents , Gram-Negative Bacteria , Anti-Bacterial Agents/chemistry , Gram-Negative Bacteria/metabolism , Kanamycin Kinase/chemistry , Kanamycin Kinase/genetics , Kanamycin Kinase/metabolism , Peptides
16.
Methods Appl Fluoresc ; 12(3)2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38537290

ABSTRACT

Polycationic photosensitizers (PS) are not susceptible to aggregation in solutions, but their high local concentrations in Gram-negative bacteria can be sufficient for aggregation and reduced effectiveness of antibacterial photodynamic treatment. By measuring fluorescence spectra and kinetics we were able to evaluate the degree of aggregation of polycationic PS ZnPcChol8in Gram-negative bacteria E.coliK12 TG1. Binding of ZnPcChol8toE.coliK12 TG1 leads to an appearance of groups of molecules with shorter PS fluorescence lifetime, a decrease in fluorescence intensity and a shift in the fluorescence spectral maximum. However, we evaluated that about 88% of the fluorescing PS molecules in the bacteria were in an unaggregated state, which indicates only a small reduction in the generation of reactive oxygen species.


Subject(s)
Photochemotherapy , Photosensitizing Agents , Polyelectrolytes , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Gram-Negative Bacteria/metabolism , Reactive Oxygen Species/metabolism
17.
Nat Commun ; 15(1): 2432, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38503735

ABSTRACT

Arrest peptides containing RAPP (ArgAlaProPro) motifs have been discovered in both Gram-positive and Gram-negative bacteria, where they are thought to regulate expression of important protein localization machinery components. Here we determine cryo-EM structures of ribosomes stalled on RAPP arrest motifs in both Bacillus subtilis and Escherichia coli. Together with molecular dynamics simulations, our structures reveal that the RAPP motifs allow full accommodation of the A-site tRNA, but prevent the subsequent peptide bond from forming. Our data support a model where the RAP in the P-site interacts and stabilizes a single hydrogen atom on the Pro-tRNA in the A-site, thereby preventing an optimal geometry for the nucleophilic attack required for peptide bond formation to occur. This mechanism to short circuit the ribosomal peptidyltransferase activity is likely to operate for the majority of other RAPP-like arrest peptides found across diverse bacterial phylogenies.


Subject(s)
Peptidyl Transferases , Peptidyl Transferases/metabolism , Anti-Bacterial Agents/metabolism , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/genetics , Protein Biosynthesis , Ribosomes/metabolism , Peptides/metabolism , RNA, Transfer/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism
18.
Nat Commun ; 15(1): 2007, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38453922

ABSTRACT

Monoclonal IgG antibodies constitute the fastest growing class of therapeutics. Thus, there is an intense interest to design more potent antibody formats, where long plasma half-life is a commercially competitive differentiator affecting dosing, frequency of administration and thereby potentially patient compliance. Here, we report on an Fc-engineered variant with three amino acid substitutions Q311R/M428E/N434W (REW), that enhances plasma half-life and mucosal distribution, as well as allows for needle-free delivery across respiratory epithelial barriers in human FcRn transgenic mice. In addition, the Fc-engineered variant improves on-target complement-mediated killing of cancer cells as well as both gram-positive and gram-negative bacteria. Hence, this versatile Fc technology should be broadly applicable in antibody design aiming for long-acting prophylactic or therapeutic interventions.


Subject(s)
Neoplasms , Receptors, Fc , Mice , Animals , Humans , Immunoglobulin G , Half-Life , Anti-Bacterial Agents/therapeutic use , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/metabolism , Mice, Transgenic , Antibodies, Monoclonal , Histocompatibility Antigens Class I/metabolism , Neoplasms/therapy , Neoplasms/drug therapy
19.
Curr Opin Microbiol ; 78: 102433, 2024 04.
Article in English | MEDLINE | ID: mdl-38350268

ABSTRACT

Our ability to control the growth of Gram-negative bacterial pathogens is challenged by rising antimicrobial resistance and requires new approaches. Endolysins are phage-derived enzymes that degrade peptidoglycan and therefore offer potential as antimicrobial agents. However, the outer membrane (OM) of Gram-negative bacteria impedes the access of externally applied endolysins to peptidoglycan. This review highlights recent advances in the discovery and characterization of natural endolysins that can breach the OM, as well as chemical and engineering approaches that increase antimicrobial efficacy of endolysins against Gram-negative pathogens.


Subject(s)
Anti-Infective Agents , Bacteriophages , Anti-Bacterial Agents/chemistry , Peptidoglycan/metabolism , Endopeptidases/genetics , Endopeptidases/pharmacology , Endopeptidases/chemistry , Anti-Infective Agents/metabolism , Gram-Negative Bacteria/metabolism , Bacteriophages/metabolism
20.
ACS Infect Dis ; 10(3): 845-857, 2024 03 08.
Article in English | MEDLINE | ID: mdl-38363869

ABSTRACT

Myeloid differentiation factor 2 (MD2), the TLR4 coreceptor, has been shown to possess opsonic activity and has been implicated in phagocytosis and intracellular killing of Gram-negative bacteria. However, any MD2 protein segment involved in phagocytosis of Gram-negative bacteria is not yet known. A short synthetic MD2 segment, MD54 (amino acid regions 54 to 69), was shown to interact with a Gram-negative bacterial outer membrane component, LPS, earlier. Furthermore, the MD54 peptide induced aggregation of LPS and facilitated its internalization in THP-1 cells. Currently, it has been investigated if MD2-derived MD54 possesses any opsonic property and role in phagocytosis of Gram-negative bacteria. Remarkably, we observed that MD54 facilitated agglutination of Gram-negative bacteria, Escherichia coli (ATCC 25922) and Pseudomonas aeruginosa (ATCC BAA-427), but not of Gram-positive bacteria, Bacillus subtilis (ATCC 6633) and Staphylococcus aureus (ATCC 25923). The MD54-opsonized Gram-negative bacteria internalized within PMA-treated THP-1 cells and were killed over a longer incubation period. However, both internalization and intracellular killing of the MD54-opsonized Gram-negative bacteria within THP-1 phagocytes were appreciably inhibited in the presence of a phagocytosis inhibitor, cytochalasin D. Furthermore, MD54 facilitated the clearance of Gram-negative bacteria E. coli (ATCC 25922) and P. aeruginosa (ATCC BAA-427) from the infected BALB/c mice whereas an MD54 analog, MMD54, was inactive. Overall, for the first time, the results revealed that a short MD2-derived peptide can specifically agglutinate Gram-negative bacteria, act as an opsonin for these bacteria, and facilitate their phagocytosis by THP-1 phagocytes. The results suggest that the MD54 segment could have a crucial role in MD2-mediated host-pathogen interaction involving the Gram-negative bacteria.


Subject(s)
Escherichia coli , Lipopolysaccharides , Animals , Mice , Lipopolysaccharides/metabolism , Escherichia coli/metabolism , Phagocytosis , Macrophages/metabolism , Gram-Negative Bacteria/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL