Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 341
Filtrar
1.
Int J Mol Sci ; 22(22)2021 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-34829983

RESUMO

The BAM is a macromolecular machine responsible for the folding and the insertion of integral proteins into the outer membrane of diderm Gram-negative bacteria. In Escherichia coli, it consists of a transmembrane ß-barrel subunit, BamA, and four outer membrane lipoproteins (BamB-E). Using BAM-specific antibodies, in E. coli cells, the complex is shown to localize in the lateral wall in foci. The machinery was shown to be enriched at midcell with specific cell cycle timing. The inhibition of septation by aztreonam did not alter the BAM midcell localization substantially. Furthermore, the absence of late cell division proteins at midcell did not impact BAM timing or localization. These results imply that the BAM enrichment at the site of constriction does not require an active cell division machinery. Expression of the Tre1 toxin, which impairs the FtsZ filamentation and therefore midcell localization, resulted in the complete loss of BAM midcell enrichment. A similar effect was observed for YidC, which is involved in the membrane insertion of cell division proteins in the inner membrane. The presence of the Z-ring is needed for preseptal peptidoglycan (PG) synthesis. As BAM was shown to be embedded in the PG layer, it is possible that BAM is inserted preferentially simultaneously with de novo PG synthesis to facilitate the insertion of OMPs in the newly synthesized outer membrane.


Assuntos
Proteínas da Membrana Bacteriana Externa/ultraestrutura , Proteínas de Bactérias/genética , Proteínas do Citoesqueleto/genética , Proteínas de Escherichia coli/genética , Proteínas de Membrana Transportadoras/genética , Proteínas da Membrana Bacteriana Externa/genética , Proteínas de Bactérias/ultraestrutura , Divisão Celular/genética , Proteínas do Citoesqueleto/ultraestrutura , Escherichia coli/química , Escherichia coli/genética , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/ultraestrutura , Bactérias Gram-Negativas/genética , Bactérias Gram-Negativas/ultraestrutura , Lipoproteínas/genética , Lipoproteínas/ultraestrutura , Proteínas de Membrana Transportadoras/ultraestrutura , Dobramento de Proteína , Multimerização Proteica/genética
2.
Science ; 373(6552)2021 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-34437126

RESUMO

Activation of cell-autonomous defense by the immune cytokine interferon-γ (IFN-γ) is critical to the control of life-threatening infections in humans. IFN-γ induces the expression of hundreds of host proteins in all nucleated cells and tissues, yet many of these proteins remain uncharacterized. We screened 19,050 human genes by CRISPR-Cas9 mutagenesis and identified IFN-γ-induced apolipoprotein L3 (APOL3) as a potent bactericidal agent protecting multiple non-immune barrier cell types against infection. Canonical apolipoproteins typically solubilize mammalian lipids for extracellular transport; APOL3 instead targeted cytosol-invasive bacteria to dissolve their anionic membranes into human-bacterial lipoprotein nanodiscs detected by native mass spectrometry and visualized by single-particle cryo-electron microscopy. Thus, humans have harnessed the detergent-like properties of extracellular apolipoproteins to fashion an intracellular lysin, thereby endowing resident nonimmune cells with a mechanism to achieve sterilizing immunity.


Assuntos
Apolipoproteínas L/metabolismo , Membrana Celular/metabolismo , Citosol/microbiologia , Bactérias Gram-Negativas/fisiologia , Interferon gama/imunologia , Apolipoproteínas L/química , Apolipoproteínas L/genética , Membrana Externa Bacteriana/metabolismo , Bacteriólise , Sistemas CRISPR-Cas , Membrana Celular/química , Membrana Celular/ultraestrutura , Permeabilidade da Membrana Celular , Células Cultivadas , Detergentes/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Edição de Genes , Bactérias Gram-Negativas/imunologia , Bactérias Gram-Negativas/patogenicidade , Bactérias Gram-Negativas/ultraestrutura , Humanos , Imunidade Inata , Lipoproteínas/química , Viabilidade Microbiana , Antígenos O/metabolismo , Domínios Proteicos , Salmonella typhimurium/imunologia , Salmonella typhimurium/patogenicidade , Salmonella typhimurium/fisiologia , Salmonella typhimurium/ultraestrutura , Solubilidade
3.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33723050

RESUMO

Multiple gram-negative bacteria encode type III secretion systems (T3SS) that allow them to inject effector proteins directly into host cells to facilitate colonization. To be secreted, effector proteins must be at least partially unfolded to pass through the narrow needle-like channel (diameter <2 nm) of the T3SS. Fusion of effector proteins to tightly packed proteins-such as GFP, ubiquitin, or dihydrofolate reductase (DHFR)-impairs secretion and results in obstruction of the T3SS. Prior observation that unfolding can become rate-limiting for secretion has led to the model that T3SS effector proteins have low thermodynamic stability, facilitating their secretion. Here, we first show that the unfolding free energy ([Formula: see text]) of two Salmonella effector proteins, SptP and SopE2, are 6.9 and 6.0 kcal/mol, respectively, typical for globular proteins and similar to published [Formula: see text] for GFP, ubiquitin, and DHFR. Next, we mechanically unfolded individual SptP and SopE2 molecules by atomic force microscopy (AFM)-based force spectroscopy. SptP and SopE2 unfolded at low force (Funfold ≤ 17 pN at 100 nm/s), making them among the most mechanically labile proteins studied to date by AFM. Moreover, their mechanical compliance is large, as measured by the distance to the transition state (Δx‡ = 1.6 and 1.5 nm for SptP and SopE2, respectively). In contrast, prior measurements of GFP, ubiquitin, and DHFR show them to be mechanically robust (Funfold > 80 pN) and brittle (Δx‡ < 0.4 nm). These results suggest that effector protein unfolding by T3SS is a mechanical process and that mechanical lability facilitates efficient effector protein secretion.


Assuntos
Fenômenos Fisiológicos Bacterianos , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Proteínas de Bactérias/química , Bactérias Gram-Negativas/fisiologia , Bactérias Gram-Negativas/ultraestrutura , Microscopia de Força Atômica , Estabilidade Proteica , Salmonella/fisiologia , Salmonella/ultraestrutura , Termodinâmica
4.
Mol Microbiol ; 115(3): 436-452, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33326642

RESUMO

Bacterial type IV secretion systems (T4SSs) are a functionally diverse translocation superfamily. They consist mainly of two large subfamilies: (i) conjugation systems that mediate interbacterial DNA transfer and (ii) effector translocators that deliver effector macromolecules into prokaryotic or eukaryotic cells. A few other T4SSs export DNA or proteins to the milieu, or import exogenous DNA. The T4SSs are defined by 6 or 12 conserved "core" subunits that respectively elaborate "minimized" systems in Gram-positive or -negative bacteria. However, many "expanded" T4SSs are built from "core" subunits plus numerous others that are system-specific, which presumptively broadens functional capabilities. Recently, there has been exciting progress in defining T4SS assembly pathways and architectures using a combination of fluorescence and cryoelectron microscopy. This review will highlight advances in our knowledge of structure-function relationships for model Gram-negative bacterial T4SSs, including "minimized" systems resembling the Agrobacterium tumefaciens VirB/VirD4 T4SS and "expanded" systems represented by the Helicobacter pylori Cag, Legionella pneumophila Dot/Icm, and F plasmid-encoded Tra T4SSs. Detailed studies of these model systems are generating new insights, some at atomic resolution, to long-standing questions concerning mechanisms of substrate recruitment, T4SS channel architecture, conjugative pilus assembly, and machine adaptations contributing to T4SS functional versatility.


Assuntos
Conjugação Genética , Fímbrias Bacterianas/fisiologia , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/fisiologia , Sistemas de Translocação de Proteínas/metabolismo , Sistemas de Secreção Tipo IV/química , Sistemas de Secreção Tipo IV/fisiologia , Agrobacterium tumefaciens/química , Agrobacterium tumefaciens/fisiologia , Motivos de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/fisiologia , Microscopia Crioeletrônica , Bactérias Gram-Negativas/ultraestrutura , Infecções por Bactérias Gram-Negativas/microbiologia , Helicobacter pylori/química , Helicobacter pylori/fisiologia , Humanos , Legionella pneumophila/química , Legionella pneumophila/fisiologia , Simulação de Acoplamento Molecular , Sistemas de Translocação de Proteínas/química , Sistemas de Translocação de Proteínas/ultraestrutura , Relação Estrutura-Atividade , Sistemas de Secreção Tipo IV/ultraestrutura
5.
Toxins (Basel) ; 12(12)2020 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-33291852

RESUMO

The abuse of antibiotics and the consequent increase of drug-resistant bacteria constitute a serious threat to human health, and new antibiotics are urgently needed. Research shows that antimicrobial peptides produced by natural organisms are potential substitutes for antibiotics. Based on Deinagkistrodonacutus (known as five-pacer viper) genome bioinformatics analysis, we discovered a new cathelicidin antibacterial peptide which was called FP-CATH. Circular dichromatic analysis showed a typical helical structure. FP-CATH showed broad-spectrum antibacterial activity. It has antibacterial activity to Gram-negative bacteria and Gram-positive bacteria including methicillin-resistant Staphylococcus aureus (MRSA). The results of transmission electron microscopy (TEM) and scanning electron microscopy (SEM) showed that FP-CATH could cause the change of bacterial cell integrity, having a destructive effect on Gram-negative bacteria and inducing Gram-positive bacterial surface formation of vesicular structure. FP-CATH could bind to LPS and showed strong binding ability to bacterial DNA. In vivo, FP-CATH can improve the survival rate of nematodes in bacterial invasion experiments, and has a certain protective effect on nematodes. To sum up, FP-CATH is likely to play a role in multiple mechanisms of antibacterial action by impacting bacterial cell integrity and binding to bacterial biomolecules. It is hoped that the study of FP-CATH antibacterial mechanisms will prove useful for development of novel antibiotics.


Assuntos
Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Crotalinae/genética , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/microbiologia , Eritrócitos/efeitos dos fármacos , Genoma , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Negativas/crescimento & desenvolvimento , Bactérias Gram-Negativas/ultraestrutura , Bactérias Gram-Positivas/efeitos dos fármacos , Bactérias Gram-Positivas/crescimento & desenvolvimento , Bactérias Gram-Positivas/ultraestrutura , Humanos , Testes de Sensibilidade Microbiana , Catelicidinas
6.
Curr Opin Chem Biol ; 59: 193-201, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33070100

RESUMO

Extracellular electron transfer via filamentous protein appendages called 'microbial nanowires' has long been studied in Geobacter and other bacteria because of their crucial role in globally-important environmental processes and their applications for bioenergy, biofuels, and bioelectronics. Thousands of papers thought these nanowires as pili without direct evidence. Here, we summarize recent discoveries that could help resolve two decades of confounding observations. Using cryo-electron microscopy with multimodal functional imaging and a suite of electrical, biochemical, and physiological studies, we find that rather than pili, nanowires are composed of cytochromes OmcS and OmcZ that transport electrons via seamless stacking of hemes over micrometers. We discuss the physiological need for two different nanowires and their potential applications for sensing, synthesis, and energy production.


Assuntos
Proteínas de Bactérias/metabolismo , Citocromos/metabolismo , Fímbrias Bacterianas/metabolismo , Bactérias Gram-Negativas/metabolismo , Proteínas de Bactérias/ultraestrutura , Citocromos/ultraestrutura , Transporte de Elétrons , Fímbrias Bacterianas/ultraestrutura , Geobacter/metabolismo , Geobacter/ultraestrutura , Bactérias Gram-Negativas/ultraestrutura , Modelos Moleculares , Nanofios/ultraestrutura
7.
Sci Rep ; 10(1): 12087, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32694655

RESUMO

Bacteriophage-encoded endolysins degrading the bacterial peptidoglycan are promising antibacterials for combating antibiotic-resistant bacteria. However, endolysins have limited use against Gram-negative bacteria, since the outer membrane prevents access to the peptidoglycan. Here, we present Innolysins, an innovative concept for engineering endolysins to exert antibacterial activity against Gram-negative bacteria. Innolysins combine the enzymatic activity of endolysins with the binding capacity of phage receptor binding proteins (RBPs). As proof-of-concept, we constructed 12 Innolysins by fusing phage T5 endolysin and RBP Pb5 in different configurations. One of these, Innolysin Ec6 displayed antibacterial activity against Escherichia coli only in the presence of Pb5 receptor FhuA, leading to 1.22 ± 0.12 log reduction in cell counts. Accordingly, other bacterial species carrying FhuA homologs such as Shigella sonnei and Pseudomonas aeruginosa were sensitive to Innolysin Ec6. To enhance the antibacterial activity, we further constructed 228 novel Innolysins by fusing 23 endolysins with Pb5. High-throughput screening allowed to select Innolysin Ec21 as the best antibacterial candidate, leading to 2.20 ± 0.09 log reduction in E. coli counts. Interestingly, Innolysin Ec21 also displayed bactericidal activity against E. coli resistant to third-generation cephalosporins, reaching a 3.31 ± 0.53 log reduction in cell counts. Overall, the Innolysin approach expands previous endolysin-engineering strategies, allowing customization of endolysins by exploiting phage RBPs to specifically target Gram-negative bacteria.


Assuntos
Endopeptidases/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Proteínas Virais/farmacologia , Sequência de Aminoácidos , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/genética , Bacteriófagos/enzimologia , Desintegrinas/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/ultraestrutura , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Bactérias Gram-Negativas/crescimento & desenvolvimento , Bactérias Gram-Negativas/ultraestrutura , Bactérias Gram-Negativas/virologia
8.
J Struct Biol ; 211(2): 107544, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32512156

RESUMO

The expression of ß-lactamases is a major mechanism of bacterial resistance to the ß-lactam antibiotics. Four molecular classes of ß-lactamases have been described (A, B, C and D), however until recently the class D enzymes were thought to exist only in Gram-negative bacteria. In the last few years, class D enzymes have been discovered in several species of Gram-positive microorganisms, such as Bacillus and Clostridia, and an investigation of their kinetic and structural properties has begun in earnest. Interestingly, it was observed that some species of Bacillus produce two distinct class D ß-lactamases, one highly active and the other with only basal catalytic activity. Analysis of amino acid sequences of active (BPU-1 from Bacillus pumilus) and inactive (BSU-2 from Bacillus subtilis and BAT-2 from Bacillus atrophaeus) enzymes suggests that presence of three additional amino acid residues in one of the surface loops of inefficient ß-lactamases may be responsible for their severely diminished activity. Our structural and docking studies show that the elongated loop of these enzymes severely restricts binding of substrates. Deletion of the three residues from the loops of BSU-2 and BAT-2 ß-lactamases relieves the steric hindrance and results in a significant increase in the catalytic activity of the enzymes. These data show that this surface loop plays an important role in modulation of the catalytic activity of Bacillus class D ß-lactamases.


Assuntos
Antibacterianos/química , Farmacorresistência Bacteriana/genética , Conformação Proteica , beta-Lactamases/ultraestrutura , Sequência de Aminoácidos/genética , Bacillus pumilus/efeitos dos fármacos , Bacillus pumilus/enzimologia , Bacillus subtilis/enzimologia , Domínio Catalítico/genética , Clostridiaceae/enzimologia , Cristalografia por Raios X , Bactérias Gram-Negativas/enzimologia , Bactérias Gram-Negativas/ultraestrutura , Humanos , Simulação de Acoplamento Molecular , Propriedades de Superfície , beta-Lactamases/química , beta-Lactamases/genética
9.
Biomolecules ; 10(3)2020 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-32120823

RESUMO

A number of Gram-negative bacteria have a membrane surrounding their flagella, referred to as the flagellar sheath, which is continuous with the outer membrane. The flagellar sheath was initially described in Vibrio metschnikovii in the early 1950s as an extension of the outer cell wall layer that completely surrounded the flagellar filament. Subsequent studies identified other bacteria that possess flagellar sheaths, most of which are restricted to a few genera of the phylum Proteobacteria. Biochemical analysis of the flagellar sheaths from a few bacterial species revealed the presence of lipopolysaccharide, phospholipids, and outer membrane proteins in the sheath. Some proteins localize preferentially to the flagellar sheath, indicating mechanisms exist for protein partitioning to the sheath. Recent cryo-electron tomography studies have yielded high resolution images of the flagellar sheath and other structures closely associated with the sheath, which has generated insights and new hypotheses for how the flagellar sheath is synthesized. Various functions have been proposed for the flagellar sheath, including preventing disassociation of the flagellin subunits in the presence of gastric acid, avoiding activation of the host innate immune response by flagellin, activating the host immune response, adherence to host cells, and protecting the bacterium from bacteriophages.


Assuntos
Flagelos/genética , Bactérias Gram-Negativas/genética , Infecções por Bactérias Gram-Negativas/microbiologia , Filogenia , Membrana Externa Bacteriana/metabolismo , Membrana Externa Bacteriana/ultraestrutura , Flagelos/metabolismo , Flagelos/ultraestrutura , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Negativas/ultraestrutura , Humanos , Vibrio/genética , Vibrio/metabolismo , Vibrio/ultraestrutura
10.
Mol Microbiol ; 113(3): 659-671, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31975449

RESUMO

How, when and why the transition between cell envelopes with one membrane (Gram-positives or monoderms) and two (Gram-negative or diderms) occurred in Bacteria is a key unanswered question in evolutionary biology. Different hypotheses have been put forward, suggesting that either the monoderm or the diderm phenotype is ancestral. The existence of diderm members in the classically monoderm Firmicutes challenges the Gram-positive/Gram-negative divide and provides a great opportunity to tackle the issue. In this review, we present current knowledge on the diversity of bacterial cell envelopes, including these atypical Firmicutes. We discuss how phylogenomic analysis supports the hypothesis that the diderm cell envelope architecture is an ancestral character in the Firmicutes, and that the monoderm phenotype in this phylum arose multiple times independently by loss of the outer membrane. Given the overwhelming distribution of diderm phenotypes with respect to monoderm ones, this scenario likely extends to the ancestor of all bacteria. Finally, we discuss the recent development of genetic tools for Veillonella parvula, a diderm Firmicute member of the human microbiome, which indicates it as an emerging new experimental model to investigate fundamental aspects of the diderm/monoderm transition.


Assuntos
Membrana Celular/genética , Bactérias Gram-Negativas/ultraestrutura , Bactérias Gram-Positivas/ultraestrutura , Bactérias/genética , Bactérias/metabolismo , Evolução Biológica , Membrana Celular/ultraestrutura , Parede Celular/genética , Parede Celular/ultraestrutura , Evolução Molecular , Firmicutes/classificação , Firmicutes/genética , Bactérias Gram-Negativas/genética , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Positivas/genética , Bactérias Gram-Positivas/metabolismo , Lipopolissacarídeos , Filogenia
11.
Biochim Biophys Acta Biomembr ; 1862(1): 183062, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31520605

RESUMO

The ß-barrel assembly machinery (BAM) is responsible for the biogenesis of outer membrane proteins (OMPs) into the outer membranes of Gram-negative bacteria. These OMPs have a membrane-embedded domain consisting of a ß-barrel fold which can vary from 8 to 36 ß-strands, with each serving a diverse role in the cell such as nutrient uptake and virulence. BAM was first identified nearly two decades ago, but only recently has the molecular structure of the full complex been reported. Together with many years of functional characterization, we have a significantly clearer depiction of BAM's structure, the intra-complex interactions, conformational changes that BAM may undergo during OMP biogenesis, and the role chaperones may play. But still, despite advances over the past two decades, the mechanism for BAM-mediated OMP biogenesis remains elusive. Over the years, several theories have been proposed that have varying degrees of support from the literature, but none has of yet been conclusive enough to be widely accepted as the sole mechanism. We will present a brief history of BAM, the recent work on the structures of BAM, and a critical analysis of the current theories for how it may function.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Conformação Proteica em Folha beta , Proteínas da Membrana Bacteriana Externa/biossíntese , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/ultraestrutura , Estrutura Secundária de Proteína
12.
J Mol Biol ; 432(4): 1008-1019, 2020 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-31870848

RESUMO

The outer membrane (OM) of Gram-negative bacteria is asymmetric, with lipopolysaccharides (LPSs) on the outer surface and phospholipids (PLs) on the inner surface. This unique organization of OM makes Gram-negative bacteria resistant to many toxic chemicals. How this asymmetric distribution of lipids is maintained has been studied for decades with previous reports of an Mla (Maintenance of OM Lipid Asymmetry) system to be involved. Furthermore, the OM of Gram-negative bacteria is about 20 nm away from inner membrane (IM) where the lipids are synthesized. Therefore, how nascent lipids travel across the periplasmic space and arrive at the inner surface of OM is another interesting question. YebT is a homologue of MlaD in the Mla pathway, but its role in lipid distribution of the OM and IM is largely unknown. Here we report the first high-resolution (~3.0 Å) cryo-EM structure of full-length E. coli YebT in a substrate-bound state. Our structure with details of lipid interaction indicates that YebT is a lipid transporter spanning between IM and OM. We also demonstrate the symmetry mismatch in YebT and the existence of many other conformations of YebT revealing the intrinsic dynamics of this lipid channel. And a brief discussion on possible mechanisms of lipid transport is also included.


Assuntos
Microscopia Crioeletrônica/métodos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/ultraestrutura , Proteínas de Membrana/química , Proteínas de Membrana/ultraestrutura , Sítios de Ligação , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Proteínas de Escherichia coli/metabolismo , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Negativas/ultraestrutura , Lipídeos de Membrana/química , Lipídeos de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
13.
Int J Mol Sci ; 20(16)2019 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-31416220

RESUMO

Antibacterial peptides (APMs) are a new type of antibacterial substance. The relationship between their structure and function remains indistinct; in particular, there is a lack of a definitive and fixed template for designing new antimicrobial peptides. Previous studies have shown that porcine Protegrin-1 (PG-1) exhibits considerable antimicrobial activity and cytotoxicity. In this study, to reduce cytotoxicity and increase cell selectivity, we designed histidine-rich peptides based on the sequence template RR(XY)2XDPGX(YX)2RR-NH2, where X represents I, W, V, and F. The results showed that the peptides form more ß-hairpin structures in a lipid-rich environment that mimics cell membranes. Among them, the antimicrobial peptide HV2 showed strong antibacterial activity against Gram-negative strains and almost no toxicity to normal cells. The results of our analysis of its antibacterial mechanism showed that peptide HV2 acts on the bacterial cell membrane to increase its permeability, resulting in cell membrane disruption and death. Furthermore, peptide HV2 inhibited bacterial movement in a concentration-dependent manner and had a more robust anti-inflammatory effect by inhibiting the production of TNF-α. In summary, peptide HV2 exhibits high bactericidal activity and cell selectivity, making it a promising candidate for future use as an antibiotic.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Histidina , Peptídeos/química , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Permeabilidade da Membrana Celular , Desenho de Fármacos , Bactérias Gram-Negativas/ultraestrutura , Histidina/química , Camundongos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Conformação Molecular , Conformação Proteica em Folha beta , Células RAW 264.7
14.
Sci Rep ; 9(1): 8815, 2019 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-31217508

RESUMO

'Black silicon' (bSi) samples with surfaces covered in nanoneedles of length ~5 µm were fabricated using a plasma etching process and then coated with a conformal uniform layer of diamond using hot filament chemical vapour deposition to produce 'black diamond' (bD) nanostructures. The diamond needles were then chemically terminated with H, O, NH2 or F using plasma treatment, and the hydrophilicity of the resulting surfaces were assessed using water droplet contact-angle measurements, and scaled in the order O > H ≈NH2 >F, with the F-terminated surface being superhydrophobic. The effectiveness of these differently terminated bD needles in killing the Gram-negative bacterium E. coli was semi-quantified by Live/Dead staining and fluorescence microscopy, and visualised by environmental scanning electron microscopy. The total number of adhered bacteria was consistent for all the nanostructured bD surfaces at around 50% of the value for the flat diamond control. This, combined with a chemical bactericidal effect of 20-30%, shows that the nanostructured bD surfaces supported significantly fewer viable E. coli than flat surfaces. Moreover, the bD surfaces were particularly effective at preventing the establishment of bacterial aggregates - a precursor to biofilm formation. The percentage of dead bacteria also decreased as a function of hydrophilicity. These results are consistent with a predominantly mechanical mechanism for bacteria death based on the stretching and disruption of the cell membrane, combined with an additional effect from the chemical nature of the surface.


Assuntos
Antibacterianos/farmacologia , Diamante/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Aderência Bacteriana/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Escherichia coli/ultraestrutura , Bactérias Gram-Negativas/ultraestrutura , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Silício/farmacologia , Água
15.
Biochem Biophys Res Commun ; 514(2): 497-502, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31056261

RESUMO

Cationic antimicrobial peptides (CAMPs) are important antibiotics because they possess a broad spectrum of activity against both Gram-positive and Gram-negative bacteria, including those resistant to traditional antibiotics. The cyclic peptide bactenecin is a 12-amino acid CAMP that contains one intramolecular disulfide bond. To improve the antibacterial activity of bactenecin, we designed and synthesized several bactenecin analogs by applying multiple approaches, including amino acid substitution, use of the d-enantiomeric form, and lipidation. Among the synthetic analogs, d-enantiomeric bactenecin conjugated to capric acid, which we named dBacK-(cap), exhibited a significantly enhanced antibacterial spectrum with MIC values ranging from 1 to 8 µM against both Gram-positive and Gram-negative bacteria, including some drug-resistant bacteria. Upon exposure to dBacK-(cap), S. aureus cells were killed within 1 h at the MIC value, but full inactivation of E. coli required over 2 h. These results indicate that covalent addition of a d-amino acid and a fatty acid to bactenecin is the most effective approach for enhancing its antibacterial activity.


Assuntos
Antibacterianos/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Peptídeos Cíclicos/farmacologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Antibacterianos/síntese química , Antibacterianos/química , Permeabilidade da Membrana Celular , Desenho de Fármacos , Bactérias Gram-Negativas/citologia , Bactérias Gram-Negativas/ultraestrutura , Bactérias Gram-Positivas/citologia , Bactérias Gram-Positivas/ultraestrutura , Cinética , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/química
16.
ACS Nano ; 13(5): 5133-5146, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-30964642

RESUMO

Medicinal leads that are also compatible with imaging technologies are attractive, as they facilitate the development of therapeutics through direct mechanistic observations at the molecular level. In this context, the uptake and antimicrobial activities of several luminescent dinuclear RuII complexes against E. coli were assessed and compared to results obtained for another ESKAPE pathogen, the Gram-positive major opportunistic pathogen Enterococcus faecalis, V583. The most promising lead displays potent activity, particularly against the Gram-negative bacteria, and potency is retained in the uropathogenic multidrug resistant EC958 ST131 strain. Exploiting the inherent luminescent properties of this complex, super-resolution STED nanoscopy was used to image its initial localization at/in cellular membranes and its subsequent transfer to the cell poles. Membrane damage assays confirm that the complex disrupts the bacterial membrane structure before internalization. Mammalian cell culture and animal model studies indicate that the complex is not toxic to eukaryotes, even at concentrations that are several orders of magnitude higher than its minimum inhibitory concentration (MIC). Taken together, these results have identified a lead molecular architecture for hard-to-treat, multiresistant, Gram-negative bacteria, which displays activities that are already comparable to optimized natural product-based leads.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Nanotecnologia/métodos , Trifosfato de Adenosina/metabolismo , Animais , Antibacterianos/toxicidade , Bactérias Gram-Negativas/ultraestrutura , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Mariposas/efeitos dos fármacos , Rutênio/química , Rutênio/farmacologia
17.
Biochim Biophys Acta Biomembr ; 1861(6): 1057-1068, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30890470

RESUMO

Polychlorinated biphenyls (PCB) are persistent organic pollutants that due to their chemical resistivity and inflammability found multiple applications. In spite of the global ban for PCB production, due to their long half-lives periods, PCB accumulate in the soils, so effective bioremediation of the polluted lands is of crucial importance. Some of the 209 PCB congeners exhibit increased toxicity to soil bacteria and their presence impoverish the soil decomposer community and slows down the degradation of environmental pollutants in the soils. The exact mechanism of PCB antimicrobial activity is unknown, but it is strictly related with the membrane activity of PCB. Therefore, to shed light on these interactions we applied Langmuir monolayers formed by selected phospholipids as model bacterial membranes. In our studies we tested 5 PCB congeners differing in the degree of chlorination and the distribution of the chlorine substituents around the biphenyl frame. Special attention was paid to tetra-substituted PCB because of their increased presence in the environment and disubstituted PCB being their degradation products. To characterize the model membranes as Langmuir monolayers, we used surface pressure measurements, Brewster angle microscopy and Grazing Incidence X-ray Diffraction. It turned out that among the tetra-substituted PCB the ortho-substituted non-dioxin like compound was much more membrane destructive than the flat dioxin-like congener. On the contrary, among the di-substituted PCB the flat para-substituted 2,2'-dichlorobiphenyl turned out to exhibit high membrane activity.


Assuntos
Membrana Celular/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Halogenação , Lipídeos de Membrana/metabolismo , Bifenilos Policlorados/farmacologia , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Negativas/ultraestrutura , Bactérias Gram-Positivas/metabolismo , Bactérias Gram-Positivas/ultraestrutura , Microscopia/métodos , Fosfolipídeos/metabolismo , Difração de Raios X/métodos
18.
J Med Chem ; 62(5): 2286-2304, 2019 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-30742437

RESUMO

Poor proteolytic resistance is an urgent problem to be solved in the clinical application of antimicrobial peptides (AMPs), yet common solutions, such as complicated chemical modifications and utilization of d-amino acids, greatly increase the difficulty and cost of producing AMPs. In this work, a set of novel peptides was synthesized based on an antitrypsin/antichymotrypsin hydrolytic peptide structure unit (XYPX) n (X represents I, L, and V; Y represents R and K), which was designed using a systematic natural amino acid arrangement. Of these peptides, 16 with seven repeat units had the highest average selectivity index (GMSI = 99.07) for all of the Gram-negative bacteria tested and remained highly effective in combating Escherichia coli infection in vivo. Importantly, 16 also had dramatic resistance to a high concentration of trypsin/chymotrypsin hydrolysis and exerted bactericidal activity through a membrane-disruptive mechanism. Overall, these findings provide new approaches for the development of antiprotease hydrolytic peptides that target Gram-negative bacteria.


Assuntos
Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Animais , Peptídeos Catiônicos Antimicrobianos/química , Materiais Biocompatíveis , Bactérias Gram-Negativas/ultraestrutura , Células HEK293 , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Proteólise , Células RAW 264.7 , Espectrometria de Fluorescência
19.
Pol J Vet Sci ; 21(3): 533-542, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30468326

RESUMO

MDAP-2 is a new antibacterial peptide with a unique structure that was isolated from house- flies. However, its biological characteristics and antibacterial mechanisms against bacteria are still poorly understood. To study the biological characteristics, antibacterial activity, hemolytic activi- ty, cytotoxicity to mammalian cells, and the secondary structure of MDAP-2 were detected; the results showed that MDAP-2 displayed high antibacterial activity against all of the tested Gram-negative bacteria. MDAP-2 had lower hemolytic activity to rabbit red blood cells; only 3.4% hemolytic activity was observed at a concentration of 800µg/ml. MDAP-2 also had lower cytotoxicity to mammalian cells; IC50 values for HEK-293 cells, VERO cells, and IPEC-J2 cells were greater than 1000 µg/ml. The circular dichroism (CD) spectra showed that the peptide most- ly has α-helical properties and some ß-fold structure in water and in membrane-like conditions. MDAP-2 is therefore a promising antibacterial agent against Gram-negative bacteria. To deter- mine the antibacterial mechanism(s) of action, fluorescent probes, flow cytometry, and transmis- sion electron microscopy (TEM) were used to study the effects of MDAP-2 on membrane perme- ability, polarization ability, and integrity of Gram-negative bacteria. The results indicated that the peptide caused membrane depolarization, increased membrane permeability, and destroyed membrane integrity. In conclusion, MDAP-2 is a broad-spectrum, lower hemolytic activity, and lower cytotoxicity antibacterial peptide, which is mainly effective on Gram-negative bacteria. It exerts its antimicrobial effects by causing bacterial cytoplasm membrane depolarization, increas- ing cell membrane permeability and disturbing the membrane integrity of Gram-negative bacte- ria. MDAP-2 may offer a new strategy to for defense against Gram-negative bacteria.


Assuntos
Antibacterianos/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Peptídeos/farmacologia , Animais , Antibacterianos/química , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Dicroísmo Circular , Bactérias Gram-Negativas/ultraestrutura , Células HEK293 , Moscas Domésticas/química , Humanos , Proteínas de Insetos/química , Proteínas de Insetos/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Transmissão , Células Vero
20.
PLoS Pathog ; 14(8): e1007240, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30138458

RESUMO

Inflammatory caspase-11/4/5 recognize cytosolic LPS from invading Gram-negative bacteria and induce pyroptosis and cytokine release, forming rapid innate antibacterial defenses. Since extracellular or vacuole-constrained bacteria are thought to rarely access the cytoplasm, how their LPS are exposed to the cytosolic sensors is a critical event for pathogen recognition. Hemolysin is a pore-forming bacterial toxin, which was generally accepted to rupture cell membrane, leading to cell lysis. Whether and how hemolysin participates in non-canonical inflammasome signaling remains undiscovered. Here, we show that hemolysin-overexpressed enterobacteria triggered significantly increased caspase-4 activation in human intestinal epithelial cell lines. Hemolysin promoted LPS cytosolic delivery from extracellular bacteria through dynamin-dependent endocytosis. Further, we revealed that hemolysin was largely associated with bacterial outer membrane vesicles (OMVs) and induced rupture of OMV-containing vacuoles, subsequently increasing LPS exposure to the cytosolic sensor. Accordingly, overexpression of hemolysin promoted caspase-11 dependent IL-18 secretion and gut inflammation in mice, which was associated with restricting bacterial colonization in vivo. Together, our work reveals a concept that hemolysin promotes noncanonical inflammasome activation via liberating OMVs for cytosolic LPS sensing, which offers insights into innate immune surveillance of dysregulated hemolysin via caspase-11/4 in intestinal antibacterial defenses.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Vesículas Extracelulares/metabolismo , Proteínas Hemolisinas/genética , Imunidade Inata/genética , Lipopolissacarídeos/metabolismo , Animais , Células CACO-2 , Caspases/genética , Caspases/metabolismo , Caspases Iniciadoras/genética , Caspases Iniciadoras/metabolismo , Citosol/metabolismo , Bactérias Gram-Negativas/imunologia , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Negativas/ultraestrutura , Células HEK293 , Células HT29 , Células HeLa , Proteínas Hemolisinas/metabolismo , Humanos , Inflamassomos/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transfecção , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...