Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 231
Filtrar
1.
Acta Microbiol Immunol Hung ; 71(2): 134-139, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38837239

RESUMO

The rate of pandrug-resistant Acinetobacter baumannii strains is on the rise in all continents. This bacterium can acquire resistance to all antibiotics, even to colistin. Alterations in the lipid A or/and the two-component pmrAB were earlier detected in colistin resistance. We investigated and analyzed two strains of A. baumannii (ABRC1 and ABRC2) isolated from two patients admitted to intensive care unit with a septic shock. Both strains were resistant to all tested antibiotics including colistin with a MIC >256 mg L-1. Colistin resistance genes (pmrA, pmrB, lpxA, lpxC, lpxD, and lpsB) of two strains (ABRC1 and ABRC2) were investigated by PCR and sequencing. Obtained nucleic acid sequences were aligned with reference sequences of ATCC 19606 and 17987. In this study two amino acid mutations, N287D in the lpxC gene and E117K in the lpxD gene, were detected in both ABRC1 and ABRC2 strains. ABRC1 had an additional H200L mutation in the pmrA gene. Both colistin resistant strains harbored the same A138T mutation in the pmrB gene. The ABRC2 strain also had an alteration in the kinase domain, specifically an R263S substitution of the histidine kinase domain. Three identical mutations were found in the lpsB gene of both A. baumannii strains: Q216K + H218G + S219E. As a result, a newly deduced protein sequence in both ABRC1 and ABRC2 strains differed from those described in ATCC 17978 and 19606 strains was determined. Colistin resistance is multifactorial in A. baumannii. In our study we detected novel mutations in colistin resistant A. baumannii clinical isolates.


Assuntos
Infecções por Acinetobacter , Acinetobacter baumannii , Antibacterianos , Proteínas de Bactérias , Lipídeo A , Testes de Sensibilidade Microbiana , Acinetobacter baumannii/genética , Acinetobacter baumannii/efeitos dos fármacos , Acinetobacter baumannii/metabolismo , Humanos , Lipídeo A/genética , Lipídeo A/metabolismo , Lipídeo A/biossíntese , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Antibacterianos/farmacologia , Infecções por Acinetobacter/microbiologia , Farmacorresistência Bacteriana/genética , Polimixinas/farmacologia , Colistina/farmacologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Mutação
2.
Nutrients ; 13(2)2021 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-33669978

RESUMO

The link between autism spectrum disorder (ASD) and the gut microbiome has received much attention, with special focus on gut-brain-axis immunological imbalances. Gastrointestinal problems are one of the major symptoms of ASD and are thought to be related to immune dysregulation. Therefore, in silico analysis was performed on mined data from 36 individuals with ASD and 21 control subjects, with an emphasis on lipid A endotoxin-producing bacteria and their lipopolysaccharide (LPS) metabolic pathways. Analysis of enzyme distribution among the 15 most abundant genera in both groups revealed that almost all these genera utilized five early-stage enzymes responsible for catalyzing the nine conserved lipid A synthesis steps. However, Haemophilus and Escherichia, which were significantly more abundant in individuals with ASD than in the control subjects, possess a complete set of essential lipid A synthesis enzymes. Furthermore, the 10 genera with the greatest increase in individuals with ASD showed high potential for producing late-stage lipid A products. Collectively, these results suggested that the synthesis rate of immunogenic LPS end products is likely to increase in individuals with ASD, which may be related to their gastrointestinal symptoms and elevated inflammatory conditions.


Assuntos
Transtorno do Espectro Autista/microbiologia , Encéfalo/microbiologia , Gastroenteropatias/microbiologia , Microbioma Gastrointestinal/fisiologia , Lipídeo A/biossíntese , Transtorno do Espectro Autista/complicações , Estudos de Casos e Controles , Criança , Simulação por Computador , Feminino , Humanos , Lipopolissacarídeos/metabolismo , Masculino , Transdução de Sinais
3.
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
4.
Biotechnol Appl Biochem ; 68(6): 1486-1500, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33150647

RESUMO

In this study, four genes encoding secondary acyltransferases of lipid A in Vibrio parahaemolyticus ATCC33846 were identified. When the four genes were overexpressed in Escherichia coli MLK1067 that which produces the penta-acylated lipid A lacking the secondary acylation at the C3' position, a C12:0 secondary acyl chain was added at the C3' position of lipid A only in E. coli overexpressing VP_RS01045, but not VP_RS00880, VP_RS08405, or VP_RS12170. When the four genes were overexpressed in E. coli MKV15b that produces lipid IVA , a C12:0 secondary acyl chain was again added at the C3' position in E. coli overexpressing VP_RS01045, but a C14:0 secondary acyl chain was added at the C2' position of lipid A in E. coli overexpressing VP_RS00880, VP_RS08405, or VP_RS12170. The results indicate that four acyltransferases of lipid A are encoded by VP_RS01045, VP_RS00880, VP_RS08405, or VP_RS12170 in V. parahaemolyticus. The acyltransferase encoded by VP_RS01045 adds a C12:0 secondary acyl chain at the C3' position of lipid A, whereas the acyltransferase encoded by VP_RS00880, VP_RS08405, or VP_RS12170 adds a C14:0 secondary acyl chain at the C2' position of lipid A. This work contributes to understanding the biosynthetic pathway of lipid A in V. parahaemolyticus.


Assuntos
Aciltransferases/análise , Lipídeo A/biossíntese , Vibrio parahaemolyticus/metabolismo , Aciltransferases/metabolismo , Configuração de Carboidratos , Lipídeo A/genética
5.
J Biol Chem ; 295(31): 10593-10609, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32424042

RESUMO

Lipopolysaccharides are critical components of bacterial outer membranes. The more conserved lipid A part of the lipopolysaccharide molecule is a major element in the permeability barrier imposed by the outer membrane and offers a pathogen-associated molecular pattern recognized by innate immune systems. In contrast, the long-chain O-antigen polysaccharide (O-PS) shows remarkable structural diversity and fulfills a range of functions, depending on bacterial lifestyles. O-PS production is vital for the success of clinically important Gram-negative pathogens. The biological properties and functions of O-PSs are mostly independent of specific structures, but the size distribution of O-PS chains is particularly important in many contexts. Despite the vast O-PS chemical diversity, most are produced in bacterial cells by two assembly strategies, and the different mechanisms employed in these pathways to regulate chain-length distribution are emerging. Here, we review our current understanding of the mechanisms involved in regulating O-PS chain-length distribution and discuss their impact on microbial cell biology.


Assuntos
Bactérias Gram-Negativas/fisiologia , Antígenos O/biossíntese , Lipídeo A/biossíntese
6.
J Antimicrob Chemother ; 75(1): 51-59, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31586411

RESUMO

BACKGROUND: LpxB is an enzyme involved in the biosynthesis pathway of lipid A, a component of LPS. OBJECTIVES: To evaluate the lpxB gene in Acinetobacter baumannii as a potential therapeutic target and to propose antisense agents such as peptide nucleic acids (PNAs) as a tool to combat bacterial infection, either alone or in combination with known antimicrobial therapies. METHODS: RNA-seq analysis of the A. baumannii ATCC 17978 strain in a murine pneumonia model was performed to study the in vivo expression of lpxB. Protein expression was studied in the presence or absence of anti-lpxB (KFF)3K-PNA (pPNA). Time-kill curve analyses and protection assays of infected A549 cells were performed. The chequerboard technique was used to test for synergy between pPNA and colistin. A Galleria mellonella infection model was used to test the in vivo efficacy of pPNA. RESULTS: The lpxB gene was overexpressed during pneumonia. Treatment with a specific pPNA inhibited LpxB expression in vitro, decreased survival of the ATCC 17978 strain and increased the survival rate of infected A549 cells. Synergy was observed between pPNA and colistin in colistin-susceptible strains. In vivo assays confirmed that a combination treatment of anti-lpxB pPNA and colistin was more effective than colistin in monotherapy. CONCLUSIONS: The lpxB gene is essential for A. baumannii survival. Anti-lpxB pPNA inhibits LpxB expression, causing bacterial death. This pPNA showed synergy with colistin and increased the survival rate in G. mellonella. The data suggest that antisense pPNA molecules blocking the lpxB gene could be used as antibacterial agents.


Assuntos
Acinetobacter baumannii/efeitos dos fármacos , Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Colistina/farmacologia , DNA Antissenso/genética , Ácidos Nucleicos Peptídicos/farmacologia , Células A549 , Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/genética , Animais , Proteínas de Bactérias/genética , Vias Biossintéticas , Sinergismo Farmacológico , Expressão Gênica , Humanos , Lipídeo A/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Mariposas/microbiologia , RNA-Seq
7.
Metab Eng ; 57: 193-202, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31786244

RESUMO

Monophosphoryl lipid A (MPLA) species, including MPL (a trade name of GlaxoSmithKline) and GLA (a trade name of Immune Design, a subsidiary of Merck), are widely used as an adjuvant in vaccines, allergy drugs, and immunotherapy to boost the immune response. Even though MPLA is a derivative of lipopolysaccharide (LPS), a component of the outer membrane of Gram-negative bacteria, bacterial strains producing MPLA have not been found in nature nor engineered. In fact, MPLA generation involves expensive and laborious procedures based on synthetic routes or chemical transformation of precursors isolated from Gram-negative bacteria. Here, we report the engineering of an Escherichia coli strain for in situ production and accumulation of MPLA. Furthermore, we establish a succinct method for purifying MPLA from the engineered E. coli strain. We show that the purified MPLA (named EcML) stimulates the mouse immune system to generate antigen-specific IgG antibodies similarly to commercially available MPLA, but with a dramatically reduced manufacturing time and cost. Our system, employing the first engineered E. coli strain that directly produces the adjuvant EcML, could transform the current standard of industrial MPLA production.


Assuntos
Adjuvantes Imunológicos , Escherichia coli , Lipídeo A/análogos & derivados , Engenharia Metabólica , Adjuvantes Imunológicos/biossíntese , Adjuvantes Imunológicos/genética , Adjuvantes Imunológicos/isolamento & purificação , Adjuvantes Imunológicos/farmacologia , Animais , Formação de Anticorpos/efeitos dos fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Imunoglobulina G/biossíntese , Lipídeo A/biossíntese , Lipídeo A/genética , Lipídeo A/isolamento & purificação , Lipídeo A/farmacologia , Camundongos , Camundongos Endogâmicos BALB C
8.
Curr Opin Struct Biol ; 59: 81-90, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31003021

RESUMO

Peripheral and integral membrane proteins feature in stepwise assembly of complex glycans and glycoconjugates. Catalysis on membrane-bound substrates features challenges with substrate solubility and active-site accessibility. However, advantages in enzyme and substrate orientation and control of lateral membrane diffusion provide order to the multistep processes. Recent glycosyltransferase (GT) studies show that substrate diversity is met by the selection of folds which do not converge upon a common mechanism. Examples of polyprenol phosphate phosphoglycosyl transferases (PGTs) highlight that divergent fold families catalyze the same reaction with different mechanisms. Lipid A biosynthesis enzymes illustrate that variations on the robust Rossmann fold allow substrate diversity. Improved understanding of GT and PGT structure and function holds promise for better function prediction and improvement of therapeutic inhibitory ligands.


Assuntos
Membrana Celular/química , Glicoconjugados/química , Sítios de Ligação , Metabolismo dos Carboidratos , Catálise , Domínio Catalítico , Membrana Celular/metabolismo , Celulose/química , Celulose/metabolismo , Glucosiltransferases/química , Glucosiltransferases/metabolismo , Glicoconjugados/biossíntese , Glicosiltransferases/química , Glicosiltransferases/metabolismo , Lipídeo A/biossíntese , Lipídeo A/química , Poliprenois/química , Poliprenois/metabolismo , Polissacarídeos/química , Relação Quantitativa Estrutura-Atividade , Especificidade por Substrato
9.
Sci Rep ; 9(1): 3947, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30850651

RESUMO

UDP-N-acetylglucosamine (UDP-GlcNAc) acyltransferase (LpxA) catalyzes the first step of lipid A biosynthesis, the transfer of an R-3-hydroxyacyl chain from its acyl carrier protein (ACP) to the 3-OH group of UDP-GlcNAc. Essential in the growth of Gram-negative bacteria, LpxA is a logical target for antibiotics design. A pentadecapeptide (Peptide 920) with high affinity towards LpxA was previously identified in a phage display library. Here we created a small library of systematically designed peptides with the length of four to thirteen amino acids using Peptide 920 as a scaffold. The concentrations of these peptides at which 50% of LpxA is inhibited (IC50) range from 50 nM to >100 µM. We determined the crystal structure of E. coli LpxA in a complex with a potent inhibitor. LpxA-inhibitor interaction, solvent model and all contributing factors to inhibitor efficacy were well resolved. The peptide primarily occludes the ACP binding site of LpxA. Interactions between LpxA and the inhibitor are different from those in the structure of Peptide 920. The inhibitory peptide library and the crystal structure of inhibitor-bound LpxA described here may further assist in the rational design of inhibitors with antimicrobial activity that target LpxA and potentially other acyltransferases.


Assuntos
Antibacterianos/farmacologia , Desenho de Fármacos , Peptídeos/farmacologia , Uridina Difosfato N-Acetilglicosamina/antagonistas & inibidores , Antibacterianos/química , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Negativas/enzimologia , Concentração Inibidora 50 , Lipídeo A/antagonistas & inibidores , Lipídeo A/biossíntese , Biblioteca de Peptídeos , Peptídeos/química
10.
J Biol Chem ; 294(20): 7982-7989, 2019 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-30926608

RESUMO

Lipopolysaccharides are anchored to the outer membrane of Gram-negative bacteria by a hydrophobic moiety known as lipid A, which potently activates the host innate immune response. Lipid A of Bordetella pertussis, the causative agent of whooping cough, displays unusual structural asymmetry with respect to the length of the acyl chains at the 3 and 3' positions, which are 3OH-C10 and 3OH-C14 chains, respectively. Both chains are attached by the acyltransferase LpxA, the first enzyme in the lipid A biosynthesis pathway, which, in B. pertussis, has limited chain length specificity. However, this only partially explains the strict asymmetry of lipid A. In attempts to modulate the endotoxicity of B. pertussis lipid A, here we expressed the gene encoding LpxA from Neisseria meningitidis, which specifically attaches 3OH-C12 chains, in B. pertussis This expression was lethal, suggesting that one of the downstream enzymes in the lipid A biosynthesis pathway in B. pertussis cannot handle precursors with a 3OH-C12 chain. We considered that the UDP-diacylglucosamine pyrophosphohydrolase LpxH could be responsible for this defect as well as for the asymmetry of B. pertussis lipid A. Expression of meningococcal LpxH in B. pertussis indeed resulted in new symmetric lipid A species with 3OH-C10 or 3OH-C14 chains at both the 3 and 3' positions, as revealed by MS analysis. Furthermore, co-expression of meningococcal lpxH and lpxA resulted in viable cells that incorporated 3OH-C12 chains in B. pertussis lipid A. We conclude that the asymmetry of B. pertussis lipid A is determined by the acyl chain length specificity of LpxH.


Assuntos
Aciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Bordetella pertussis/enzimologia , Lipídeo A/biossíntese , Aciltransferases/química , Aciltransferases/genética , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Bordetella pertussis/genética , Células HEK293 , Humanos , Lipídeo A/química , Lipídeo A/genética , Camundongos , Neisseria meningitidis/enzimologia , Neisseria meningitidis/genética , Especificidade por Substrato/fisiologia
11.
J Bacteriol ; 200(19)2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30012728

RESUMO

Members of the Rickettsia genus are obligate intracellular, Gram-negative coccobacilli that infect mammalian and arthropod hosts. Several rickettsial species are human pathogens and are transmitted by blood-feeding arthropods. In Gram-negative parasites, the outer membrane (OM) sits at the nexus of the host-pathogen interaction and is rich in lipopolysaccharide (LPS). The lipid A component of LPS anchors the molecule to the bacterial surface and is an endotoxic agonist of Toll-like receptor 4 (TLR4). Despite the apparent importance of lipid A in maintaining OM integrity, as well as its inflammatory potential during infection, this molecule is poorly characterized in Rickettsia pathogens. In this work, we have identified and characterized new members of the recently discovered LpxJ family of lipid A acyltransferases in both Rickettsia typhi and Rickettsia rickettsii, the etiological agents of murine typhus and Rocky Mountain spotted fever, respectively. Our results demonstrate that these enzymes catalyze the addition of a secondary acyl chain (C14/C16) to the 3'-linked primary acyl chain of the lipid A moiety in the final steps of the Raetz pathway of lipid A biosynthesis. Since lipid A architecture is fundamental to bacterial OM integrity, we believe that rickettsial LpxJ may be important in maintaining membrane dynamics to facilitate molecular interactions at the host-pathogen interface that are required for adhesion and invasion of mammalian cells. This work contributes to our understanding of rickettsial outer membrane physiology and sets a foundation for further exploration of the envelope and its role in pathogenesis.IMPORTANCE Lipopolysaccharide (LPS) triggers an inflammatory response through the TLR4-MD2 receptor complex and inflammatory caspases, a process mediated by the lipid A moiety of LPS. Species of Rickettsia directly engage both extracellular and intracellular immunosurveillance, yet little is known about rickettsial lipid A. Here, we demonstrate that the alternative lipid A acyltransferase, LpxJ, from Rickettsia typhi and R. rickettsii catalyzes the addition of C16 fatty acid chains into the lipid A 3'-linked primary acyl chain, accounting for major structural differences relative to the highly inflammatory lipid A of Escherichia coli.


Assuntos
Aciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Ácidos Graxos/metabolismo , Lipídeo A/biossíntese , Rickettsia rickettsii/metabolismo , Rickettsia typhi/metabolismo , Aciltransferases/genética , Proteínas de Bactérias/genética , Genoma Bacteriano , Interações Hospedeiro-Patógeno , Rickettsia rickettsii/genética , Rickettsia typhi/genética
12.
FASEB J ; 32(2): 1085-1098, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29079699

RESUMO

Colistin is considered a last-resort antibiotic against most gram-negative bacteria. Recent discoveries of a plasmid-mediated, transferable mobilized colistin-resistance gene ( mcr-1) on all continents have heralded the imminent emergence of pan-drug-resistant superbacteria. The inner-membrane protein MCR-1 can catalyze the transfer of phosphoethanolamine (PEA) to lipid A, resulting in colistin resistance. However, little is known about the mechanism, and few drugs exist to address this issue. We present crystal structures revealing the MCR-1 catalytic domain (cMCR-1) as a monozinc metalloprotein with ethanolamine (ETA) and d-glucose, respectively, thus highlighting 2 possible substrate-binding pockets in the MCR-1-catalyzed PEA transfer reaction. Mutation of the residues involved in ETA and d-glucose binding impairs colistin resistance in recombinant Escherichia coli containing full-length MCR-1. Partial analogs of the substrate are used for cocrystallization with cMCR-1, providing valuable information about the family of PEA transferases. One of the analogs, ETA, causes clear inhibition of polymyxin B resistance, highlighting its potential for drug development. These data demonstrate the crucial role of the PEA- and lipid A-binding pockets and provide novel insights into the structure-based mechanisms, important drug-target hot spots, and a drug template for further drug development to combat the urgent, rising threat of MCR-1-mediated antibiotic resistance.-Wei, P., Song, G., Shi, M., Zhou, Y., Liu, Y., Lei, J., Chen, P., Yin, L. Substrate analog interaction with MCR-1 offers insight into the rising threat of the plasmid-mediated transferable colistin resistance.


Assuntos
Colistina/química , Farmacorresistência Bacteriana , Proteínas de Escherichia coli/química , Escherichia coli/enzimologia , Plasmídeos , Catálise , Colistina/farmacologia , Proteínas de Escherichia coli/metabolismo , Etanolaminas/química , Etanolaminas/metabolismo , Lipídeo A/biossíntese , Lipídeo A/química , Domínios Proteicos
13.
Med Res Rev ; 38(2): 556-601, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28621828

RESUMO

Lipid A is one of the core structures of bacterial lipopolysaccharides (LPSs), and it is mainly responsible for the strong immunostimulatory activities of LPS through interactions with the Toll-like receptors and other molecules in the human immune system. To obtain structurally homogeneous and well-defined lipid As and its derivatives in quantities meaningful for various biological studies and applications, their chemical synthesis has become a focal point. This review has provided a survey of significant progresses made in the synthesis of lipid A, and its derivatives that carry diverse saturated and unsaturated lipids, have the phosphate group at its reducing end replaced with a more stable phosphate or carboxyl group, or lack the reducing end phosphate or both phosphate groups, as well as progresses in the synthesis of LPS analogs and other lipid A conjugates. These synthetic molecules have facilitated the elucidation of the structure-activity relationships of lipid A useful for the design and development of lipid A based therapeutics, such as those utilized to treat sepsis, and other medical applications, for example the use of monophosphoryl lipid A as a carrier molecule for the study of fully synthetic self-adjuvanting conjugate vaccines. These topics are also briefly covered in the current review.


Assuntos
Lipídeo A/biossíntese , Lipídeo A/farmacologia , Animais , Vias Biossintéticas , Humanos , Lipídeo A/análogos & derivados , Lipídeo A/química , Vacinas/imunologia
14.
Mol Microbiol ; 106(4): 582-596, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28906060

RESUMO

In the environment and during infection, the human intestinal pathogen Vibrio cholerae must overcome noxious compounds that damage the bacterial outer membrane. The El Tor and classical biotypes of O1 V. cholerae show striking differences in their resistance to membrane disrupting cationic antimicrobial peptides (CAMPs), such as polymyxins. The classical biotype is susceptible to CAMPs, but current pandemic El Tor biotype isolates gain CAMP resistance by altering the net charge of their cell surface through glycine modification of lipid A. Here we report a second lipid A modification mechanism that only functions in the V. cholerae El Tor biotype. We identify a functional EptA ortholog responsible for the transfer of the amino-residue phosphoethanolamine (pEtN) to the lipid A of V. cholerae El Tor that is not functional in the classical biotype. We previously reported that mildly acidic growth conditions (pH 5.8) downregulate expression of genes encoding the glycine modification machinery. In this report, growth at pH 5.8 increases expression of eptA with concomitant pEtN modification suggesting coordinated regulation of these LPS modification systems. Similarly, efficient pEtN lipid A substitution is seen in the absence of lipid A glycinylation. We further demonstrate EptA orthologs from non-cholerae Vibrio species are functional.


Assuntos
Lipídeo A/metabolismo , Lipopolissacarídeos/metabolismo , Vibrio cholerae/metabolismo , Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas de Bactérias/metabolismo , Cólera/microbiologia , Etanolaminas/metabolismo , Glicina/metabolismo , Humanos , Lipídeo A/biossíntese , Lipopolissacarídeos/genética , Vibrio cholerae/genética
15.
mBio ; 8(4)2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28743813

RESUMO

The infectious diseases caused by multidrug-resistant bacteria pose serious threats to humankind. It has been suggested that an antibiotic targeting LpxC of the lipid A biosynthetic pathway in Gram-negative bacteria is a promising strategy for curing Gram-negative bacterial infections. However, experimental proof of this concept is lacking. Here, we describe our discovery and characterization of a biphenylacetylene-based inhibitor of LpxC, an essential enzyme in the biosynthesis of the lipid A component of the outer membrane of Gram-negative bacteria. The compound LPC-069 has no known adverse effects in mice and is effective in vitro against a broad panel of Gram-negative clinical isolates, including several multiresistant and extremely drug-resistant strains involved in nosocomial infections. Furthermore, LPC-069 is curative in a murine model of one of the most severe human diseases, bubonic plague, which is caused by the Gram-negative bacterium Yersinia pestis Our results demonstrate the safety and efficacy of LpxC inhibitors as a new class of antibiotic against fatal infections caused by extremely virulent pathogens. The present findings also highlight the potential of LpxC inhibitors for clinical development as therapeutics for infections caused by multidrug-resistant bacteria.IMPORTANCE The rapid spread of antimicrobial resistance among Gram-negative bacilli highlights the urgent need for new antibiotics. Here, we describe a new class of antibiotics lacking cross-resistance with conventional antibiotics. The compounds inhibit LpxC, a key enzyme in the lipid A biosynthetic pathway in Gram-negative bacteria, and are active in vitro against a broad panel of clinical isolates of Gram-negative bacilli involved in nosocomial and community infections. The present study also constitutes the first demonstration of the curative treatment of bubonic plague by a novel, broad-spectrum antibiotic targeting LpxC. Hence, the data highlight the therapeutic potential of LpxC inhibitors against a wide variety of Gram-negative bacterial infections, including the most severe ones caused by Y. pestis and by multidrug-resistant and extensively drug-resistant carbapenemase-producing strains.


Assuntos
Antibacterianos/uso terapêutico , Proteínas de Bactérias/antagonistas & inibidores , Benzamidas/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Bactérias Gram-Negativas/efeitos dos fármacos , Morfolinas/uso terapêutico , Peste/tratamento farmacológico , Yersinia pestis/efeitos dos fármacos , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Benzamidas/química , Benzamidas/farmacologia , Modelos Animais de Doenças , Farmacorresistência Bacteriana Múltipla , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Feminino , Bactérias Gram-Negativas/enzimologia , Infecções por Bactérias Gram-Negativas/tratamento farmacológico , Infecções por Bactérias Gram-Negativas/microbiologia , Lipídeo A/biossíntese , Camundongos , Morfolinas/química , Morfolinas/farmacologia , Peste/microbiologia , Yersinia pestis/enzimologia
16.
mBio ; 8(3)2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28487429

RESUMO

Adjuvant properties of bacterial cell wall components like MPLA (monophosphoryl lipid A) are well described and have gained FDA approval for use in vaccines such as Cervarix. MPLA is the product of chemically modified lipooligosaccharide (LOS), altered to diminish toxic proinflammatory effects while retaining adequate immunogenicity. Despite the virtually unlimited number of potential sources among bacterial strains, the number of useable compounds within this promising class of adjuvants are few. We have developed bacterial enzymatic combinatorial chemistry (BECC) as a method to generate rationally designed, functionally diverse lipid A. BECC removes endogenous or introduces exogenous lipid A-modifying enzymes to bacteria, effectively reprogramming the lipid A biosynthetic pathway. In this study, BECC is applied within an avirulent strain of Yersinia pestis to develop structurally distinct LOS molecules that elicit differential Toll-like receptor 4 (TLR4) activation. Using reporter cell lines that measure NF-κB activation, BECC-derived molecules were screened for the ability to induce a lower proinflammatory response than Escherichia coli LOS. Their structures exhibit varied, dose-dependent, TLR4-driven NF-κB activation with both human and mouse TLR4 complexes. Additional cytokine secretion screening identified molecules that induce levels of tumor necrosis factor alpha (TNF-α) and interleukin-8 (IL-8) comparable to the levels induced by phosphorylated hexa-acyl disaccharide (PHAD). The lead candidates demonstrated potent immunostimulation in mouse splenocytes, human primary blood mononuclear cells (PBMCs), and human monocyte-derived dendritic cells (DCs). This newly described system allows directed programming of lipid A synthesis and has the potential to generate a diverse array of TLR4 agonist candidates.IMPORTANCE There is an urgent need to develop effective vaccines against infectious diseases that continue to be major causes of morbidity and mortality worldwide. Making effective vaccines requires selecting an adjuvant to strengthen an appropriate and protective immune response. This work describes a practical method, bacterial enzymatic combinatorial chemistry (BECC), for generating functionally diverse molecules for adjuvant use. These molecules were analyzed in cell culture for their ability to initiate immune stimulatory activity. Several of the assays described herein show promising in vitro cytokine production and costimulatory molecule expression results, suggesting that the BECC molecules may be useful in future vaccine preparations.


Assuntos
Adjuvantes Imunológicos/química , Descoberta de Drogas , Lipídeo A/biossíntese , Lipopolissacarídeos/química , Receptor 4 Toll-Like/imunologia , Adjuvantes Imunológicos/isolamento & purificação , Animais , Linhagem Celular , Técnicas de Química Combinatória , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Escherichia coli/química , Humanos , Imunomodulação , Interleucina-8/biossíntese , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Ligantes , Lipídeo A/análogos & derivados , Lipídeo A/química , Lipídeo A/imunologia , Lipídeo A/metabolismo , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Camundongos , NF-kappa B/metabolismo , Receptor 4 Toll-Like/agonistas , Fator de Necrose Tumoral alfa/biossíntese , Yersinia pestis/química
17.
Antonie Van Leeuwenhoek ; 110(11): 1413-1433, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28409238

RESUMO

The structure of lipid A from lipopolysaccharide of Phyllobacterium trifolii PETP02T, a nitrogen-fixing symbiotic bacterium, was studied. It was found that the lipid A backbone was composed of two 2,3-diamino-2,3-dideoxy-D-glucose (GlcpN3N) residues connected by a ß-(1 â†’ 6) glycosidic linkage, substituted by galacturonic acid (GalpA) at position C-1 and partly decorated by a phosphate residue at C-4' of the non-reducing GlcpN3N. Both diaminosugars were symmetrically substituted by 3-hydroxy fatty acids (14:0(3-OH) and 16:0(3-OH)). Ester-linked secondary acyl residues [i.e. 19:0cyc and 28:0(27-OH) or 28:0(27-4:0(3-OMe))] were located in the distal part of lipid A. A high similarity between the lipid A of P. trifolii and Mesorhizobium was observed and discussed from the perspective of the genetic context of both genomes.


Assuntos
Lipídeo A/química , Lipopolissacarídeos/química , Phyllobacteriaceae/química , Ácidos Graxos/análise , Glucosamina/análogos & derivados , Glucosamina/química , Ácidos Hexurônicos/química , Lipídeo A/biossíntese , Lipídeo A/isolamento & purificação , Lipopolissacarídeos/isolamento & purificação , Espectroscopia de Ressonância Magnética , Mesorhizobium/química , Mesorhizobium/genética , Redes e Vias Metabólicas/genética , Phyllobacteriaceae/genética , Homologia de Sequência de Aminoácidos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
18.
J Bacteriol ; 199(11)2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28320881

RESUMO

Porphyromonas gingivalis produces outer membrane vesicles (OMVs) rich in virulence factors, including cysteine proteases and A-LPS, one of the two lipopolysaccharides (LPSs) produced by this organism. Previous studies had suggested that A-LPS and PG0027, an outer membrane (OM) protein, may be involved in OMV formation. Their roles in this process were examined by using W50 parent and the ΔPG0027 mutant strains. Inactivation of PG0027 caused a reduction in the yield of OMVs. Lipid A from cells and OMVs of P. gingivalis W50 and the ΔPG0027 mutant strains were analyzed by matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS). Lipid A from W50 cells contained bis-P-pentaacyl, mono-P-pentaacyl, mono-P-tetraacyl, non-P-pentaacyl, and non-P-tetraacyl species, whereas lipid A from ΔPG0027 mutant cells contained only phosphorylated species; nonphosphorylated species were absent. MALDI-TOF/TOF tandem MS of mono-P-pentaacyl (m/z 1,688) and mono-P-tetraacyl (m/z 1,448) lipid A from ΔPG0027 showed that both contained lipid A 1-phosphate, suggesting that the ΔPG0027 mutant strain lacked lipid A 1-phosphatase activity. The total phosphatase activities in the W50 and the ΔPG0027 mutant strains were similar, whereas the phosphatase activity in the periplasm of the ΔPG0027 mutant was lower than that in W50, supporting a role for PG0027 in lipid A dephosphorylation. W50 OMVs were enriched in A-LPS, and its lipid A did not contain nonphosphorylated species, whereas lipid A from the ΔPG0027 mutant (OMVs and cells) contained similar species. Thus, OMVs in P. gingivalis are apparently formed in regions of the OM enriched in A-LPS devoid of nonphosphorylated lipid A. Conversely, dephosphorylation of lipid A through a PG0027-dependent process is required for optimal formation of OMVs. Hence, the relative proportions of nonphosphorylated and phosphorylated lipid A appear to be crucial for OMV formation in this organism.IMPORTANCE Gram-negative bacteria produce outer membrane vesicles (OMVs) by "blebbing" of the outer membrane (OM). OMVs can be used offensively as delivery systems for virulence factors and defensively to aid in the colonization of a host and in the survival of the bacterium in hostile environments. Earlier studies using the oral anaerobe Porphyromonas gingivalis as a model organism to study the mechanism of OMV formation suggested that the OM protein PG0027 and one of the two lipopolysaccharides (LPSs) synthesized by this organism, namely, A-LPS, played important roles in OMV formation. We suggest a novel mechanism of OMV formation in P. gingivalis involving dephosphorylation of lipid A of A-LPS controlled/regulated by PG0027, which causes destabilization of the OM, resulting in blebbing and generation of OMVs.


Assuntos
Proteínas de Bactérias/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Porphyromonas gingivalis/metabolismo , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Lipídeo A/biossíntese , Monoéster Fosfórico Hidrolases/genética , Porphyromonas gingivalis/enzimologia , Porphyromonas gingivalis/genética
19.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(11): 1439-1450, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28108356

RESUMO

Strategies utilizing Toll-like receptor 4 (TLR4) agonists for treatment of cancer, infectious diseases, and other targets report promising results. Potent TLR4 antagonists are also gaining attention as therapeutic leads. Though some principles for TLR4 modulation by lipid A have been described, a thorough understanding of the structure-activity relationship (SAR) is lacking. Only through a complete definition of lipid A-TLR4 SAR is it possible to predict TLR4 signaling effects of discrete lipid A structures, rendering them more pharmacologically relevant. A limited 'toolbox' of lipid A-modifying enzymes has been defined and is largely composed of enzymes from mesophile human and zoonotic pathogens. Expansion of this 'toolbox' will result from extending the search into lipid A biosynthesis and modification by bacteria living at the extremes. Here, we review the fundamentals of lipid A structure, advances in lipid A uses in TLR4 modulation, and the search for novel lipid A-modifying systems in extremophile bacteria. This article is part of a Special Issue entitled: Bacterial Lipids edited by Russell E. Bishop.


Assuntos
Bactérias/enzimologia , Proteínas de Bactérias/metabolismo , Desenho de Fármacos , Enzimas/metabolismo , Lipídeo A/farmacologia , Receptor 4 Toll-Like/efeitos dos fármacos , Animais , Proteínas de Bactérias/química , Enzimas/química , Humanos , Lipídeo A/biossíntese , Lipídeo A/química , Lipogênese , Conformação Proteica , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade , Receptor 4 Toll-Like/química , Receptor 4 Toll-Like/metabolismo
20.
Mol Microbiol ; 104(1): 144-162, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28085228

RESUMO

Lipid A anchors the lipopolysaccharide (LPS) to the outer membrane and is usually composed of a hexa-acylated diglucosamine backbone. Burkholderia cenocepacia, an opportunistic pathogen, produces a mixture of tetra- and penta-acylated lipid A. "Late" acyltransferases add secondary acyl chains to lipid A after the incorporation of four primary acyl chains to the diglucosamine backbone. Here, we report that B. cenocepacia has only one late acyltransferase, LpxL (BCAL0508), which adds a myristoyl chain to the 2' position of lipid A resulting in penta-acylated lipid A. We also identified PagL (BCAL0788), which acts as an outer membrane lipase by removing the primary ß-hydroxymyristate (3-OH-C14:0) chain at the 3 position, leading to tetra-acylated lipid A. Unlike PagL, LpxL depletion caused reduced cell growth and defects in cell morphology, both of which were suppressed by overexpressing the LPS flippase MsbA (BCAL2408), suggesting that lipid A molecules lacking the fifth acyl chain contributed by LpxL are not good substrates for the flippase. We also show that intracellular B. cenocepacia within macrophages produced more penta-acylated lipid A, suggesting lipid A penta-acylation in B. cenocepacia is required not only for bacterial growth and morphology but also for adaptation to intracellular lifestyle.


Assuntos
Aciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Lipídeo A/biossíntese , Lipídeo A/metabolismo , Acilação , Burkholderia cenocepacia/crescimento & desenvolvimento , Burkholderia cenocepacia/metabolismo , Lipopolissacarídeos/metabolismo , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA