Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 652
Filtrar
1.
J Chem Theory Comput ; 20(4): 1704-1716, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-37676287

RESUMO

The outer lipopolysaccharide (LPS) membrane of Gram-negative bacteria forms the main barrier for transport of antimicrobial molecules into the bacterial cell. In this study we develop coarse-grained models for the outer membrane of Escherichia coli in the Martini-3 framework. The coarse-grained model force field was parametrized and validated using all-atom simulations of symmetric membranes of lipid A and rough LPS as well as a complete asymmetric membrane of LPS with the O-antigen. The bonded parameters were obtained using an iterative refinement procedure with target bonded distributions obtained from all-atom simulations. The membrane thickness, area of the LPS, and density distributions for the different regions as well as the water and ion densities in Martini-3 simulations show excellent agreement with the all-atom data. Additionally the solvent accessible surface area for individual molecules in water was found to be in good agreement. The binding of calcium ions with phosphate and carboxylate moieties of LPS is accurately captured in the Martini-3 model, indicative of the integrity of the highly negatively charged LPS molecules in the outer membranes of Gram-negative bacteria. The melting transition of the coarse-grained lipid A membrane model was found to occur between 300 and 310 K, and the model captured variations in area per LPS, order parameter, and membrane thickness across the melting transition. Our study reveals that the proposed Martini-3 models for LPS are able to capture the physicochemical balance of the complex sugar architecture of the outer membrane of Escherichia coli. The coarse-grained models developed in this study would be useful for determining membrane protein interactions and permeation of potential antimicrobials through bacterial membranes at mesoscopic spatial and temporal scales.


Assuntos
Lipídeo A , Lipopolissacarídeos , Lipopolissacarídeos/química , Escherichia coli , Simulação de Dinâmica Molecular , Bactérias Gram-Negativas/química , Água
2.
Nature ; 626(7999): 617-625, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38081298

RESUMO

The outer membrane in Gram-negative bacteria consists of an asymmetric phospholipid-lipopolysaccharide bilayer that is densely packed with outer-membrane ß-barrel proteins (OMPs) and lipoproteins1. The architecture and composition of this bilayer is closely monitored and is essential to cell integrity and survival2-4. Here we find that SlyB, a lipoprotein in the PhoPQ stress regulon, forms stable stress-induced complexes with the outer-membrane proteome. SlyB comprises a 10 kDa periplasmic ß-sandwich domain and a glycine zipper domain that forms a transmembrane α-helical hairpin with discrete phospholipid- and lipopolysaccharide-binding sites. After loss in lipid asymmetry, SlyB oligomerizes into ring-shaped transmembrane complexes that encapsulate ß-barrel proteins into lipid nanodomains of variable size. We find that the formation of SlyB nanodomains is essential during lipopolysaccharide destabilization by antimicrobial peptides or acute cation shortage, conditions that result in a loss of OMPs and compromised outer-membrane barrier function in the absence of a functional SlyB. Our data reveal that SlyB is a compartmentalizing transmembrane guard protein that is involved in cell-envelope proteostasis and integrity, and suggest that SlyB represents a larger family of broadly conserved lipoproteins with 2TM glycine zipper domains with the ability to form lipid nanodomains.


Assuntos
Proteínas da Membrana Bacteriana Externa , Membrana Celular , Bactérias Gram-Negativas , Bicamadas Lipídicas , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Glicina/metabolismo , Lipopolissacarídeos/metabolismo , Lipoproteínas/química , Lipoproteínas/metabolismo , Fosfolipídeos/metabolismo , Sítios de Ligação , Proteostase , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Proteoma/química , Proteoma/metabolismo , Regulon , Domínios Proteicos , Peptídeos Antimicrobianos/metabolismo , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/citologia , Bactérias Gram-Negativas/metabolismo
3.
Nature ; 623(7988): 814-819, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37938784

RESUMO

Gram-negative bacteria are surrounded by two membranes. A special feature of the outer membrane is its asymmetry. It contains lipopolysaccharide (LPS) in the outer leaflet and phospholipids in the inner leaflet1-3. The proper assembly of LPS in the outer membrane is required for cell viability and provides Gram-negative bacteria intrinsic resistance to many classes of antibiotics. LPS biosynthesis is completed in the inner membrane, so the LPS must be extracted, moved across the aqueous periplasm that separates the two membranes and translocated through the outer membrane where it assembles on the cell surface4. LPS transport and assembly requires seven conserved and essential LPS transport components5 (LptA-G). This system has been proposed to form a continuous protein bridge that provides a path for LPS to reach the cell surface6,7, but this model has not been validated in living cells. Here, using single-molecule tracking, we show that Lpt protein dynamics are consistent with the bridge model. Half of the inner membrane Lpt proteins exist in a bridge state, and bridges persist for 5-10 s, showing that their organization is highly dynamic. LPS facilitates Lpt bridge formation, suggesting a mechanism by which the production of LPS can be directly coupled to its transport. Finally, the bridge decay kinetics suggest that there may be two different types of bridges, whose stability differs according to the presence (long-lived) or absence (short-lived) of LPS. Together, our data support a model in which LPS is both a substrate and a structural component of dynamic Lpt bridges that promote outer membrane assembly.


Assuntos
Membrana Externa Bacteriana , Proteínas de Transporte , Bactérias Gram-Negativas , Lipopolissacarídeos , Proteínas de Membrana , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/metabolismo , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Transporte Biológico , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Escherichia coli/química , Escherichia coli/citologia , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/citologia , Bactérias Gram-Negativas/metabolismo , Lipopolissacarídeos/química , Lipopolissacarídeos/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo
4.
Nature ; 613(7945): 721-728, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36450355

RESUMO

The microbial cell wall is essential for maintenance of cell shape and resistance to external stressors1. The primary structural component of the cell wall is peptidoglycan, a glycopolymer with peptide crosslinks located outside of the cell membrane1. Peptidoglycan biosynthesis and structure are responsive to shifting environmental conditions such as pH and salinity2-6, but the mechanisms underlying such adaptations are incompletely understood. Precursors of peptidoglycan and other cell surface glycopolymers are synthesized in the cytoplasm and then delivered across the cell membrane bound to the recyclable lipid carrier undecaprenyl phosphate7 (C55-P, also known as UndP). Here we identify the DUF368-containing and DedA transmembrane protein families as candidate C55-P translocases, filling a critical gap in knowledge of the proteins required for the biogenesis of microbial cell surface polymers. Gram-negative and Gram-positive bacteria lacking their cognate DUF368-containing protein exhibited alkaline-dependent cell wall and viability defects, along with increased cell surface C55-P levels. pH-dependent synthetic genetic interactions between DUF368-containing proteins and DedA family members suggest that C55-P transporter usage is dynamic and modulated by environmental inputs. C55-P transporter activity was required by the cholera pathogen for growth and cell shape maintenance in the intestine. We propose that conditional transporter reliance provides resilience in lipid carrier recycling, bolstering microbial fitness both inside and outside the host.


Assuntos
Proteínas de Bactérias , Proteínas de Transporte , Aptidão Genética , Bactérias Gram-Negativas , Bactérias Gram-Positivas , Fosfatos de Poli-Isoprenil , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Parede Celular/química , Parede Celular/metabolismo , Lipídeos/análise , Peptidoglicano/metabolismo , Fosfatos de Poli-Isoprenil/metabolismo , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/citologia , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Positivas/química , Bactérias Gram-Positivas/citologia , Bactérias Gram-Positivas/metabolismo , Viabilidade Microbiana
5.
J Chem Phys ; 157(15): 154902, 2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36272789

RESUMO

Lipopolysaccharides (LPSs) are negatively charged molecules covering the surface of Gram-negative bacteria (GNB). Adding divalent cations (DCs) is important to stabilize the LPS bilayer. Thus, DCs are always only considered as membrane stabilizing ions. Here, on the basis of a coarse-grained (CG) Martini force field, we conduct molecular dynamic (MD) simulations to study the divalent cation mediated LPS interaction and the stability of the LPS membrane in a wide range of DC concentrations. By measuring the LPS binding free energy and the LPS-LPS aggregate from the association course between two LPS molecules, we find that the initial addition of DCs may significantly facilitate the aggregation of LPSs into a compact structure, while sequentially adding more DCs only unpacks the LPS aggregate and drives the dissolution of LPSs. With an increasing concentration of DCs, we find a gradual replacement of DCs to monovalent cations as condensed counterions on the LPS, which follows a sign change from negative to positive in terms of the LPS effective charge and a switch of LPSs in solution from undergoing precipitation to resolubilization on adding DCs. This interaction change in the level of two LPSs accounts for the structure variation of the LPS assembly on a larger scale, where the LPS packing rigidity in the assembly bilayer is found with a similar nonmonotonic dependence with the DC concentration. Thus, our results demonstrate for the first time the presence of a re-entrant condensation behavior for LPS molecules, which can be exploited for developing novel membrane-perturbing agents based on multivalent ions as efficient GNB antibiotics.


Assuntos
Bactérias Gram-Negativas , Lipopolissacarídeos , Cátions Bivalentes/química , Lipopolissacarídeos/química , Bactérias Gram-Negativas/química , Cátions Monovalentes , Antibacterianos
6.
Biophys J ; 121(18): 3486-3498, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-35964158

RESUMO

Polymyxins are increasingly used as the last-line therapeutic option for the treatment of infections caused by multidrug-resistant Gram-negative bacteria. However, efforts to address the resistance in superbugs are compromised by a poor understanding of the bactericidal modes because high-resolution detection of the cell structure is still lacking. By performing molecular dynamics simulations at a coarse-grained level, here we show that polymyxin B (PmB) disrupts Gram-negative bacterial membranes by altering lipid homeostasis and asymmetry. We found that the binding of PmBs onto the asymmetric outer membrane (OM) loosens the packing of lipopolysaccharides (LPS) and induces unbalanced bending torque between the inner and outer leaflets, which in turn triggers phospholipids to flip from the inner leaflet to the outer leaflet to compensate for the stress deformation. Meanwhile, some LPSs may be detained on the inner membrane (IM). Then, the lipid-scrambled OM undergoes phase separation. Defects are created at the boundaries between LPS-rich domains and phospholipid-rich domains, which consequently facilitate the uptake of PmB across the OM. Finally, PmBs target LPSs detained on the IM and similarly perturb the IM. This lipid Scramble, membrane phase Separation, and peptide Translocation model depicts a novel mechanism by which polymyxins kill bacteria and sheds light on developing a new generation of polymyxins or antibiotic adjuvants with improved killing activities and higher therapeutic indices.


Assuntos
Lipopolissacarídeos , Polimixinas , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Membrana Celular/metabolismo , Bactérias Gram-Negativas/química , Homeostase , Lipopolissacarídeos/química , Lipopolissacarídeos/farmacologia , Fosfolipídeos/química , Polimixina B/farmacologia , Polimixinas/análise , Polimixinas/metabolismo , Polimixinas/farmacologia
7.
PLoS One ; 17(2): e0263593, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35202421

RESUMO

Magnetotactic bacteria (MTB) rely on magnetotaxis to effectively reach their preferred living habitats, whereas experimental investigation of magnetotactic advantage in stable sediment is currently lacking. We studied two wild type MTB (cocci and rod-shaped M. bavaricum) in sedimentary environment under exposure to geomagnetic field in the laboratory, zero field and an alternating field whose polarity was switched every 24 hours. The mean concentration of M. bavaricum dropped by ~50% during 6 months in zero field, with no clear temporal trend suggesting an extinction. Cell numbers recovered to initial values within ~1.5 months after the Earth's field was reset. Cocci displayed a larger temporal variability with no evident population changes in zero field. The alternating field experiment produced a moderate decrease of M. bavaricum concentrations and nearby extinction of cocci, confirming the active role of magnetotaxis in sediment and might point to a different magnetotactic mechanism for M. bavaricum which possibly benefited them to survive field reversals in geological periods. Our findings provide a first quantification of magnetotaxis advantage in sedimentary environment.


Assuntos
Fenômenos Fisiológicos Bacterianos , Bactérias Gram-Negativas/fisiologia , Magnetismo , Magnetossomos/química , Planeta Terra , Ecossistema , Sedimentos Geológicos , Bactérias Gram-Negativas/química , Magnetossomos/fisiologia , Filogenia , RNA Ribossômico 16S
8.
Carbohydr Polym ; 278: 118995, 2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-34973797

RESUMO

Driven by its excellent physical and chemical properties, BC (bacterial cellulose) has achieved significant progress in the last decade, rendering with many novel applications. Due to its resemblance to the structure of extracellular matrix, BC-based biomaterials have been widely explored for biomedical applications such as tissue engineering and drug delivery. The recent advances in nanotechnology endow further modifications on BC and generate BC-based composites for different applications. This article presents a review on the research advancement on BC-based biomaterials from fabrication methods to biomedical applications, including wound dressing, artificial skin, vascular tissue engineering, bone tissue regeneration, drug delivery, and other applications. The preparation of these materials and their potential applications are reviewed and summarized. Important factors for the applications of BC in biomedical applications including degradation and pore structure characteristic are discussed in detail. Finally, the challenges in future development and potential advances of these materials are also discussed.


Assuntos
Materiais Biocompatíveis/química , Celulose/química , Sistemas de Liberação de Medicamentos , Bactérias Gram-Negativas/química , Engenharia Tecidual , Configuração de Carboidratos
9.
J Biochem ; 170(6): 775-785, 2022 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-34557892

RESUMO

In this study, we investigated the activation of TRPV1 and TRPA1 by N-acyl homoserine lactones, quorum sensing molecules produced by Gram-negative bacteria, and the inhibitory effect of TRPV1 and TRPA1 by autoinducing peptides (AIPs), quorum sensing molecules produced by Gram-positive bacteria, using human embryonic kidney 293T cell lines stably expressing human TRPV1 and TRPA1, respectively. As a result, we found that some N-acyl homoserine lactones, such as N-octanoyl-L-homoserine lactone (C8-HSL), N-nonanoyl-L-homoserine lactone (C9-HSL) and N-decanoyl-L-homoserine lactone (C10-HSL), activated both TRPV1 and TRPA1. In addition, we clarified that some N-acyl homoserine lactones, such as N-3-oxo-dodecanoyl-L-homoserine lactone (3-oxo-C12-HSL), only activated TRPV1 and N-acyl homoserine lactones having saturated short acyl chain, such as N-acetyl-L-homoserine lactone (C2-HSL) and N-butyryl-L-homoserine lactone (C4-HSL), only activated TRPA1. Furthermore, we found that an AIP, simple linear peptide CHWPR, inhibited both TRPV1 and TRPA1 and peptide having thiolactone ring DICNAYF, the thiolactone ring were formed between C3 to F7, strongly inhibited only the TRPV1. Although the specificity of TRPV1 and TRPA1 for quorum sensing molecules was different, these data suggest that both TRPV1 and TRPA1 would function as receptors for quorum sensing molecule produced by bacteria. Graphical Abstract.


Assuntos
Acil-Butirolactonas/farmacologia , Bactérias Gram-Negativas/química , Percepção de Quorum , Canal de Cátion TRPA1/metabolismo , Canais de Cátion TRPV/metabolismo , Acil-Butirolactonas/química , Bactérias Gram-Negativas/genética , Bactérias Gram-Negativas/metabolismo , Células HEK293 , Humanos , Canal de Cátion TRPA1/química , Canal de Cátion TRPA1/genética , Canais de Cátion TRPV/química , Canais de Cátion TRPV/genética
10.
ACS Chem Biol ; 16(10): 1841-1865, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34569792

RESUMO

Bacterial cells present a wide diversity of saccharides that decorate the cell surface and help mediate interactions with the environment. Many Gram-negative cells express O-antigens, which are long sugar polymers that makeup the distal portion of lipopolysaccharide (LPS) that constitutes the surface of the outer membrane. This review highlights chemical biology tools that have been developed in recent years to facilitate the modulation of O-antigen synthesis and composition, as well as related bacterial polysaccharide pathways, and the detection of unique glycan sequences. Advances in the biochemistry and structural biology of O-antigen biosynthetic machinery are also described, which provide guidance for the design of novel chemical and biomolecular probes. Many of the tools noted here have not yet been utilized in biological systems and offer researchers the opportunity to investigate the complex sugar architecture of Gram-negative cells.


Assuntos
Bactérias Gram-Negativas/química , Antígenos O/metabolismo , Inibidores Enzimáticos/farmacologia , Glicosiltransferases/antagonistas & inibidores , Glicosiltransferases/química , Glicosiltransferases/metabolismo , Bactérias Gram-Negativas/enzimologia , Humanos , Engenharia Metabólica , Sondas Moleculares/química , Sondas Moleculares/farmacologia , Nucleotidiltransferases/antagonistas & inibidores , Nucleotidiltransferases/química , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Antígenos O/química , Engenharia de Proteínas , Especificidade por Substrato/genética
11.
J Mol Model ; 27(10): 291, 2021 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-34546425

RESUMO

Human α-defensin 5 (HD5) is one of cationic antimicrobial peptides which plays a crucial role in an innate immune system in human body. HD5 shows the killing activity against a broad spectrum of pathogenic bacteria by making a pore in a bacterial membrane and penetrating into a cytosol. Nonetheless, its pore-forming mechanisms remain unclear. Thus, in this work, the constant-velocity steered molecular dynamics (SMD) simulation was used to simulate the permeation of a dimeric HD5 into a gram-negative lipopolysaccharide (LPS) membrane model. Arginine-rich HD5 is found to strongly interact with a LPS surface. Upon arrival, arginines on HD5 interact with lipid A head groups (a top part of LPS) and then drag these charged moieties down into a hydrophobic core resulting in the formation of water-filled pore. Although all arginines are found to interact with a membrane, Arg13 and Arg32 appear to play a dominant role in the HD5 adsorption on a gram-negative membrane. Furthermore, one chain of a dimeric HD5 is required for HD5 adhesion. The interactions of arginine-lipid A head groups play a major role in adhering a cationic HD5 on a membrane surface and retarding a HD5 passage in the meantime.


Assuntos
Membrana Externa Bacteriana/química , alfa-Defensinas/química , Arginina/química , Membrana Externa Bacteriana/metabolismo , Bactérias Gram-Negativas/química , Humanos , Ligação de Hidrogênio , Lipopolissacarídeos/química , Simulação de Dinâmica Molecular , Multimerização Proteica , alfa-Defensinas/metabolismo
12.
J Microbiol Methods ; 186: 106241, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33992679

RESUMO

Endotoxin is a kind of lipopolysaccharide that exits on the cell wall of Gram-negative bacteria. It can cause fever, shock or even death when is delivered into human body. So, it is necessary to control the endotoxin contamination for biopharmaceutical products that are mainly administered by intravenous route. Limulus Amebocyte Lysate (LAL)-based tests are usually used to detect endotoxin content in biologics formulations. However, an undesirable phenomenon called "Low Endotoxin Recovery (LER)" often occurs in formulation buffers that usually contain chelating component, such as sodium citrate, and amphiphilic surfactant, such as Tween-20. The occurrence of this LER phenomenon may interfere with endotoxin detection and cause false negative results. In this study, we compared the effect of different sample treatment methods on endotoxin detection and found that the LER phenomenon was better controlled under the conditions of low pH (pH = 5.0), low temperature (2-8 °C) and in the presence of divalent cations in the solution. In addition, although the endotoxin activity was found to have decreased due to LER phenomenon, the particle size distribution of endotoxin determined by dynamic light scattering (DLS) in LER solution did not change obviously, which is different from previous hypothesis about LER phenomenon in literature that the particle size of endotoxin aggregates would decrease under LER conditions. These findings provide some insights into different sample treatment methods for endotoxin detection and give a better understanding and solution on minimizing the LER phenomenon.


Assuntos
Métodos Analíticos de Preparação de Amostras/métodos , Endotoxinas/isolamento & purificação , Bactérias Gram-Negativas/química , Métodos Analíticos de Preparação de Amostras/instrumentação , Animais , Cátions Bivalentes/química , Endotoxinas/química , Endotoxinas/farmacologia , Caranguejos Ferradura , Concentração de Íons de Hidrogênio , Teste do Limulus , Lipopolissacarídeos/química , Lipopolissacarídeos/isolamento & purificação , Tamanho da Partícula , Tensoativos/química
13.
Nat Commun ; 12(1): 2560, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33963193

RESUMO

The commensal fungus Candida albicans often causes life-threatening infections in patients who are immunocompromised with high mortality. A prominent but poorly understood risk factor for the C. albicans commensal‒pathogen transition is the use of broad-spectrum antibiotics. Here, we report that ß-lactam antibiotics cause bacteria to release significant quantities of peptidoglycan fragments that potently induce the invasive hyphal growth of C. albicans. We identify several active peptidoglycan subunits, including tracheal cytotoxin, a molecule produced by many Gram-negative bacteria, and fragments purified from the cell wall of Gram-positive Staphylococcus aureus. Feeding mice with ß-lactam antibiotics causes a peptidoglycan storm that transforms the gut from a niche usually restraining C. albicans in the commensal state to promoting invasive growth, leading to systemic dissemination. Our findings reveal a mechanism underlying a significant risk factor for C. albicans infection, which could inform clinicians regarding future antibiotic selection to minimize this deadly disease incidence.


Assuntos
Candida albicans/patogenicidade , Candidíase/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Peptidoglicano/toxicidade , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , beta-Lactamas/efeitos adversos , Animais , Antibacterianos/efeitos adversos , Candida albicans/crescimento & desenvolvimento , Candidíase/complicações , Candidíase/tratamento farmacológico , Candidíase/patologia , Parede Celular/química , Parede Celular/efeitos dos fármacos , Cromatografia Líquida , Feminino , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Negativas/metabolismo , Humanos , Hifas/crescimento & desenvolvimento , Hifas/patogenicidade , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Peptidoglicano/química , Infecções Estafilocócicas/complicações , Staphylococcus aureus/química , Staphylococcus aureus/metabolismo
14.
ACS Appl Mater Interfaces ; 13(14): 16062-16074, 2021 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-33797891

RESUMO

Gram-negative bacteria are covered by both an inner cytoplasmic membrane (IM) and an outer membrane (OM). Antimicrobial peptides (AMPs) must first permeate through the OM and cell wall before attacking the IM to cause cytoplasmic leakage and kill the bacteria. The bacterial OM is an asymmetric bilayer with the outer leaflet primarily composed of lipopolysaccharides (LPSs) and the inner leaflet composed of phospholipids (PLs). Two cationic α-helical AMPs were designed to target Gram-negative bacteria, a full peptide G(IIKK)3I-NH2 (G3), and a hydrophobic lipopeptide C8-G(IIKK)2I-NH2 (C8G2, with C8 denoting the octanoyl chain). LPS dominates OM functions as the first line of defense against antibiotics, thereby reducing drug susceptibility. This work explores how the two AMPs interact with LPS through several carefully chosen OM models that facilitated measurements from solid-state nuclear magnetic resonance (ss-NMR), small-angle neutron scattering (SANS), and neutron reflectivity (NR). The results revealed that G3 molecules bound preferably to the LPS head region and functioned as bridge molecules to reassemble the dislocated lipids into bilayer stacks. In contrast, C8G2 lipopeptides could quickly penetrate into the central region of the OM to cause direct removal of some membrane lipids. Different structural disruptions implicated different antimicrobial efficacies from these AMPs. The demonstration of the structural features underlying different susceptibilities of the OM to AMPs offers a useful route for the future development of strain-specific AMPs against antimicrobial-resistant pathogens.


Assuntos
Parede Celular/química , Bactérias Gram-Negativas/química , Proteínas Citotóxicas Formadoras de Poros/química , Desenho de Fármacos , Eritrócitos/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Humanos , Bicamadas Lipídicas , Testes de Sensibilidade Microbiana , Proteínas Citotóxicas Formadoras de Poros/farmacologia , Conformação Proteica
15.
Chem Rev ; 121(9): 5479-5596, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33909410

RESUMO

Tripartite efflux pumps and the related type 1 secretion systems (T1SSs) in Gram-negative organisms are diverse in function, energization, and structural organization. They form continuous conduits spanning both the inner and the outer membrane and are composed of three principal components-the energized inner membrane transporters (belonging to ABC, RND, and MFS families), the outer membrane factor channel-like proteins, and linking the two, the periplasmic adaptor proteins (PAPs), also known as the membrane fusion proteins (MFPs). In this review we summarize the recent advances in understanding of structural biology, function, and regulation of these systems, highlighting the previously undescribed role of PAPs in providing a common architectural scaffold across diverse families of transporters. Despite being built from a limited number of basic structural domains, these complexes present a staggering variety of architectures. While key insights have been derived from the RND transporter systems, a closer inspection of the operation and structural organization of different tripartite systems reveals unexpected analogies between them, including those formed around MFS- and ATP-driven transporters, suggesting that they operate around basic common principles. Based on that we are proposing a new integrated model of PAP-mediated communication within the conformational cycling of tripartite systems, which could be expanded to other types of assemblies.


Assuntos
Bactérias Gram-Negativas/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Sistemas de Secreção Tipo I/metabolismo , Transportadores de Cassetes de Ligação de ATP , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Bactérias Gram-Negativas/química , Proteínas de Membrana Transportadoras/química , Simulação de Dinâmica Molecular , Conformação Proteica , Relação Estrutura-Atividade , Sistemas de Secreção Tipo I/química
16.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33836615

RESUMO

Gram-positive bacteria assemble a multilayered cell wall that provides tensile strength to the cell. The cell wall is composed of glycan strands cross-linked by nonribosomally synthesized peptide stems. Herein, we modify the peptide stems of the Gram-positive bacterium Bacillus subtilis with noncanonical electrophilic d-amino acids, which when in proximity to adjacent stem peptides form novel covalent 5,3-cross-links. Approximately 20% of canonical cell-wall cross-links can be replaced with synthetic cross-links. While a low level of synthetic cross-link formation does not affect B. subtilis growth and phenotype, at higher levels cell growth is perturbed and bacteria elongate. A comparison of the accumulation of synthetic cross-links over time in Gram-negative and Gram-positive bacteria highlights key differences between them. The ability to perturb cell-wall architecture with synthetic building blocks provides a novel approach to studying the adaptability, elasticity, and porosity of bacterial cell walls.


Assuntos
Parede Celular/química , Bacilos Gram-Positivos/química , Peptidoglicano/química , Aminoácidos/química , Aminoácidos/metabolismo , Bacillus subtilis/química , Bacillus subtilis/citologia , Bacillus subtilis/crescimento & desenvolvimento , Bacillus subtilis/metabolismo , Parede Celular/metabolismo , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/citologia , Bactérias Gram-Negativas/metabolismo , Bacilos Gram-Positivos/citologia , Bacilos Gram-Positivos/crescimento & desenvolvimento , Bacilos Gram-Positivos/metabolismo , Peptidoglicano/metabolismo , Peptidil Transferases/genética , Peptidil Transferases/metabolismo , Fenótipo
17.
Org Lett ; 23(9): 3477-3480, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33885313

RESUMO

Myrindole A, a bis-indole alkaloid, was isolated from the deep-sea sponge Myrmekioderma sp. The high degree of unsaturation of the molecule complicated the assignment of its structure by standard 2D-NMR experiments but was ultimately achieved by a combination of 1H-15N-HMBC and 1,n-ADEQUATE experiments as well as the comparison of measured and calculated CD spectra. Myrindole A showed antimicrobial activity against Gram-positive and Gram-negative bacteria.


Assuntos
Antibacterianos/farmacologia , Bactérias Gram-Negativas/química , Bactérias Gram-Positivas/química , Alcaloides Indólicos/química , Animais , Antibacterianos/química , Espectroscopia de Ressonância Magnética , Poríferos/química
18.
Chem Biodivers ; 18(4): e2100046, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33636028

RESUMO

From arid, high desert soil samples collected near Bend, Oregon, 19 unique bacteria were isolated. Each strain was identified by 16S rRNA gene sequencing, and their organic extracts were tested for antibacterial and antiproliferative activities. Noteworthy, six extracts (30 %) exhibited strong inhibition resulting in less than 50 % cell proliferation in more than one cancer cell model, tested at 10 µg/mL. Principal component analysis (PCA) of LC/MS data revealed drastic differences in the metabolic profiles found in the organic extracts of these soil bacteria. In total, fourteen potent antibacterial and/or cytotoxic metabolites were isolated via bioactivity-guided fractionation, including two new natural products: a pyrazinone containing tetrapeptide and 7-methoxy-2,3-dimethyl-4H-chromen-4-one, as well as twelve known compounds: furanonaphthoquinone I, bafilomycin C1 and D, FD-594, oligomycin A, chloramphenicol, MY12-62A, rac-sclerone, isosclerone, tunicamycin VII, tunicamycin VIII, and (6S,16S)-anthrabenzoxocinone 1.264-C.


Assuntos
Antibacterianos/farmacologia , Antineoplásicos/farmacologia , Produtos Biológicos/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Solo/química , Antibacterianos/química , Antibacterianos/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Produtos Biológicos/química , Produtos Biológicos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/isolamento & purificação , Bactérias Gram-Positivas/química , Bactérias Gram-Positivas/isolamento & purificação , Humanos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Análise de Componente Principal , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo , Microbiologia do Solo
19.
Chem Rev ; 121(9): 5597-5631, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33596653

RESUMO

Cell envelope plays a dual role in the life of bacteria by simultaneously protecting it from a hostile environment and facilitating access to beneficial molecules. At the heart of this ability lie the restrictive properties of the cellular membrane augmented by efflux transporters, which preclude intracellular penetration of most molecules except with the help of specialized uptake mediators. Recently, kinetic properties of the cell envelope came into focus driven on one hand by the urgent need in new antibiotics and, on the other hand, by experimental and theoretical advances in studies of transmembrane transport. A notable result from these studies is the development of a kinetic formalism that integrates the Michaelis-Menten behavior of individual transporters with transmembrane diffusion and offers a quantitative basis for the analysis of intracellular penetration of bioactive compounds. This review surveys key experimental and computational approaches to the investigation of transport by individual translocators and in whole cells, summarizes key findings from these studies and outlines implications for antibiotic discovery. Special emphasis is placed on Gram-negative bacteria, whose envelope contains two separate membranes. This feature sets these organisms apart from Gram-positive bacteria and eukaryotic cells by providing them with full benefits of the synergy between slow transmembrane diffusion and active efflux.


Assuntos
Bactérias Gram-Negativas/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Membrana Externa Bacteriana/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/efeitos dos fármacos , Humanos , Lipídeos de Membrana/química , Lipídeos de Membrana/metabolismo , Proteínas de Membrana Transportadoras/química , Modelos Biológicos , Simulação de Dinâmica Molecular
20.
J Med Chem ; 64(4): 1816-1834, 2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33538159

RESUMO

The outer membrane (OM) of Gram-negative bacteria provides an efficient barrier against external noxious compounds such as antimicrobial agents. Associated with drug target modification, it contributes to the overall failure of chemotherapy. In the complex OM architecture, Lipid A plays an essential role by anchoring the lipopolysaccharide in the membrane and ensuring the spatial organization between lipids, proteins, and sugars. Currently, the targets of almost all antibiotics are intracellularly located and require translocation across membranes. We report herein an integrated view of Lipid A synthesis, membrane assembly, a structure comparison at the molecular structure level of numerous Gram-negative bacterial species, as well as its recent use as a target for original antibacterial molecules. This review paves the way for a new vision of a key membrane component that acts during bacterial adaptation to environmental stresses and for the development of new weapons against microbial resistance to usual antibiotics.


Assuntos
Adaptação Biológica/fisiologia , Bactérias Gram-Negativas/metabolismo , Lipídeo A/metabolismo , Sequência de Aminoácidos , Animais , Antibacterianos/farmacologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Farmacorresistência Bacteriana/fisiologia , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistência a Múltiplos Medicamentos/fisiologia , Inibidores Enzimáticos/farmacologia , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/efeitos dos fármacos , Humanos , Lipídeo A/biossíntese , Lipídeo A/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...