Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.523
Filtrar
1.
Appl Environ Microbiol ; 90(8): e0004424, 2024 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-39007603

RESUMEN

The Soudan Underground Mine State Park, found in the Vermilion Iron Range in northern Minnesota, provides access to a ~ 2.7 billion-year-old banded iron formation. Exploratory boreholes drilled between 1958 and 1962 on the 27th level (713 m underground) of the mine intersect calcium and iron-rich brines that have recently been subject to metagenomic analysis and microbial enrichments. Using concentrated brine samples pumped from a borehole depth of up to 55 m, a novel Gram-positive bacterium was enriched under anaerobic, acetate-oxidizing, and Fe(III) citrate-reducing conditions. The isolated bacterium, designated strain MK1, is non-motile, rod-shaped, spore-forming, anaerobic, and mesophilic, with a growth range between 24°C and 30°C. The complete circular MK1 genome was found to be 3,720,236 bp and encodes 25 putative multiheme cytochromes, including homologs to inner membrane cytochromes in the Gram-negative bacterium Geobacter sulfurreducens and cytoplasmic membrane and periplasmic cytochromes in the Gram-positive bacterium Thermincola potens. However, MK1 does not encode homologs of the peptidoglycan (CwcA) and cell surface-associated (OcwA) multiheme cytochromes proposed to be required by T. potens to perform extracellular electron transfer. The 16S rRNA gene sequence of MK1 indicates that its closest related isolate is Desulfitibacter alkalitolerans strain sk.kt5 (91% sequence identity), which places MK1 in a novel genus within the Desulfitibacteraceae family and Moorellales order. Within the Moorellales order, only Calderihabitans maritimus strain KKC1 has been reported to reduce Fe(III), and only D. alkalitolerans can also grow in temperatures below 40°C. Thus, MK1 represents a novel species within a novel genus, for which we propose the name "Metallumcola ferriviriculae" strain MK1, and provides a unique opportunity to study a cytochrome-rich, mesophilic, Gram-positive, spore-forming Fe(III)-reducing bacterium.IMPORTANCEThe Soudan Underground Mine State Park gives access to understudied regions of the deep terrestrial subsurface that potentially predate the Great Oxidation Event. Studying organisms that have been relatively unperturbed by surface conditions for as long as 2.7 billion years may give us a window into ancient life before oxygen dominated the planet. Additionally, studying microbes from anoxic and iron-rich environments can help us better understand the requirements of life in analogous environments, such as on Mars. The isolation and characterization of "Metallumcola ferriviriculae" strain MK1 give us insights into a novel genus and species that is distinct both from its closest related isolates and from iron reducers characterized to date. "M. ferriviriculae" strain MK1 may also act as a model organism to study how the processes of sporulation and germination are affected by insoluble extracellular acceptors, as well as the impact of spores in the deep terrestrial biosphere.


Asunto(s)
Genoma Bacteriano , Oxidación-Reducción , Filogenia , Minería , Hierro/metabolismo , ARN Ribosómico 16S/genética , Compuestos Férricos/metabolismo , Minnesota , Bacterias Grampositivas/genética , Bacterias Grampositivas/clasificación , Bacterias Grampositivas/metabolismo , Bacterias Grampositivas/aislamiento & purificación
2.
Bull Exp Biol Med ; 177(1): 140-146, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38960962

RESUMEN

The dynamics of lung microbiota in tuberculosis remains poorly understood. Sequencing of variable regions of the 16S rRNA gene from surgically excised tuberculosis foci and biopsy specimens of normal lung tissue allowed characterization of the diversity and predictive potential of bacterial communities. Taxonomic diversity indices attested to differences in the structure of microbial communities between "healthy" lungs and tuberculomas. The microbial composition of "healthy" lungs varied in taxonomic diversity and was presented by both gram-positive and gram-negative bacteria with sufficiently similar metabolic potential. The microbiota of the examined tuberculomas consisted of Mycobacterium tuberculosis in 99.9% of cases. A significant part of the metabolic pathways predicted by PICRUSt2 included cholesterol catabolism, sulfate assimilation, and various pathways for the biosynthesis of cell wall components.


Asunto(s)
Pulmón , Mycobacterium tuberculosis , ARN Ribosómico 16S , Tuberculoma , Humanos , ARN Ribosómico 16S/genética , Mycobacterium tuberculosis/genética , Tuberculoma/microbiología , Tuberculoma/patología , Tuberculoma/genética , Pulmón/microbiología , Pulmón/patología , Pulmón/metabolismo , Microbiota/genética , Microbiota/fisiología , Masculino , Adulto , Tuberculosis Pulmonar/microbiología , Femenino , Persona de Mediana Edad , Bacterias Gramnegativas/genética , Bacterias Grampositivas/genética , Bacterias Grampositivas/metabolismo , Bacterias Grampositivas/clasificación
3.
Curr Biol ; 34(13): 2932-2947.e7, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38897200

RESUMEN

Many bacteria glycosylate flagellin on serine or threonine residues using pseudaminic acid (Pse) or other sialic acid-like donor sugars. Successful reconstitution of Pse-dependent sialylation by the conserved Maf-type flagellin glycosyltransferase (fGT) may require (a) missing component(s). Here, we characterize both Maf paralogs in the Gram-negative bacterium Shewanella oneidensis MR-1 and reconstitute Pse-dependent glycosylation in heterologous hosts. Remarkably, we uncovered distinct acceptor determinants and target specificities for each Maf. Whereas Maf-1 uses its C-terminal tetratricopeptide repeat (TPR) domain to confer flagellin acceptor and O-glycosylation specificity, Maf-2 requires the newly identified conserved specificity factor, glycosylation factor for Maf (GlfM), to form a ternary complex with flagellin. GlfM orthologs are co-encoded with Maf-2 in Gram-negative and Gram-positive bacteria and require an invariant aspartate in their four-helix bundle to function with Maf-2. Thus, convergent fGT evolution underlies distinct flagellin-binding modes in tripartite versus bipartite systems and, consequently, distinct O-glycosylation preferences of acceptor serine residues with Pse.


Asunto(s)
Flagelina , Flagelina/metabolismo , Flagelina/genética , Glicosilación , Shewanella/metabolismo , Shewanella/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Glicosiltransferasas/metabolismo , Glicosiltransferasas/genética , Bacterias Grampositivas/metabolismo , Bacterias Grampositivas/genética , Evolución Molecular
4.
J Colloid Interface Sci ; 664: 275-283, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38471190

RESUMEN

Planktonic bacterial presence in many industrial and environmental applications and personal health-care products is generally countered using antimicrobials. However, antimicrobial chemicals present an environmental threat, while emerging resistance reduces their efficacy. Suspended bacteria have no defense against mechanical attack. Therefore, we synthesized silica hexapods on an α-Fe2O3 core that can be magnetically-rotated to inflict lethal cell-wall-damage to planktonic Gram-negative and Gram-positive bacteria. Hexapods possessed 600 nm long nano-spikes, composed of SiO2, as shown by FTIR and XPS. Fluorescence staining revealed cell wall damage caused by rotating hexapods. This damage was accompanied by DNA/protein release and bacterial death that increased with increasing rotational frequency up to 500 rpm. Lethal puncturing was more extensive on Gram-negative bacteria than on Gram-positive bacteria, which have a thicker peptidoglycan layer with a higher Young's modulus. Simulations confirmed that cell-wall-puncturing occurs at lower nano-spike penetration levels in the cell walls of Gram-negative bacteria. This approach offers a new way to kill bacteria in suspension, not based on antimicrobial chemicals.


Asunto(s)
Antiinfecciosos , Bacterias Gramnegativas , Antibacterianos/farmacología , Antibacterianos/metabolismo , Dióxido de Silicio/farmacología , Dióxido de Silicio/metabolismo , Bacterias Grampositivas/metabolismo , Plancton , Bacterias , Pared Celular
5.
Chem Commun (Camb) ; 60(27): 3697-3700, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38477080

RESUMEN

We introduce a biotinylated D-amino acid probe capable of metabolically incorporating into bacterial PG. Leveraging the robust affinity between biotin and streptavidin, the probe has demonstrated efficacy in imaging, capture, and targeted inactivation of Gram-positive bacteria through synergistic pairings with commercially available streptavidin-modified fluorescent dyes and nanomaterials. The versatility of the probe is underscored by its compatibility with a variety of commercially available streptavidin-modified reagents. This adaptability allows the probe to be applied across diverse scenarios by integrating with these commercial reagents.


Asunto(s)
Bacterias , Biotina , Estreptavidina/química , Biotina/química , Bacterias/metabolismo , Colorantes Fluorescentes/química , Bacterias Grampositivas/metabolismo
6.
Nat Commun ; 15(1): 2007, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38453922

RESUMEN

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.


Asunto(s)
Neoplasias , Receptores Fc , Ratones , Animales , Humanos , Inmunoglobulina G , Semivida , Antibacterianos/uso terapéutico , Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/metabolismo , Ratones Transgénicos , Anticuerpos Monoclonales , Antígenos de Histocompatibilidad Clase I/metabolismo , Neoplasias/terapia , Neoplasias/tratamiento farmacológico
7.
Eur J Med Chem ; 268: 116303, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38458107

RESUMEN

Methionyl-tRNA synthetase (MetRS) catalyzes the attachment of l-methionine (l-Met) to tRNAMet to generate methionyl-tRNAMet, an essential substrate for protein translation within ribosome. Owing to its indispensable biological function and the structural discrepancies with human counterpart, bacterial MetRS is considered an ideal target for developing antibacterials. Herein, chlorhexidine (CHX) was identified as a potent binder of Staphylococcus aureus MetRS (SaMetRS) through an ATP-aided affinity screening. The co-crystal structure showed that CHX simultaneously occupies the enlarged l-Met pocket (EMP) and the auxiliary pocket (AP) of SaMetRS with its two chlorophenyl groups, while its central hexyl linker swings upwards to interact with some conserved hydrophobic residues. ATP adopts alternative conformations in the active site cavity, and forms ionic bonds and water-mediated hydrogen bonds with CHX. Consistent with this synergistic binding mode, ATP concentration-dependently enhanced the binding affinity of CHX to SaMetRS from 10.2 µM (no ATP) to 0.45 µM (1 mM ATP). While it selectively inhibited two representative type 1 MetRSs from S. aureus and Enterococcus faecalis, CHX did not show significant interactions with three tested type 2 MetRSs, including human cytoplasmic MetRS, in the enzyme inhibition and biophysical binding assays, probably due to the conformational differences between two types of MetRSs at their EMP and AP. Our findings on CHX may inspire the design of MetRS-directed antimicrobials in future.


Asunto(s)
Metionina-ARNt Ligasa , Humanos , Metionina-ARNt Ligasa/química , Metionina-ARNt Ligasa/genética , Metionina-ARNt Ligasa/metabolismo , Clorhexidina/farmacología , Staphylococcus aureus , ARN de Transferencia de Metionina/metabolismo , Bacterias Grampositivas/metabolismo , Adenosina Trifosfato/metabolismo
8.
Int J Biol Macromol ; 260(Pt 1): 129493, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38224804

RESUMEN

Endolysins are lytic enzymes produced by bacteriophages at the end of their lytic cycle and degrade the peptidoglycan layer of the bacterial cell wall. Thus, they have been extensively explored as a promising antibacterial agent to replace or supplement current antibiotics. Gram-negative bacteria, however, are prone to resist exogenous endolysins owing to their protective outer membrane. We previously engineered endolysin EC340, encoded by the Escherichia coli phage PBEC131, by substituting its seven amino acids and fusing an antimicrobial peptide cecropin A at its N-terminus. The engineered endolysin LNT113 exerted superior activity to its intrinsic form. This study investigated how cecropin A fusion facilitated the bactericidal activity of LNT113 toward Gram-negative bacteria. Cecropin A of LNT113 markedly increased the interaction with lipopolysaccharides, while the E. coli defective in the core oligosaccharide was less susceptible to endolysins, implicating the interaction between the core oligosaccharide and endolysins. In fact, E. coli with compromised lipid A construction was more vulnerable to LNT113 treatment, suggesting that the integrity of the lipid A layer was important to resist the internalization of LNT113 across the outer membrane. Cecropin A fusion further accelerated the inner membrane destabilization, thereby enabling LNT113 to deconstruct it promptly. Owing to the increased membrane permeability, LNT113 could inactivate some Gram-positive bacteria as well. This study demonstrates that cecropin A fusion is a feasible method to improve the membrane permeability of endolysins in both Gram-negative and Gram-positive bacteria.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Escherichia coli , Lípido A , Escherichia coli/metabolismo , Endopeptidasas/química , Bacterias Gramnegativas/metabolismo , Antibacterianos/farmacología , Antibacterianos/química , Bacterias Grampositivas/metabolismo , Oligosacáridos
9.
Glycobiology ; 34(2)2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-37847605

RESUMEN

Bacteria possess diverse metabolic and genetic processes, resulting in the inability of certain bacteria to degrade trehalose. However, some bacteria do have the capability to degrade trehalose, utilizing it as a carbon source, and for defense against environmental stress. Trehalose, a disaccharide, serves as a carbon source for many bacteria, including some that are vital for pathogens. The degradation of trehalose is carried out by enzymes like trehalase (EC 3.2.1.28) and trehalose phosphorylase (EC 2.4.1.64/2.4.1.231), which are classified under the glycoside hydrolase families GH37, GH15, and GH65. Numerous studies and reports have explored the physiological functions, recombinant expression, enzymatic characteristics, and potential applications of these enzymes. However, further research is still being conducted to understand their roles in bacteria. This review aims to provide a comprehensive summary of the current understanding of trehalose degradation pathways in various bacteria, focusing on three key areas: (i) identifying different trehalose-degrading enzymes in Gram-positive and Gram-negative bacteria, (ii) elucidating the mechanisms employed by trehalose-degrading enzymes belonging to the glycoside hydrolases GH37, GH15, and GH65, and (iii) discussing the potential applications of these enzymes in different sectors. Notably, this review emphasizes the bacterial trehalose-degrading enzymes, specifically trehalases (GH37, GH15, and GH65) and trehalose phosphorylases (GH65), in both Gram-positive and Gram-negative bacteria, an aspect that has not been highlighted before.


Asunto(s)
Glucosiltransferasas , Trehalasa , Trehalosa , Humanos , Trehalosa/metabolismo , Trehalasa/genética , Trehalasa/metabolismo , Antibacterianos , Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/metabolismo , Bacterias/metabolismo , Carbono
10.
J Bacteriol ; 206(1): e0024123, 2024 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-37975670

RESUMEN

Serine-rich-repeat proteins (SRRPs) are large mucin-like glycoprotein adhesins expressed by a plethora of pathogenic and symbiotic Gram-positive bacteria. SRRPs play major functional roles in bacterial-host interactions, like adhesion, aggregation, biofilm formation, virulence, and pathogenesis. Through their functional roles, SRRPs aid in the development of host microbiomes but also diseases like infective endocarditis, otitis media, meningitis, and pneumonia. SRRPs comprise shared domains across different species, including two or more heavily O-glycosylated long stretches of serine-rich repeat regions. With loci that can be as large as ~40 kb and can encode up to 10 distinct glycosyltransferases that specifically facilitate SRRP glycosylation, the SRRP loci makes up a significant portion of the bacterial genome. The significance of SRRPs and their glycans in host-microbe communications is becoming increasingly evident. Studies are beginning to reveal the glycosylation pathways and mature O-glycans presented by SRRPs. Here we review the glycosylation machinery of SRRPs across species and discuss the functional roles and clinical manifestations of SRRP glycosylation.


Asunto(s)
Adhesinas Bacterianas , Serina , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Serina/metabolismo , Glicosilación , Bacterias Grampositivas/metabolismo , Polisacáridos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Adhesión Bacteriana
11.
Microbiol Spectr ; 12(1): e0203023, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38032193

RESUMEN

IMPORTANCE: Tigecycline, a glycecycline antibiotic with broad-spectrum activity against almost all Gram-positive and Gram-negative bacteria, is a highly concerned "last-resort" antibiotic. In addition to plasmid-hosted mobile tet(X) conferring high-level resistance to tigecycline, there are many reports suggesting increased expression of AcrAB-TolC efflux pump leads to tigecycline non-susceptibility. However, the role of mutations in AcrAB-TolC on tigecycline resistance has not been identified. This study reports a novel T188A mutation of the AcrA subunit of AcrAB-TolC complex in a clinical tigecycline-resistant Klebsiella pneumoniae strain and reveals the role of AcrA mutation on tigecycline resistance in K. pneumoniae. High prevalence of A188 type AcrA in hypervirulent multidrug-resistant K. pneumoniae indicates that mutations of the AcrAB-TolC complex may play a larger role in determining bacterial pathogenesis and antibiotic susceptibility than previously expected.


Asunto(s)
Antibacterianos , Infecciones por Klebsiella , Humanos , Tigeciclina/farmacología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/metabolismo , Minociclina/farmacología , Aminoácidos , Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/metabolismo , Mutación , Pruebas de Sensibilidad Microbiana , Infecciones por Klebsiella/microbiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana/genética
12.
Front Cell Infect Microbiol ; 13: 1282258, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37942477

RESUMEN

Hfq is required by many Gram-negative bacteria to chaperone the interaction between small non-coding RNA (sRNA) and mRNA to facilitate annealing. Conversely and despite the presence of Hfq in many Gram-positive bacteria, sRNAs in Gram-positive bacteria bind the mRNA target independent of Hfq. Details provided by the Hfq structures from both Gram-negative and Gram-positive bacteria have demonstrated that despite a conserved global structure of the protein, variations of residues on the binding surfaces of Hfq results in the recognition of different RNA sequences as well as the ability of Hfq to facilitate the annealing of the sRNA to the mRNA target. Additionally, a subset of Gram-negative bacteria has an extended C-terminal Domain (CTD) that has been shown to affect the stability of the Hfq hexamer and increase the rate of release of the annealed sRNA-mRNA product. Here we review the structures of Hfq and biochemical data that have defined the interactions of the Gram-negative and Gram-positive homologues to highlight the similarities and differences in the interactions with RNA. These interactions provided a deeper understanding of the how Hfq functions to facilitate the annealing of sRNA-mRNA, the selectivity of the interactions with RNA, and the role of the CTD of Hfq in the interactions with sRNA.


Asunto(s)
Proteínas de Escherichia coli , ARN Pequeño no Traducido , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , Bacterias Grampositivas/genética , Bacterias Grampositivas/metabolismo , Secuencia de Bases , Bacterias Gramnegativas/genética , Bacterias Gramnegativas/metabolismo , ARN Mensajero/metabolismo , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo , Proteína de Factor 1 del Huésped/genética , Proteínas de Escherichia coli/genética
13.
J Bacteriol ; 205(11): e0031023, 2023 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-37905811

RESUMEN

IMPORTANCE: With the lack of new antibiotics in the drug discovery pipeline, coupled with accelerated evolution of antibiotic resistance, new sources of antibiotics that target pathogens of clinical importance are paramount. Here, we use bacterial cytological profiling to identify the mechanism of action of the monounsaturated fatty acid (Z)-13-methyltetra-4-decenoic acid isolated from the marine bacterium Olleya marilimosa with antibacterial effects against Gram-positive bacteria. The fatty acid antibiotic was found to rapidly destabilize the cell membrane by pore formation and membrane aggregation in Bacillus subtilis, suggesting that this fatty acid may be a promising adjuvant used in combination to enhance antibiotic sensitivity.


Asunto(s)
Antibacterianos , Ácidos Grasos , Ácidos Grasos/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo , Bacterias Grampositivas/metabolismo , Membrana Celular/metabolismo , Bacillus subtilis/metabolismo , Pruebas de Sensibilidad Microbiana , Bacterias Gramnegativas/metabolismo
14.
Int J Mol Sci ; 24(18)2023 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-37762406

RESUMEN

The current study describes the encapsulation of hydroxychloroquine, widely used in traditional medicine due to its diverse pharmacological and medicinal uses, in chitosan nanoparticles (CNPs). This work aims to combine the HCQ drug with CS NPs to generate a novel nanocomposite with improved characteristics and bioavailability. HCQ@CS NPs are roughly shaped like roadways and have a smooth surface with an average size of 159.3 ± 7.1 nm, a PDI of 0.224 ± 0.101, and a zeta potential of +46.6 ± 0.8 mV. To aid in the development of pharmaceutical systems for use in cancer therapy, the binding mechanism and affinity of the interaction between HCQ and HCQ@CS NPs and BSA were examined using stopped-flow and other spectroscopic approaches, supplemented by molecular docking analysis. HCQ and HCQ@CS NPs binding with BSA is driven by a ground-state complex formation that may be accompanied by a non-radiative energy transfer process, and binding constants indicate that HCQ@CS NPs-BSA was more stable than HCQ-BSA. The stopped-flow analysis demonstrated that, in addition to increasing BSA affinity, the nanoformulation HCQ@CS NPS changes the binding process and may open new routes for interaction. Docking experiments verified the development of the HCQ-BSA complex, with HCQ binding to site I on the BSA structure, primarily with the amino acids, Thr 578, Gln 579, Gln 525, Tyr 400, and Asn 404. Furthermore, the nanoformulation HCQ@CS NPS not only increased cytotoxicity against the A549 lung cancer cell line (IC50 = 28.57 ± 1.72 µg/mL) compared to HCQ (102.21 ± 0.67 µg/mL), but also exhibited higher antibacterial activity against both Gram-positive and Gram-negative bacteria when compared to HCQ and chloramphenicol, which is in agreement with the binding constants. The nanoformulation developed in this study may offer a viable therapy option for A549 lung cancer.


Asunto(s)
Quitosano , Neoplasias Pulmonares , Nanopartículas , Humanos , Simulación del Acoplamiento Molecular , Quitosano/química , Hidroxicloroquina/farmacología , Liberación de Fármacos , Antibacterianos , Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/metabolismo , Nanopartículas/química , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo
15.
Int J Biol Macromol ; 253(Pt 5): 126825, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37696369

RESUMEN

Teichoic acid (TA) is a weakly anionic polymer present in the cell walls of Gram-positive bacteria. It can be classified into wall teichoic acid (WTA) and lipoteichoic acid (LTA) based on its localization in the cell wall. The structure and biosynthetic pathway of TAs are strain-specific and have a significant role in maintaining cell wall stability. TAs have various beneficial functions, such as immunomodulatory, anticancer and antioxidant activities. However, the purity and yield of TAs are generally not high, and different isolation methods may even affect their structural integrity, which limits the research progress on the probiotic functions of TA. This paper reviews an overview of the structure and biosynthetic pathway of TAs in different strains, as well as the research progress of the isolation and purification methods of TAs. Furthermore, this review also highlights the current research status on the biological functions of TAs. Through a comprehensive understanding of this review, it is expected to pave the way for advancements in isolating and purifying high-quality TAs and, in turn, lay a foundation for contributing to the development of targeted probiotic therapies.


Asunto(s)
Pared Celular , Bacterias Grampositivas , Pared Celular/química , Bacterias Grampositivas/metabolismo , Glicosilación , Ácidos Teicoicos/química , Lipopolisacáridos/química , Vías Biosintéticas , Polímeros/metabolismo
16.
Elife ; 122023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37750680

RESUMEN

Generating specific, robust protective responses to different bacteria is vital for animal survival. Here, we address the role of transforming growth factor ß (TGF-ß) member DBL-1 in regulating signature host defense responses in Caenorhabditis elegans to human opportunistic Gram-negative and Gram-positive pathogens. Canonical DBL-1 signaling is required to suppress avoidance behavior in response to Gram-negative, but not Gram-positive bacteria. We propose that in the absence of DBL-1, animals perceive some bacteria as more harmful. Animals activate DBL-1 pathway activity in response to Gram-negative bacteria and strongly repress it in response to select Gram-positive bacteria, demonstrating bacteria-responsive regulation of DBL-1 signaling. DBL-1 signaling differentially regulates expression of target innate immunity genes depending on the bacterial exposure. These findings highlight a central role for TGF-ß in tailoring a suite of bacteria-specific host defenses.


Asunto(s)
Proteínas de Caenorhabditis elegans , Neuropéptidos , Animales , Humanos , Caenorhabditis elegans/fisiología , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Neuropéptidos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal , Bacterias Grampositivas/metabolismo
17.
PLoS Biol ; 21(8): e3002186, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37561817

RESUMEN

Antibiotic resistance is a continuously increasing concern for public healthcare. Understanding resistance mechanisms and their emergence is crucial for the development of new antibiotics and their effective use. The peptide antibiotic albicidin is such a promising candidate that, as a gyrase poison, shows bactericidal activity against a wide range of gram-positive and gram-negative bacteria. Here, we report the discovery of a gene amplification-based mechanism that imparts an up to 1000-fold increase in resistance levels against albicidin. RNA sequencing and proteomics data show that this novel mechanism protects Salmonella Typhimurium and Escherichia coli by increasing the copy number of STM3175 (YgiV), a transcription regulator with a GyrI-like small molecule binding domain that traps albicidin with high affinity. X-ray crystallography and molecular docking reveal a new conserved motif in the binding groove of the GyrI-like domain that can interact with aromatic building blocks of albicidin. Phylogenetic studies suggest that this resistance mechanism is ubiquitous in gram-negative bacteria, and our experiments confirm that STM3175 homologs can confer resistance in pathogens such as Vibrio vulnificus and Pseudomonas aeruginosa.


Asunto(s)
Antibacterianos , Amplificación de Genes , Antibacterianos/farmacología , Simulación del Acoplamiento Molecular , Filogenia , Bacterias Gramnegativas/genética , Bacterias Grampositivas/metabolismo
18.
Int J Antimicrob Agents ; 62(4): 106941, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37536571

RESUMEN

Wall teichoic acid (WTA) and lipoteichoic acid (LTA) are structural components of Gram-positive bacteria's peptidoglycan and cell membrane, which are mostly anionic glycopolymers. WTA confers numerous physiological, virulence, and pathogenic features to bacterial pathogens. It controls cell shape, cell division, and the localisation of autolytic enzymes and ion homeostasis. In the context of virulence and pathogenicity, it aids bacterial cell attachment and colonisation and protects against the host defence system and antibiotics. Having such a broad function in pathogenic bacteria's lifecycle, WTA/LTA become one of the potential targets for antibacterial agents to reduce bacterial infection in the host. The number of reports for targeting the WTA/LTA pathway has risen, mostly by focusing on three distinct targets: antivirulence targets, ß-lactam potentiator targets, and essential targets. The current review looked at the role of WTA/LTA in biofilm development and virulence in a range of Gram-positive pathogenic bacteria. Furthermore, alternate strategies, such as the application of natural and synthetic compounds that target the WTA/LTA pathway, have been thoroughly discussed. Moreover, the application of nanomaterials and a combination of drugs have also been discussed as a viable method for targeting the WTA/LTA in numerous Gram-positive bacteria. In addition, a future perspective for controlling bacterial infection by targeting the WTA/LTA is proposed.


Asunto(s)
Infecciones Bacterianas , Lipopolisacáridos , Humanos , Virulencia , Lipopolisacáridos/metabolismo , Ácidos Teicoicos/metabolismo , Pared Celular/metabolismo , Antibacterianos/metabolismo , Biopelículas , Bacterias Grampositivas/metabolismo
19.
Exp Eye Res ; 234: 109601, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37488008

RESUMEN

Bandage contact lenses have an increased affinity to accumulate tear film proteins and bacteria during wear. Among the wide variety of tear film proteins, lysozyme has attracted the most attention for several reasons, including the fact that it is found at a high concentration in the tear film, has exceptional antibacterial and antibiofilm properties, and its significant deposits onto contact lenses. This study aims to evaluate the effect of lysozyme on bacterial biofilm formation on bandage contact lenses. For this purpose, several methods, including microtiter plate test and Colony Forming Unit (CFU) assay have been used to determine antibacterial and antibiofilm characteristics of lysozyme against the two most frequent contact lens-induced bacterial ocular infections, Staphylococcus aureus, and Pseudomonas aeruginosa. The results of these assays demonstrate lysozyme potential to inhibit 57.9% and 80.7% of the growth of S. aureus and P. aeruginosa, respectively. In addition, biofilm formations of P. aeruginosa and S. aureus reduced by 38.3% and 62.7%, respectively due to the antibiofilm effect of lysozyme. SEM and AFM imaging were utilized to visualize lysozyme antibacterial activity and topography changes of the contact lens surface, respectively, in the presence/absence of lysozyme. The results indicated that lysozyme can efficiently attack both gram-positive and gram-negative bacteria and consequently lysozyme-functionalized bandage contact lenses can reduce the risk of ocular infection after eye surgery.


Asunto(s)
Lentes de Contacto Hidrofílicos , Muramidasa , Muramidasa/farmacología , Adhesión Bacteriana , Antibacterianos/farmacología , Staphylococcus aureus , Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/metabolismo , Pseudomonas aeruginosa , Biopelículas , Vendajes
20.
Microb Pathog ; 181: 106182, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37263448

RESUMEN

Efflux proteins are transporter molecules that actively pump out a variety of substrates, including antibiotics, from cells to the environment. They are found in both Gram-positive and Gram-negative bacteria and eukaryotic cells. Based on their protein sequence homology, energy source, and overall structure, efflux proteins can be divided into seven groups. Multidrug efflux pumps are transmembrane proteins produced by microbes to enhance their survival in harsh environments and contribute to antibiotic resistance. These pumps are present in all bacterial genomes studied, indicating their ancestral origins. Many bacterial genes encoding efflux pumps are involved in transport, a significant contributor to antibiotic resistance in microbes. Efflux pumps are widely implicated in the extrusion of clinically relevant antibiotics from cells to the extracellular environment and, as such, represent a significant challenge to antimicrobial therapy. This review aims to provide an overview of the structures and mechanisms of action, substrate profiles, regulation, and possible inhibition of clinically relevant efflux pumps. Additionally, recent advances in research and the pharmacological exploitation of efflux pump inhibitors as a promising intervention for combating drug resistance will be discussed.


Asunto(s)
Proteínas Bacterianas , Bacterias Gramnegativas , Proteínas Bacterianas/metabolismo , Bacterias Gramnegativas/genética , Bacterias Gramnegativas/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo , Farmacorresistencia Bacteriana Múltiple/genética , Bacterias Grampositivas/genética , Bacterias Grampositivas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA